Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Biomed Pharmacother ; 151: 113124, 2022 Jul.
Article in English | MEDLINE | ID: covidwho-1850707

ABSTRACT

The substrate-analog furin inhibitor MI-1851 can suppress the cleavage of SARS-CoV-2 spike protein and consequently produces significant antiviral effect on infected human airway epithelial cells. In this study, the interaction of inhibitor MI-1851 was examined with human serum albumin using fluorescence spectroscopy and ultrafiltration techniques. Furthermore, the impacts of MI-1851 on human microsomal hepatic cytochrome P450 (CYP) 1A2, 2C9, 2C19, 2D6 and 3A4 activities were assessed based on fluorometric assays. The inhibitory action was also examined on human recombinant CYP3A4 enzyme and on hepatocytes. In addition, microsomal stability (60 min) and cytotoxicity were tested as well. MI-1851 showed no relevant interaction with human serum albumin and was significantly depleted by human microsomes. Furthermore, it did not inhibit CYP1A2, 2C9, 2C19 and 2D6 enzymes. In human hepatocytes, CYP3A4 was significantly suppressed by MI-1851 and weak inhibition was noticed in regard to human microsomes and human recombinant CYP3A4. Finally, MI-1851 did not impair the viability and the oxidative status of primary human hepatocytes (up to 100 µM concentration). Based on these observations, furin inhibitor MI-1851 appears to be potential drug candidates in the treatment of COVID-19, due to the involvement of furin in S protein priming and thus activation of the pandemic SARS-CoV-2.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Furin , Albumins/pharmacology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme Inhibitors/toxicity , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Furin/antagonists & inhibitors , Furin/metabolism , Furin/pharmacology , Humans , Microsomes, Liver , SARS-CoV-2/drug effects , Serum Albumin, Human/metabolism , Spike Glycoprotein, Coronavirus , COVID-19 Drug Treatment
2.
Molecules ; 27(3)2022 Jan 29.
Article in English | MEDLINE | ID: covidwho-1667252

ABSTRACT

Aspirin (also known as acetylsalicylic acid) is a drug intended to treat fever, pain, or inflammation. Treatment of moderate to severe cases of COVID-19 using aspirin along with dexamethasone has gained major attention globally in recent times. Thus, the purpose of this study was to use High-Performance Liquid Chromatography (HPLC) to evaluate the in vitro inhibition of CYP3A2 enzyme activity using aspirin in rat liver microsomes (RLMs). In this study, an efficient and sensitive HPLC method was developed using a reversed phase C18 column (X Bridge 4.6 mm × 150 mm, 3.5 µm) at 243 nm using acetonitrile and water (70:30 v/v). The linearity (r2 > 0.999), precision (<15%), accuracy and recovery (80-120%), limit of detection (5.60 µM and 0.06 µM), limit of quantification (16.98 µM and 0.19 µM), and stability of the newly developed method were validated for dexamethasone and 6ß-hydroxydexamethasone, respectively, following International Conference on Harmonization (ICH) guidelines. This method was applied in vitro to measure CYP3A2 activity. The results showed that aspirin competitively inhibits 6ß-hydroxylation (CYP3A2 activity) with an inhibition constant (Ki) = 95.46 µM and the concentration of the inhibitor causing 50% inhibition of original enzyme activity (IC50) = 190.92 µM. This indicated that there is a minimal risk of toxicity when dexamethasone and aspirin are co-administrated and a very low risk of toxicity and drug interaction with drugs that are a substrate for CYP3A2 in healthcare settings.


Subject(s)
Aspirin/pharmacology , Chromatography, High Pressure Liquid/methods , Cytochrome P-450 CYP3A/metabolism , Animals , Aspirin/chemistry , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Male , Microsomes, Liver/metabolism , Pharmaceutical Preparations/metabolism , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Reproducibility of Results , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , COVID-19 Drug Treatment
3.
Drug Deliv ; 28(1): 1150-1165, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1271847

ABSTRACT

The objective of this paper is to confine piperine, a poor oral bioavailable herbal drug into bile salt based nano vesicles for improving its aqueous solubility, hence, its therapeutic activity. Piperine-loaded bilosomes were fabricated adopting thin film hydration technique according to 32.21 full factorial design to investigate the impact of different formulation variables on the characters of bilosomes: entrapment efficiency (EE%), particle size, and % of drug released post 8 h (Q8hr). The selected optimum formula was F2 (enclosing 1% bile salt, brij72 as a surfactant, and ratio of surfactant:cholesterol was 9:1) with desirability value 0.801, exhibiting high EE% (97.2 ± 0.8%) nanosized spherical vesicles (220.2 ± 20.5 nm) and Q8hr (88.2%±5.6). The superiority of the optimized formula (F2) over the drug suspension was revealed via ex vivo permeation study, also pharmacokinetic study denoted to the boosted oral bioavailability of piperine-loaded bilosome compared to piperine suspension. Moreover, antiviral activity and safety margin of F2 was significantly higher than that of the drug suspension. The ability of piperine to interact with the key amino acids in the receptor binding domain 4L3N as indicated by its docking configuration, rationalized its observed activity. Furthermore, F2 significantly reduce oxidant markers, inflammatory cytokines in MERS-CoV-infected mice. Hence, bilosomes can be considered as a carrier of choice for piperine with potential antiviral and anti-inflammatory activities.


Subject(s)
Alkaloids , Benzodioxoles , Bile Acids and Salts/pharmacokinetics , Drug Delivery Systems/methods , Middle East Respiratory Syndrome Coronavirus/drug effects , Piperidines , Polyunsaturated Alkamides , Administration, Oral , Alkaloids/administration & dosage , Alkaloids/pharmacokinetics , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacokinetics , Biological Availability , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Drug Liberation , Liposomes , Mice , Molecular Docking Simulation , Nanostructures , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Plants, Medicinal , Polyunsaturated Alkamides/administration & dosage , Polyunsaturated Alkamides/pharmacokinetics , Surface-Active Agents/pharmacokinetics
4.
Curr Drug Metab ; 21(14): 1127-1135, 2020.
Article in English | MEDLINE | ID: covidwho-968953

ABSTRACT

BACKGROUND: In clinical practice, chloroquine and hydroxychloroquine are often co-administered with other drugs in the treatment of malaria, chronic inflammatory diseases, and COVID-19. Therefore, their metabolic properties and the effects on the activity of cytochrome P450 (P450, CYP) enzymes and drug transporters should be considered when developing the most efficient treatments for patients. METHODS: Scientific literature on the interactions of chloroquine and hydroxychloroquine with human P450 enzymes and drug transporters, was searched using PUBMED.Gov (https://pubmed.ncbi.nlm.nih.gov/) and the ADME database (https://life-science.kyushu.fujitsu.com/admedb/). RESULTS: Chloroquine and hydroxychloroquine are metabolized by P450 1A2, 2C8, 2C19, 2D6, and 3A4/5 in vitro and by P450s 2C8 and 3A4/5 in vivo by N-deethylation. Chloroquine effectively inhibited P450 2D6 in vitro; however, in vivo inhibition was not apparent except in individuals with limited P450 2D6 activity. Chloroquine is both an inhibitor and inducer of the transporter MRP1 and is also a substrate of the Mate and MRP1 transport systems. Hydroxychloroquine also inhibited P450 2D6 and the transporter OATP1A2. CONCLUSIONS: Chloroquine caused a statistically significant decrease in P450 2D6 activity in vitro and in vivo, also inhibiting its own metabolism by the enzyme. The inhibition indicates a potential for clinical drug-drug interactions when taken with other drugs that are predominant substrates of the P450 2D6. When chloroquine and hydroxychloroquine are used clinically with other drugs, substrates of P450 2D6 enzyme, attention should be given to substrate-specific metabolism by P450 2D6 alleles present in individuals taking the drugs.


Subject(s)
Chloroquine/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Hydroxychloroquine/metabolism , Membrane Transport Proteins/metabolism , Animals , COVID-19/metabolism , Chloroquine/therapeutic use , Cytochrome P-450 Enzyme Inhibitors/therapeutic use , Drug Interactions/physiology , Humans , Hydroxychloroquine/therapeutic use , Pharmaceutical Preparations/metabolism , COVID-19 Drug Treatment
5.
Curr Heart Fail Rep ; 17(6): 365-383, 2020 12.
Article in English | MEDLINE | ID: covidwho-834057

ABSTRACT

PURPOSE OF REVIEW: Cardiotoxicity by anticancer agents has emerged as a multifaceted issue and is expected to affect both mortality and morbidity. This review summarizes clinical challenges in the management of oncological patients requiring anticoagulants for atrial fibrillation (AF) also considering the current outbreak of the COVID-19 (coronavirus disease 2019) pandemic, since this infection can add challenges to the management of both conditions. Specifically, the aims are manyfold: (1) describe the evolving use of direct oral anticoagulants (DOACs) in AF patients with cancer; (2) critically appraise the risk of clinically important drug-drug interactions (DDIs) between DOACs and oral targeted anticancer agents; (3) address expected DDIs between DOACs and candidate anti-COVID drugs, with implications on management of the underlying thrombotic risk; and (4) characterize the proarrhythmic liability in cardio-oncology in the setting of COVID-19, focusing on QT prolongation. RECENT FINDINGS: AF in cardio-oncology poses diagnostic and management challenges, also due to the number of anticancer drugs recently associated with AF onset/worsening. Oral targeted drugs can potentially interact with DOACs, with increased bleeding risk mainly due to pharmacokinetic DDIs. Moreover, the vast majority of oral anticancer agents cause QT prolongation with direct and indirect mechanisms, potentially resulting in the occurrence of torsade de pointes, especially in susceptible patients with COVID-19 receiving additional drugs with QT liability. Oncologists and cardiologists must be aware of the increased bleeding risk and arrhythmic susceptibility of patients with AF and cancer due to DDIs. High-risk individuals with COVID-19 should be prioritized to target preventive strategies, including optimal antithrombotic management, medication review, and stringent monitoring.


Subject(s)
Antineoplastic Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Atrial Fibrillation/drug therapy , COVID-19 Drug Treatment , Factor Xa Inhibitors/adverse effects , Hemorrhage/chemically induced , Neoplasms/drug therapy , Thromboembolism/prevention & control , Atrial Fibrillation/complications , COVID-19/complications , Cytochrome P-450 Enzyme Inhibitors/adverse effects , Cytochrome P-450 Enzyme System , Drug Interactions , Humans , Long QT Syndrome/chemically induced , Neoplasms/complications , Thromboembolism/etiology
SELECTION OF CITATIONS
SEARCH DETAIL