Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Value Health ; 26(7): 1085-1097, 2023 07.
Article in English | MEDLINE | ID: mdl-36878311

ABSTRACT

OBJECTIVES: To describe how subclinical hypothyroidism (SubHypo) influences the quality of life (QoL) during pregnancy. METHODS: In primary data collection (NCT04167423), thyroid stimulating hormone (TSH), free thyroxine (FT4), thyroid peroxidase antibodies, generic quality of life (QoL; 5-level version of EQ-5D [EQ-5D-5L]), and disease-specific QoL (ThyPRO-39) were measured among pregnant women. SubHypo during each trimester was defined according to the 2014 European Thyroid Association guidelines (TSH > 2.5, 3.0, and 3.5 IU/L, respectively; with normal FT4). Path analysis described relationships and tested mediation. Linear ordinary least squares, beta, tobit, and two-part regressions were used to map ThyPRO-39 and EQ-5D-5L. Alternative SubHypo definition was tested in sensitivity analysis. RESULTS: A total of 253 women at 14 sites (31 ± 5 years old, 15 ± 6 weeks pregnant) completed the questionnaires. Sixty-one (26%) had SubHypo and differed from 174 (74%) euthyroid women in smoking history (61% vs 41%), primiparity (62% vs 43%) and TSH level (4.1 ± 1.4 vs 1.5 ± 0.7 mIU/L, P < .001). EQ-5D-5L utility in SubHypo (0.89 ± 0.12) was lower than that in euthyroid (0.92 ± 0.11; P = .028) even after adjustment (difference -0.04, P = .033), whereas ocular (P = .001, ThyPRO-39), cognitive symptoms (P = .043), anxiety (P < .0001), and the composite score were higher. The impact of SubHypo on utility was mediated by anxiety. Results were confirmed by sensitivity analysis. Final mapping equation (ordinary least squares) includes goiter symptoms, anxiety, upset stomach, composite score (ThyPRO-39), FT4 levels, and week of pregnancy (determination coefficient 0.36). CONCLUSION: This is the first QoL mapping of SubHypo during pregnancy and the first evidence that SubHypo is associated with a negative impact on QoL. The effect is mediated by anxiety. EQ-5D-5L utilities can be generated based on ThyPRO-39 scores collected in pregnant euthyroid and patients with SubHypo.


Subject(s)
Hypothyroidism , Quality of Life , Adult , Female , Humans , Pregnancy , Anxiety , Quality of Life/psychology , Surveys and Questionnaires , Thyrotropin
2.
Toxicol Appl Pharmacol ; 434: 115823, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34896433

ABSTRACT

Alisertib (MLN8237), a novel Aurora A kinase inhibitor, is currently being clinically tested in late-phase trials for the therapy of various malignancies. In the present work, we describe alisertib's potential to perpetrate pharmacokinetic drug-drug interactions (DDIs) and/or to act as an antagonist of multidrug resistance (MDR). In accumulation assays, alisertib potently inhibited ABCC1 transporter, but not ABCB1 or ABCG2. The results of molecular modeling suggested a bifunctional mechanism for interaction on ABCC1. In addition, alisertib was characterized as a low- to moderate-affinity inhibitor of recombinant CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2D6 isoenzymes, but without potential clinical relevance. Drug combination studies revealed the capability of alisertib to synergistically antagonize ABCC1-mediated resistance to daunorubicin. Although alisertib exhibited substrate characteristics toward ABCB1 transporter in monolayer transport assays, comparative proliferation studies showed lack of its MDR-victim behavior in cells overexpressing ABCB1 as well as ABCG2 and ABCC1. Lastly, alisertib did not affect the expression of ABCC1, ABCG2, ABCB1 transporters and CYP1A2, CYP3A4, CYP2B6 isozymes on mRNA level in various systemic and tumoral models. In conclusion, our study suggests that alisertib is a drug candidate with negligible potential for perpetrating systemic pharmacokinetic DDIs on ABCB1, ABCG2 and cytochromes P450. In addition, we introduce alisertib as an effective dual-activity chemosensitizer whose MDR-antagonistic capacities are not impaired by efflux or effect on MDR phenotype. Our in vitro findings provide important pieces of information for clinicians when introducing alisertib into the clinical area.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Azepines/pharmacology , Azepines/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Catalytic Domain , Cell Line , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression Regulation/drug effects , Humans , Models, Molecular , Molecular Docking Simulation , Protein Conformation
3.
Arch Toxicol ; 96(12): 3265-3277, 2022 12.
Article in English | MEDLINE | ID: mdl-35972551

ABSTRACT

Targeting mutations that trigger acute myeloid leukaemia (AML) has emerged as a refined therapeutic approach in recent years. Enasidenib (Idhifa) is the first selective inhibitor of mutated forms of isocitrate dehydrogenase 2 (IDH2) approved against relapsed/refractory AML. In addition to its use as monotherapy, a combination trial of enasidenib with standard intensive induction therapy (daunorubicin + cytarabine) is being evaluated. This study aimed to decipher enasidenib off-target molecular mechanisms involved in anthracycline resistance, such as reduction by carbonyl reducing enzymes (CREs) and drug efflux by ATP-binding cassette (ABC) transporters. We analysed the effect of enasidenib on daunorubicin (Daun) reduction by several recombinant CREs and different human cell lines expressing aldo-keto reductase 1C3 (AKR1C3) exogenously (HCT116) or endogenously (A549 and KG1a). Additionally, A431 cell models overexpressing ABCB1, ABCG2, or ABCC1 were employed to evaluate enasidenib modulation of Daun efflux. Furthermore, the potential synergism of enasidenib over Daun cytotoxicity was quantified amongst all the cell models. Enasidenib selectively inhibited AKR1C3-mediated inactivation of Daun in vitro and in cell lines expressing AKR1C3, as well as its extrusion by ABCB1, ABCG2, and ABCC1 transporters, thus synergizing Daun cytotoxicity to overcome resistance. This work provides in vitro evidence on enasidenib-mediated targeting of the anthracycline resistance actors AKR1C3 and ABC transporters under clinically achievable concentrations. Our findings may encourage its combination with intensive chemotherapy and even suggest that the effectiveness of enasidenib as monotherapy against AML could lie beyond the targeting of mIDH2.


Subject(s)
Daunorubicin , Leukemia, Myeloid, Acute , Humans , Daunorubicin/pharmacology , ATP-Binding Cassette Transporters/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Anthracyclines , Antibiotics, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Adenosine Triphosphate
4.
Int J Mol Sci ; 23(22)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36430819

ABSTRACT

Talazoparib (Talzenna) is a novel poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor that is clinically used for the therapy of breast cancer. Furthermore, the drug has shown antitumor activity against different cancer types, including non-small cell lung cancer (NSCLC). In this work, we investigated the possible inhibitory interactions of talazoparib toward selected ATP-binding cassette (ABC) drug efflux transporters and cytochrome P450 biotransformation enzymes (CYPs) and evaluated its position in multidrug resistance (MDR). In accumulation studies, talazoparib interacted with the ABCC1 and ABCG2 transporters, but there were no significant effects on ABCB1. Furthermore, incubation assays revealed a negligible capacity of the tested drug to inhibit clinically relevant CYPs. In in vitro drug combination experiments, talazoparib synergistically reversed daunorubicin and mitoxantrone resistance in cells with ABCC1 and ABCG2 expression, respectively. Importantly, the position of an effective MDR modulator was further confirmed in drug combinations performed in ex vivo NSCLC patients-derived explants, whereas the possible victim role was refuted in comparative proliferation experiments. In addition, talazoparib had no significant effects on the mRNA-level expressions of MDR-related ABC transporters in the MCF-7 cellular model. In summary, our study presents a comprehensive overview on the pharmacokinetic drug-drug interactions (DDI) profile of talazoparib. Moreover, we introduced talazoparib as an efficient MDR antagonist.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Multiple , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cytochrome P-450 Enzyme System/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics
5.
Klin Mikrobiol Infekc Lek ; 28(1): 4-9, 2022 Mar.
Article in Czech | MEDLINE | ID: mdl-36183411

ABSTRACT

Spontaneous pneumothorax, pneumomediastinum and subcutaneous emphysema are serious complications of COVID-19 infection caused by SARS-CoV-2, occurring in approximately 1% of hospitalized patients. The risk increases with the accumulation of risk factors, namely moderate or severe illness, high-flow oxygen therapy and noninvasive ventilation. The pathophysiology may be contributed to by patient self-inflicted lung injury. Hypoxia-induced respiratory effort in patients with pneumonia puts an enormous load on certain parts of their lungs, leading to subsequent progression of lung tissue damage. These complications start with destruction of the alveolar membrane, accompanied by emergence of a pulmonary bulla. Rupture of the bulla results in air leaking to the pleural space (pneumothorax). In the case of pneumomediastinum, the air spreads within the peribronchial interstitium along the airways to the mediastinum and subcutaneous tissues (Macklin effect). While pneumomediastinum and subcutaneous emphysema resolve spontaneously in most cases, pneumothorax treatment depends on its magnitude. While small pneumothorax may be managed conservatively, large pneumothorax usually requires active treatment with an acute chest drain; the latter is also associated with worse prognosis and a higher chance of death. We report air-leak complications in nine COVID-19 patients, of whom seven had spontaneous pneumothorax and four of them died. Three patients developed spontaneous pneumomediastinum together with subcutaneous emphysema, with two of them surviving. One patient with combination of pneumothorax, pneumomediastinum and subcutaneous emphysema died. These complications may accompany moderate or severe COVID-19-associated pneumonia, mostly the late phase of the disease. They should be considered when patients complain of sudden chest or back pain or worsening respiratory insufficiency.


Subject(s)
COVID-19 , Mediastinal Emphysema , Pneumothorax , Subcutaneous Emphysema , Blister/complications , COVID-19/complications , Humans , Mediastinal Emphysema/diagnosis , Mediastinal Emphysema/etiology , Mediastinal Emphysema/therapy , Oxygen , Pneumothorax/etiology , Pneumothorax/therapy , SARS-CoV-2 , Subcutaneous Emphysema/complications
6.
Arch Toxicol ; 95(1): 67-78, 2021 01.
Article in English | MEDLINE | ID: mdl-33025066

ABSTRACT

Midostaurin is an FMS-like tyrosine kinase 3 receptor (FLT3) inhibitor that provides renewed hope for treating acute myeloid leukaemia (AML). The limited efficacy of this compound as a monotherapy contrasts with that of its synergistic combination with standard cytarabine and daunorubicin (Dau), suggesting a therapeutic benefit that is not driven only by FLT3 inhibition. In an AML context, the activity of the enzyme aldo-keto reductase 1C3 (AKR1C3) is a crucial factor in chemotherapy resistance, as it mediates the intracellular transformation of anthracyclines to less active hydroxy metabolites. Here, we report that midostaurin is a potent inhibitor of Dau inactivation mediated by AKR1C3 in both its recombinant form as well as during its overexpression in a transfected cell model. Likewise, in the FLT3- AML cell line KG1a, midostaurin was able to increase the cellular accumulation of Dau and significantly decrease its metabolism by AKR1C3 simultaneously. The combination of those mechanisms increased the nuclear localization of Dau, thus synergizing its cytotoxic effects on KG1a cells. Our results provide new in vitro evidence of how the therapeutic activity of midostaurin could operate beyond targeting the FLT3 receptor.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Daunorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Staurosporine/analogs & derivatives , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Biotransformation , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Daunorubicin/metabolism , Drug Synergism , HCT116 Cells , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Staurosporine/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
7.
Int J Mol Sci ; 22(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34769363

ABSTRACT

Tepotinib is a novel tyrosine kinase inhibitor recently approved for the treatment of non-small cell lung cancer (NSCLC). In this study, we evaluated the tepotinib's potential to perpetrate pharmacokinetic drug interactions and modulate multidrug resistance (MDR). Accumulation studies showed that tepotinib potently inhibits ABCB1 and ABCG2 efflux transporters, which was confirmed by molecular docking. In addition, tepotinib inhibited several recombinant cytochrome P450 (CYP) isoforms with varying potency. In subsequent drug combination experiments, tepotinib synergistically reversed daunorubicin and mitoxantrone resistance in cells with ABCB1 and ABCG2 overexpression, respectively. Remarkably, MDR-modulatory properties were confirmed in ex vivo explants derived from NSCLC patients. Furthermore, we demonstrated that anticancer effect of tepotinib is not influenced by the presence of ABC transporters associated with MDR, although monolayer transport assays designated it as ABCB1 substrate. Finally, tested drug was observed to have negligible effect on the expression of clinically relevant drug efflux transporters and CYP enzymes. In conclusion, our findings provide complex overview on the tepotinib's drug interaction profile and suggest a promising novel therapeutic strategy for future clinical investigations.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytostatic Agents/pharmacology , Drug Interactions , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Piperidines/pharmacology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , In Vitro Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
8.
Arch Toxicol ; 94(9): 3059-3068, 2020 09.
Article in English | MEDLINE | ID: mdl-32588086

ABSTRACT

Although novel anticancer drugs are being developed intensively, anthracyclines remain the gold standard in the treatment of acute myeloid leukaemia (AML). The reductive conversion of daunorubicin (Dau) to less active daunorubicinol (Dau-ol) is an important mechanism that contributes to the development of pharmacokinetic anthracycline resistance. Dau is a key component in many AML regimes, in which it is combined with many drugs, including all-trans-retinoic acid (ATRA), cytarabine, cladribine and prednisolone. In the present study, we investigated the influence of these anticancer drugs on the reductive Dau metabolism mediated by the aldo-keto reductases AKR1A1, 1B10, 1C3, and 7A2 and carbonyl reductase 1 (CBR1). In incubation experiments with recombinant enzymes, cladribine and cytarabine did not significantly inhibit the activity of the tested enzymes. Prednisolone inhibited AKR1C3 with an IC50 of 41.73 µM, while ATRA decreased the activity of AKR1B10 (IC50 = 78.33 µM) and AKR1C3 (IC50 = 1.17 µM). Subsequent studies showed that AKR1C3 inhibition mediated by ATRA exhibited tight binding (Kiapp = 0.54 µM). Further, the combination of 1 µM ATRA with different concentrations of Dau demonstrated synergistic effects in HCT116 and KG1a human cells expressing AKR1C3. Our results suggest that ATRA-mediated inhibition of AKR1C3 can contribute to the mechanisms that are hidden beyond the beneficial clinical outcome of the ATRA-Dau combination.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carbonyl Reductase (NADPH)/metabolism , Leukemia, Myeloid, Acute/drug therapy , Alcohol Oxidoreductases , Anthracyclines , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents , Daunorubicin , Humans
9.
Drug Metab Dispos ; 47(7): 699-709, 2019 07.
Article in English | MEDLINE | ID: mdl-31068367

ABSTRACT

Alectinib is a tyrosine kinase inhibitor currently used as a first-line treatment of anaplastic lymphoma kinase-positive metastatic nonsmall cell lung cancer (NSCLC). In the present work, we investigated possible interactions of this novel drug with ATP-binding cassette (ABC) drug efflux transporters and cytochrome P450 (P450) biotransformation enzymes that play significant roles in the phenomenon of multidrug resistance (MDR) of cancer cells as well as in pharmacokinetic drug-drug interactions. Using accumulation studies in Madin-Darby canine kidney subtype 2 (MDCKII) cells alectinib was identified as an inhibitor of ABCB1 and ABCG2 but not of ABCC1. In subsequent drug combination studies, we demonstrated the ability for alectinib to effectively overcome MDR in ABCB1- and ABCG2-overexpressing MDCKII and A431 cells. To describe the pharmacokinetic interaction profile of alectinib in a complete fashion, its possible inhibitory properties toward clinically relevant P450 enzymes (i.e., CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, or CYP3A5) were evaluated using human P450-expressing insect microsomes, revealing alectinib as a poor interactor. Advantageously for its use in pharmacotherapy, alectinib further exhibited negligible potential to cause any changes in expression of ABCB1, ABCG2, ABCC1, CYP1A2, CYP3A4, and CYP2B6 in intestine, liver, and NSCLC models. Our in vitro observations might serve as a valuable foundation for future in vivo studies that could support the rationale for our conclusions and possibly enable providing more efficient and safer therapy to many oncological patients.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Carbazoles/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Biotransformation , Carbazoles/pharmacokinetics , Dogs , Humans , Madin Darby Canine Kidney Cells , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics
10.
Arch Toxicol ; 92(9): 2845-2857, 2018 09.
Article in English | MEDLINE | ID: mdl-29992508

ABSTRACT

Dinaciclib is a multi-specific cyclin-dependent kinase (CDK) inhibitor with significant preclinical and clinical activity. It inhibits CDK1, CDK2, CDK5, CDK9 and CDK12 in the nanomolar range and exhibits potent antiproliferative effects on various cancers in vitro and in vivo. Aldo-keto reductases (AKR) and carbonyl reductases (CBR) are enzymes involved at the biosynthesis, intermediary metabolism and detoxification processes, but can also play a significant role in cancer resistance. Here, we report that dinaciclib is a strong inhibitor of aldo-keto reductase 1C3 (AKR1C3), an enzyme that is known to be an important regulator of cell proliferation and differentiation. AKR1C3 is overexpressed in a range of cancer types and is also involved in tumour cell resistance to anthracyclines. In our study, dinaciclib displayed tight-binding inhibition of human recombinant AKR1C3 (Kiapp = 0.07 µM) and was also active at the cellular level (IC50 = 0.23 µM). Dinaciclib acts as a noncompetitive inhibitor with respect to daunorubicin and as an uncompetitive inhibitor with respect to the NADPH. In subsequent experiments, pretreatment with dinaciclib (0.1 µM) significantly sensitized AKR1C3-overexpressing anthracycline-resistant cancer cells to daunorubicin. In conclusion, our results indicate that dinaciclib may potentially increase the therapeutic efficacy and safety of anthracyclines by preventing anthracycline resistance and minimizing their adverse effects.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Anthracyclines/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cyclic N-Oxides , Daunorubicin/metabolism , Daunorubicin/pharmacokinetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , HCT116 Cells , Hep G2 Cells , Humans , Indolizines , Pyridinium Compounds/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Arch Pharm (Weinheim) ; 350(8)2017 Aug.
Article in English | MEDLINE | ID: mdl-28635184

ABSTRACT

A series of benzaldehyde and salicylaldehyde-S-benzylisothiosemicarbazones was synthesized and tested against 12 different strains of mycobacteria, Gram-positive and Gram-negative bacteria, and the significant selectivity toward mycobacteria was proved. Twenty-eight derivatives were evaluated for the inhibition of isocitrate lyase, which is a key enzyme of the glyoxylate cycle necessary for latent tuberculosis infection, and their iron-chelating properties were investigated. Two derivatives, 5-bromosalicylaldehyde-S-(4-fluorobenzyl)-isothiosemicarbazone and salicylaldehyde-S-(4-bromobenzyl)-isothiosemicarbazone, influenced the isocitrate lyase activity and caused a better inhibition at 10 µmol/L than 3-nitropropionic acid, a standard inhibitor. The compounds were also found to act as exogenous chelators of iron, which is an obligate cofactor for many mycobacterial enzymes. Due to their low cytotoxicity, together with the activity against isocitrate lyase and the ability to sequester iron ions, the compounds belong to potential antibiotics with the main effect on mycobacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antitubercular Agents/pharmacology , Mycobacterium/drug effects , Thiosemicarbazones/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Isocitrate Lyase/antagonists & inhibitors , Structure-Activity Relationship , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry
12.
J Nat Prod ; 78(11): 2666-74, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26529431

ABSTRACT

AKR1B10 is an NADPH-dependent reductase that plays an important function in several physiological reactions such as the conversion of retinal to retinol, reduction of isoprenyl aldehydes, and biotransformation of procarcinogens and drugs. A growing body of evidence points to the important role of the enzyme in the development of several types of cancer (e.g., breast, hepatocellular), in which it is highly overexpressed. AKR1B10 is regarded as a therapeutic target for the treatment of these diseases, and potent and specific inhibitors may be promising therapeutic agents. Several inhibitors of AKR1B10 have been described, but the area of natural plant products has been investigated sparingly. In the present study almost 40 diverse phenolic compounds and alkaloids were examined for their ability to inhibit the recombinant AKR1B10 enzyme. The most potent inhibitors-apigenin, luteolin, and 7-hydroxyflavone-were further characterized in terms of IC50, selectivity, and mode of action. Molecular docking studies were also conducted, which identified putative binding residues important for the interaction. In addition, cellular studies demonstrated a significant inhibition of the AKR1B10-mediated reduction of daunorubicin in intact cells by these inhibitors without a considerable cytotoxic effect. Although these compounds are moderately potent and selective inhibitors of AKR1B10, they constitute a new structural type of AKR1B10 inhibitor and may serve as a template for the development of better inhibitors.


Subject(s)
Aldehyde Reductase/drug effects , Flavones/pharmacology , Neoplasms/drug therapy , Aldehyde Reductase/antagonists & inhibitors , Aldo-Keto Reductases , Apigenin/pharmacology , Daunorubicin/pharmacology , Enzyme Inhibitors/chemistry , Flavones/chemistry , Flavonoids/pharmacology , HCT116 Cells , Humans , Luteolin/pharmacology , Molecular Conformation , Molecular Structure
13.
Toxicol Appl Pharmacol ; 278(3): 238-48, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24832494

ABSTRACT

Pharmacokinetic drug resistance is a serious obstacle that emerges during cancer chemotherapy. In this study, we investigated the possible role of aldo-keto reductase 1C3 (AKR1C3) in the resistance of cancer cells to anthracyclines. First, the reducing activity of AKR1C3 toward anthracyclines was tested using incubations with a purified recombinant enzyme. Furthermore, the intracellular reduction of daunorubicin and idarubicin was examined by employing the transfection of A549, HeLa, MCF7 and HCT 116 cancer cells with an AKR1C3 encoding vector. To investigate the participation of AKR1C3 in anthracycline resistance, we conducted MTT cytotoxicity assays with these cells, and observed that AKR1C3 significantly contributes to the resistance of cancer cells to daunorubicin and idarubicin, whereas this resistance was reversible by the simultaneous administration of 2'-hydroxyflavanone, a specific AKR1C3 inhibitor. In the final part of our work, we tracked the changes in AKR1C3 expression after anthracycline exposure. Interestingly, a reciprocal correlation between the extent of induction and endogenous levels of AKR1C3 was recorded in particular cell lines. Therefore, we suggest that the induction of AKR1C3 following exposure to daunorubicin and idarubicin, which seems to be dependent on endogenous AKR1C3 expression, eventually might potentiate an intrinsic resistance given by the normal expression of AKR1C3. In conclusion, our data suggest a substantial impact of AKR1C3 on the metabolism of daunorubicin and idarubicin, which affects their pharmacokinetic and pharmacodynamic behavior. In addition, we demonstrate that the reduction of daunorubicin and idarubicin, which is catalyzed by AKR1C3, contributes to the resistance of cancer cells to anthracycline treatment.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Carcinoma/drug therapy , Drug Resistance, Neoplasm , Hydroxyprostaglandin Dehydrogenases/metabolism , Neoplasm Proteins/metabolism , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3 , Anthracyclines/agonists , Anthracyclines/metabolism , Antibiotics, Antineoplastic/agonists , Antibiotics, Antineoplastic/metabolism , Biotransformation , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/agonists , Daunorubicin/metabolism , Daunorubicin/pharmacology , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Flavanones/pharmacology , Humans , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/genetics , Idarubicin/agonists , Idarubicin/metabolism , Idarubicin/pharmacology , Kinetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
14.
Bioorg Med Chem ; 22(15): 4073-82, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24953953

ABSTRACT

The development of novel antimicrobial drugs represents a cutting edge research topic. In this study, 20 salicylanilide N,N-disubstituted carbamates and thiocarbamates were designed, synthesised and characterised by IR, (1)H NMR and (13)C NMR. The compounds were evaluated in vitro as potential antimicrobial agents against Mycobacterium tuberculosis and nontuberculous mycobacteria (Mycobacterium avium and Mycobacterium kansasii) as well as against eight bacterial and fungal strains. Additionally, we investigated the inhibitory effect of these compounds on mycobacterial isocitrate lyase and cellular toxicity. The minimum inhibitory concentrations (MICs) against mycobacteria were from 4 µM for thiocarbamates and from 16 µM for carbamates. Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus, were inhibited with MICs from 0.49 µM by thiocarbamates, whilst Gram-negative bacteria and most of the fungi did not display any significant susceptibility. All (thio)carbamates mildly inhibited isocitrate lyase (up to 22%) at a concentration of 10 µM. The (thio)carbamoylation of the parent salicylanilides led to considerably decreased cytotoxicity and thus improved the selectivity indices (up to 175). These values indicate that some derivatives are attractive candidates for future research.


Subject(s)
Anti-Infective Agents/chemical synthesis , Carbamates/chemistry , Salicylanilides/chemistry , Thiocarbamates/chemistry , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Carbamates/chemical synthesis , Carbamates/pharmacology , Cell Survival/drug effects , Fungi/drug effects , Gram-Positive Bacteria/drug effects , Hep G2 Cells , Humans , Isocitrate Lyase/antagonists & inhibitors , Isocitrate Lyase/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Mycobacterium avium/drug effects , Mycobacterium kansasii/drug effects , Structure-Activity Relationship , Thiocarbamates/chemical synthesis , Thiocarbamates/pharmacology
15.
Arch Pharm (Weinheim) ; 347(6): 381-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24664885

ABSTRACT

New quaternary ammonium salt-type compounds with lipophilic cholesterol and terpene moieties were synthesized. The compounds showed promising antibacterial and antimycobacterial activities. Those compounds containing the cholesterol moiety showed significant activity against Staphylococcus aureus, Staphylococcus epidermidis, and Enterococcus faecium. On the contrary, the antimycobacterial activity increased with the presence of the terpene unit in the molecule.


Subject(s)
Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Cholesterol/chemical synthesis , Cholesterol/pharmacology , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/pharmacology , Terpenes/pharmacology , Cholesterol/analogs & derivatives , Drug Design , Enterococcus faecium/drug effects , Enterococcus faecium/growth & development , Fungi/drug effects , Fungi/growth & development , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium/drug effects , Mycobacterium/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/growth & development , Structure-Activity Relationship
16.
ScientificWorldJournal ; 2014: 703053, 2014.
Article in English | MEDLINE | ID: mdl-25538961

ABSTRACT

Antimycobacterially active salicylanilide diethyl phosphates were evaluated to identify their potential drug target(s) for the inhibition of several mycobacterial enzymes, including isocitrate lyase, L-alanine dehydrogenase (MtAlaDH), lysine ε-aminotransferase, chorismate mutase, and pantothenate synthetase. The enzymes are related to the nongrowing state of Mycobacterium tuberculosis. Salicylanilide diethyl phosphates represent new candidates with significant inhibitory activity especially against L-alanine dehydrogenase. The most active MtAlaDH inhibitor, 5-chloro-2-[(3-chlorophenyl)carbamoyl]phenyl diethyl phosphate, has an IC50 of 4.96 µM and the best docking results. Other mycobacterial enzymes were mostly inhibited by some derivatives but at higher concentrations; isocitrate lyase showed the highest resistance to salicylanilide diethyl phosphates.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Mycobacterium tuberculosis/enzymology , Salicylanilides/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry
17.
Birth Defects Res B Dev Reprod Toxicol ; 98(6): 493-502, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24395216

ABSTRACT

Ochratoxin A (OTA) is nephrotoxic, hepatotoxic, reprotoxic, embryotoxic, teratogenic, neurotoxic, immunotoxic, and carcinogenic for laboratory and farm animals. Male and female reproductive health has deteriorated in many countries during the last few decades. A number of toxins in environment are suspected to affect reproductive system in male and female. OTA is one of them. OTA has been found to be teratogenic in several animal models including rat, mouse, hamster, quail, and chick, with reduced birth weight and craniofacial abnormalities being the most common signs. The presence of OTA also results in congenital defects in the fetus. Neither the potential of OTA to cause malformations in human nor its teratogenic mode of action is known. Exposure to OTA leads to increased embryo lethality manifested as resorptions or dead fetuses. The mechanism of OTA transfer across human placenta (e.g., which transporters are involved in the transfer mechanism) is not fully understood. Some of the toxic effects of OTA are potentiated by other mycotoxins or other contaminants. Therefore, OTA exposure of pregnant women should be minimized. OTA has been shown to be an endocrine disruptor and a reproductive toxicant, with abilities of altering sperm quality. Other studies have shown that OTA is a testicular toxin in animals. Thus, OTA is a biologically plausible cause of testicular cancer in man.


Subject(s)
Environmental Exposure/adverse effects , Ochratoxins/toxicity , Teratogenesis/drug effects , Animals , Calcium Channel Blockers/toxicity , Carcinogens/toxicity , Cricetinae , Endocrine Disruptors/toxicity , Female , Humans , Male , Mice , Pregnancy , Rats , Reproduction/drug effects , Spermatozoa/abnormalities , Teratogens/toxicity , Testicular Neoplasms/chemically induced
18.
Radiat Environ Biophys ; 52(4): 471-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934411

ABSTRACT

We compared the effects of inhibitors of kinases ATM (KU55933) and ATR (VE-821) (incubated for 30 min before irradiation) on the radiosensitization of human promyelocyte leukaemia cells (HL-60), lacking functional protein p53. VE-821 reduces phosphorylation of check-point kinase 1 at serine 345, and KU55933 reduces phosphorylation of check-point kinase 2 on threonine 68 as assayed 4 h after irradiation by the dose of 6 Gy. Within 24 h after gamma-irradiation with a dose of 3 Gy, the cells accumulated in the G2 phase (67 %) and the number of cells in S phase decreased. KU55933 (10 µM) did not affect the accumulation of cells in G2 phase and did not affect the decrease in the number of cells in S phase after irradiation. VE-821 (2 and 10 µM) reduced the number of irradiated cells in the G2 phase to the level of non-irradiated cells and increased the number of irradiated cells in S phase, compared to irradiated cells not treated with inhibitors. In the 144 h interval after irradiation with 3 Gy, there was a considerable induction of apoptosis in the VE-821 group (10 µM). The repair of the radiation damage, as observed 72 h after irradiation, was more rapid in the group exposed solely to irradiation and in the group treated with KU55933 (80 and 77 % of cells, respectively, were free of DSBs), whereas in the group incubated with 10 µM VE-821, there were only 61 % of cells free of DSBs. The inhibition of kinase ATR with its specific inhibitor VE-821 resulted in a more pronounced radiosensitizing effect in HL-60 cells as compared to the inhibition of kinase ATM with the inhibitor KU55933. In contrast to KU55933, the VE-821 treatment prevented HL-60 cells from undergoing G2 cell cycle arrest. Taken together, we conclude that the ATR kinase inhibition offers a new possibility of radiosensitization of tumour cells lacking functional protein p53.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Leukemia, Promyelocytic, Acute/pathology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Radiation Tolerance/drug effects , Sulfones/pharmacology , Apoptosis/drug effects , DNA Repair/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , HL-60 Cells , Humans , Morpholines/pharmacology , Pyrones/pharmacology
19.
Molecules ; 18(4): 3674-88, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23529028

ABSTRACT

The development of novel antimicrobial agents represents a timely research topic. Eighteen salicylanilide 4-(trifluoromethyl)benzoates were evaluated against Mycobacterium tuberculosis, M. avium and M. kansasii, eight bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA) and for the inhibition of mycobacterial isocitrate lyase. Some compounds were further screened against drug-resistant M. tuberculosis and for their cytotoxicity. Minimum inhibitory concentrations (MICs) for all mycobacterial strains were within 0.5-32 µmol/L, with 4-chloro-2-[4-(trifluoromethyl)phenylcarbamoyl]phenyl 4-(trifluoromethyl)benzoate superiority. Gram-positive bacteria including MRSA were inhibited with MICs ³ 0.49 µmol/L, while Gram-negative ones were much less susceptible. Salicylanilide 4-(trifluoromethyl)benzoates showed significant antibacterial properties, for many strains being comparable to standard drugs (isoniazid, benzylpenicillin) with no cross-resistance. All esters showed mild inhibition of mycobacterial isocitrate lyase and four compounds were comparable to 3-nitropropionic acid without a direct correlation between in vitro MICs and enzyme inhibition.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzoates/pharmacology , Salicylanilides/pharmacology , Isocitrate Lyase/antagonists & inhibitors , Isocitrate Lyase/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Mycobacterium avium/drug effects , Mycobacterium kansasii/drug effects , Mycobacterium tuberculosis/drug effects , Nitro Compounds/pharmacology , Propionates/pharmacology
20.
Biochem Pharmacol ; 216: 115769, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37634597

ABSTRACT

ATP-binding cassette (ABC) drug efflux transporters and drug metabolizing enzymes play crucial roles in pharmacokinetic drug-drug interactions and multidrug tumor resistance (MDR). Tazemetostat (EPZ-6438, Tazverik) is a novel epigenetic drug that has been recently approved for the therapy of advanced epithelioid sarcoma and follicular lymphoma. Additionally, this medication is currently being clinically tested to treat several other cancers such as non-small cell lung cancer (NSCLC). This study aimed to investigate the inhibitory effects of tazemetostat on selected ABC transporters/cytochrome P450 3A4 (CYP3A4) enzyme to comprehensively explore its role in MDR. First, our accumulation and molecular docking studies showed that tazemetostat is a unique triple inhibitor of ABCB1, ABCC1, and ABCG2 transporters. In contrast, tazemetostat exhibited only low level of interaction with the CYP3A4 isozyme. Drug combination assays confirmed that tazemetostat is a multipotent MDR modulator able to synergize with various conventional chemotherapeutics in vitro. Subsequent caspase activity assays and microscopic staining of apoptotic nuclei proved that the effective induction of apoptosis is behind the observed synergies. Notably, a potent MDR-modulatory capacity of tazemetostat was recorded in primary ex vivo NSCLC explants generated from patients' biopsies. On the contrary, its possible position of pharmacokinetic MDR's victim was excluded in comparative proliferation assays. Finally, tested drug has not been identified as an inducer of resistant phenotype in NSCLC cell lines. In conclusion, we demonstrated that tazemetostat is a unique multispecific chemosensitizer, which has strong potential to overcome limitations seen in the era of traditional MDR modulators.

SELECTION OF CITATIONS
SEARCH DETAIL