Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Drug Metab Rev ; 56(2): 127-144, 2024.
Article in English | MEDLINE | ID: mdl-38445647

ABSTRACT

Severe acute respiratory syndrome 2 (SARS-CoV-2) caused the emergence of the COVID-19 pandemic all over the world. Several studies have suggested that antiviral drugs such as favipiravir (FAV), remdesivir (RDV), and lopinavir (LPV) may potentially prevent the spread of the virus in the host cells and person-to-person transmission. Simultaneously with the widespread use of these drugs, their stability and action mechanism studies have also attracted the attention of many researchers. This review focuses on the action mechanism, metabolites and degradation products of these antiviral drugs (FAV, RDV and LPV) and demonstrates various methods for their quantification and discrimination in the different biological samples. Herein, the instrumental methods for analysis of the main form of drugs or their metabolite and degradation products are classified into two types: optical and chromatography methods which the last one in combination with various detectors provides a powerful method for routine and stability analyses. Some representative studies are reported in this review and the details of them are carefully explained. It is hoped that this review will be a good guideline study and provide a better understanding of these drugs from the aspects investigated in this study.


Subject(s)
Adenosine Monophosphate , Adenosine Monophosphate/analogs & derivatives , Alanine , Alanine/analogs & derivatives , Amides , Antiviral Agents , COVID-19 Drug Treatment , Lopinavir , Pyrazines , Pyrazines/metabolism , Amides/metabolism , Amides/chemistry , Antiviral Agents/pharmacology , Adenosine Monophosphate/metabolism , Humans , Alanine/metabolism , Lopinavir/therapeutic use , Lopinavir/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Animals
2.
Eur Rev Med Pharmacol Sci ; 26(18): 6845-6855, 2022 09.
Article in English | MEDLINE | ID: mdl-36196733

ABSTRACT

OBJECTIVE: This study was conducted to examine whether lopinavir/ritonavir (Lop/r), an HIV protease inhibitor, can improve disc physiology and slow down intervertebral disc (IVD) degeneration through in vitro experimental methods, as well as whether it can suppress inflammation with interleukin-1 beta (IL-1ß) and sex-determining region Y (SRY) protein-related high-mobility group box genes-9 (SOX9) through hypoxia-inducible factor 1-alpha (HIF-1α) and the nuclear factor kappa B (NF-κB) signaling pathway. The aim was to investigate whether Lop/r application is toxic to IVD cells and the microenvironment simultaneously. PATIENTS AND METHODS: Human primary cell cultures were prepared using herniated IVD tissues obtained from patients with lumbar disc hernia who were unresponsive to conservative and medical treatment, and thereby, were operated on. The untreated culture samples served as control group, and the samples treated with Lop/r served as study group. Microscopic evaluations were performed simultaneously using fluorescent and supravital dyes in all groups. In addition to cell viability, toxicity, and proliferation analysis through a commercial kit, IL-1ß, SOX9, HIF-1α, and NF-κB protein expressions were evaluated using Western blotting. In the statistical comparison of the obtained data, an alpha value less than 0.05 was considered significant. RESULTS: Cell proliferation decreased in the Lop/r group, but no cell death was observed (p < 0.05). Moreover, at the end of 72 hours after Lop/r application, IL-1ß and NF-kB protein expressions decreased by 40% and 52%, respectively, while HIF-1α and SOX9 protein expressions increased by 4% and 59%, respectively (p< 0.05). CONCLUSIONS: Although these data were obtained from an in vitro experimental study, it is believed that these findings could make significant contributions to the pharmaco-regenerative treatment modalities of IVD degeneration. Lop/r suppresses the IL-1ß and NF-κB and induces SOX9 and HIF-1α, since these signaling pathways may be related to human IVD degeneration.


Subject(s)
HIV Protease Inhibitors , Intervertebral Disc Degeneration , Intervertebral Disc , Nucleus Pulposus , Cells, Cultured , Coloring Agents/metabolism , Coloring Agents/pharmacology , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacology , Humans , Hypoxia-Inducible Factor 1/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Lopinavir/metabolism , NF-kappa B/metabolism , Nucleus Pulposus/metabolism , Ritonavir , Signal Transduction
3.
Adv Sci (Weinh) ; 9(30): e2203388, 2022 10.
Article in English | MEDLINE | ID: mdl-36055796

ABSTRACT

Coronavirus disease 2019 continues to spread worldwide. Given the urgent need for effective treatments, many clinical trials are ongoing through repurposing approved drugs. However, clinical data regarding the cardiotoxicity of these drugs are limited. Human pluripotent stem cell-derived cardiomyocytes (hCMs) represent a powerful tool for assessing drug-induced cardiotoxicity. Here, by using hCMs, it is demonstrated that four antiviral drugs, namely, apilimod, remdesivir, ritonavir, and lopinavir, exhibit cardiotoxicity in terms of inducing cell death, sarcomere disarray, and dysregulation of calcium handling and contraction, at clinically relevant concentrations. Human engineered heart tissue (hEHT) model is used to further evaluate the cardiotoxic effects of these drugs and it is found that they weaken hEHT contractile function. RNA-seq analysis reveals that the expression of genes that regulate cardiomyocyte function, such as sarcomere organization (TNNT2, MYH6) and ion homeostasis (ATP2A2, HCN4), is significantly altered after drug treatments. Using high-throughput screening of approved drugs, it is found that ceftiofur hydrochloride, astaxanthin, and quetiapine fumarate can ameliorate the cardiotoxicity of remdesivir, with astaxanthin being the most prominent one. These results warrant caution and careful monitoring when prescribing these therapies in patients and provide drug candidates to limit remdesivir-induced cardiotoxicity.


Subject(s)
COVID-19 Drug Treatment , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/physiology , Calcium/metabolism , Lopinavir/metabolism , Lopinavir/pharmacology , Ritonavir/metabolism , Ritonavir/pharmacology , Quetiapine Fumarate/metabolism , Quetiapine Fumarate/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Pluripotent Stem Cells/metabolism , Antiviral Agents/adverse effects
4.
SAR QSAR Environ Res ; 32(11): 889-915, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34551634

ABSTRACT

HIV-1 protease (PR) is thought to be efficient targets of anti-AIDS drug design. Molecular dynamics (MD) simulations and multiple post-processing analysis technologies were applied to decipher molecular mechanism underlying binding of three drugs Lopinavir (LPV), Nelfinavir (NFV) and Atazanavir (ATV) to the PR. Binding free energies calculated by molecular mechanics generalized Born surface area (MM-GBSA) suggest that compensation between binding enthalpy and entropy plays a vital role in binding of drugs to PR. Dynamics analyses show that binding of LPV, NFV and ATV highly affects structural flexibility, motion modes and dynamics behaviour of the PR, especially for two flaps. Computational alanine scanning and interaction network analysis verify that although three drugs have structural difference, they share similar binding modes to the PR and common interaction clusters with the PR. The current findings also confirm that residues located interaction clusters, such as Asp25/Asp25', Gly27/Gly27', Ala28/Ala28', Asp29, Ile47/Ile47', Gly49/Gly49', Ile50/Ile50', Val82/Val82' and Ile84/Ile84, can be used as efficient targets of clinically available inhibitors towards the PR.


Subject(s)
Anti-HIV Agents/metabolism , Atazanavir Sulfate/metabolism , HIV Protease/metabolism , Lopinavir/metabolism , Molecular Dynamics Simulation , Nelfinavir/metabolism , Binding Sites
5.
Sci Rep ; 11(1): 17810, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34497279

ABSTRACT

Transporters in the human liver play a major role in the clearance of endo- and xenobiotics. Apical (canalicular) transporters extrude compounds to the bile, while basolateral hepatocyte transporters promote the uptake of, or expel, various compounds from/into the venous blood stream. In the present work we have examined the in vitro interactions of some key repurposed drugs advocated to treat COVID-19 (lopinavir, ritonavir, ivermectin, remdesivir and favipiravir), with the key drug transporters of hepatocytes. These transporters included ABCB11/BSEP, ABCC2/MRP2, and SLC47A1/MATE1 in the canalicular membrane, as well as ABCC3/MRP3, ABCC4/MRP4, SLC22A1/OCT1, SLCO1B1/OATP1B1, SLCO1B3/OATP1B3, and SLC10A1/NTCP, residing in the basolateral membrane. Lopinavir and ritonavir in low micromolar concentrations inhibited BSEP and MATE1 exporters, as well as OATP1B1/1B3 uptake transporters. Ritonavir had a similar inhibitory pattern, also inhibiting OCT1. Remdesivir strongly inhibited MRP4, OATP1B1/1B3, MATE1 and OCT1. Favipiravir had no significant effect on any of these transporters. Since both general drug metabolism and drug-induced liver toxicity are strongly dependent on the functioning of these transporters, the various interactions reported here may have important clinical relevance in the drug treatment of this viral disease and the existing co-morbidities.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Antiviral Agents/pharmacology , Liver-Specific Organic Anion Transporter 1/metabolism , Liver/drug effects , Organic Cation Transport Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/metabolism , Alanine/pharmacology , Alanine/therapeutic use , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Comorbidity , Drug Repositioning , Humans , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Lopinavir/chemistry , Lopinavir/metabolism , Lopinavir/pharmacology , Lopinavir/therapeutic use , Multidrug Resistance-Associated Protein 2 , Organic Cation Transport Proteins/antagonists & inhibitors , Ritonavir/chemistry , Ritonavir/metabolism , Ritonavir/pharmacology , Ritonavir/therapeutic use , SARS-CoV-2/isolation & purification , Substrate Specificity , COVID-19 Drug Treatment
6.
Eur Rev Med Pharmacol Sci ; 25(11): 4163-4173, 2021 06.
Article in English | MEDLINE | ID: mdl-34156698

ABSTRACT

OBJECTIVE: As a beta-coronavirus, Coronavirus disease-2019 (COVID-19) has caused one of the most significant historical pandemics, as well as various health and medical challenges. Our purpose in this report is to collect, summarize, and articulate all essential information about antiviral drugs that may or may not be efficient for treating COVID-19. Clinical evidence about these drugs and their possible mechanisms of action are also discussed. MATERIALS AND METHODS: To conduct a comprehensive review, different keywords in various databases, including Web of Science, Scopus, Medline, PubMed, and Google Scholar, were searched relevant articles, especially the most recent ones, were selected and studied. These selected original research articles, review papers, systematic reviews, and even letters to the editors were then carefully reviewed for data collection. RESULTS: SARS-CoV-2 is the newest member of the coronavirus family, and there are still no promising therapies or particular antiviral compounds to fight it. After entering the body, SARS-CoV-2 penetrates the cells by attaching to specific lung cell receptors, called angiotensin-converting enzyme-2. Then, by employing cell division machinery, it replicates through a complex mechanism and spreads throughout the patient's body. Various antiviral drugs, including anti-influenza/HIV/HCV drugs, have been applied for treating COVID-19 patients. Due to the similarity of the structure and transcriptional mechanism of COVID-19 to a number of viruses, some of the listed drugs have been beneficial against SARS-CoV-2. However, the effectiveness of others is in an aura of ambiguity and doubt. CONCLUSIONS: Some of the antiviral medications listed and discussed in this article have been effective in the treatment of COVID-19 patients or preventing the virus from spreading further. However, other drugs have to be investigated to reach a reliable conclusion about their effectiveness or ineffectiveness.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , COVID-19/epidemiology , Data Analysis , SARS-CoV-2/drug effects , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , COVID-19/metabolism , Humans , Lopinavir/metabolism , Lopinavir/pharmacology , Lopinavir/therapeutic use , SARS-CoV-2/metabolism
7.
Molecules ; 26(7)2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33916461

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent for the COVID-19 pandemic, which generated more than 1.82 million deaths in 2020 alone, in addition to 83.8 million infections. Currently, there is no antiviral medication to treat COVID-19. In the search for drug leads, marine-derived metabolites are reported here as prospective SARS-CoV-2 inhibitors. Two hundred and twenty-seven terpene natural products isolated from the biodiverse Red-Sea ecosystem were screened for inhibitor activity against the SARS-CoV-2 main protease (Mpro) using molecular docking and molecular dynamics (MD) simulations combined with molecular mechanics/generalized Born surface area binding energy calculations. On the basis of in silico analyses, six terpenes demonstrated high potency as Mpro inhibitors with ΔGbinding ≤ -40.0 kcal/mol. The stability and binding affinity of the most potent metabolite, erylosides B, were compared to the human immunodeficiency virus protease inhibitor, lopinavir. Erylosides B showed greater binding affinity towards SARS-CoV-2 Mpro than lopinavir over 100 ns with ΔGbinding values of -51.9 vs. -33.6 kcal/mol, respectively. Protein-protein interactions indicate that erylosides B biochemical signaling shares gene components that mediate severe acute respiratory syndrome diseases, including the cytokine- and immune-signaling components BCL2L1, IL2, and PRKC. Pathway enrichment analysis and Boolean network modeling were performed towards a deep dissection and mining of the erylosides B target-function interactions. The current study identifies erylosides B as a promising anti-COVID-19 drug lead that warrants further in vitro and in vivo testing.


Subject(s)
Invertebrates/chemistry , SARS-CoV-2/metabolism , Terpenes/chemistry , Viral Matrix Proteins/antagonists & inhibitors , Animals , Binding Sites , COVID-19/virology , Humans , Hydrogen Bonding , Invertebrates/metabolism , Lopinavir/chemistry , Lopinavir/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/isolation & purification , Protease Inhibitors/therapeutic use , Protein Binding , SARS-CoV-2/isolation & purification , Terpenes/isolation & purification , Terpenes/metabolism , Terpenes/therapeutic use , Thermodynamics , Viral Matrix Proteins/metabolism , COVID-19 Drug Treatment
8.
Virology ; 555: 10-18, 2021 03.
Article in English | MEDLINE | ID: mdl-33421743

ABSTRACT

Novel coronavirus (SARS-CoV-2), turned out to be a global pandemic with unstoppable morbidity and mortality rate. However, till date there is no effective treatment found against SARS-CoV-2. We report on the major in-depth molecular and docking analysis by using antiretroviral (Lopinavir and ritonavir), antimalarial (Hydroxychloroquine), antibiotics (Azithromycin), and dietary supplements (Vitamin C and E) to provide new insight into drug repurposing molecular events involved in SARS-CoV-2. We constructed three drug-target-pathways-disease networks to predict the targets and drugs interactions as well as important pathways involved in SARS-CoV-2. The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Gene ontology biological process analysis further confirmed multiple viral infection-related processes (P < 0.001), including viral life cycle, modulation by virus, C-C chemokine receptor activity, and platelet activation. KEGG pathway analysis involves multiple pathways (P < 0.05), including FoxO, GnRH, ErbB, Neurotrophin, Toll-like receptor, IL-17, TNF, Insulin, HIF-1, JAK-STAT, Estrogen, NF-kappa, Chemokine, VEGF, and Thyroid hormone signaling pathway in SARS-CoV-2. Docking study was carried out to predict the molecular mechanism Thus, the potential drug combinations could reduce viral infectivity, viral replication, and abnormal host inflammatory responses and may be useful for multi-target drugs against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Drug Repositioning , SARS-CoV-2/drug effects , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , COVID-19/virology , Drug Development , Drug Therapy, Combination , Humans , Hydroxychloroquine/metabolism , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Lopinavir/metabolism , Lopinavir/pharmacology , Lopinavir/therapeutic use , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Ritonavir/metabolism , Ritonavir/pharmacology , Ritonavir/therapeutic use , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Signal Transduction , Virus Replication/drug effects
9.
Sci Rep ; 10(1): 16986, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046764

ABSTRACT

We performed molecular dynamics simulation of the dimeric SARS-CoV-2 (severe acute respiratory syndrome corona virus 2) main protease (Mpro) to examine the binding dynamics of small molecular ligands. Seven HIV inhibitors, darunavir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir, and tipranavir, were used as the potential lead drugs to investigate access to the drug binding sites in Mpro. The frequently accessed sites on Mpro were classified based on contacts between the ligands and the protein, and the differences in site distributions of the encounter complex were observed among the ligands. All seven ligands showed binding to the active site at least twice in 28 simulations of 200 ns each. We further investigated the variations in the complex structure of the active site with the ligands, using microsecond order simulations. Results revealed a wide variation in the shapes of the binding sites and binding poses of the ligands. Additionally, the C-terminal region of the other chain often interacted with the ligands and the active site. Collectively, these findings indicate the importance of dynamic sampling of protein-ligand complexes and suggest the possibilities of further drug optimisations.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Cysteine Endopeptidases/metabolism , Drug Repositioning/methods , HIV Protease Inhibitors/pharmacology , Pneumonia, Viral/drug therapy , Viral Nonstructural Proteins/metabolism , Betacoronavirus/metabolism , Binding Sites/drug effects , Biophysical Phenomena , COVID-19 , Catalytic Domain/drug effects , Computational Biology , Coronavirus 3C Proteases , Darunavir/metabolism , Darunavir/pharmacology , HIV Protease Inhibitors/metabolism , Humans , Indinavir/metabolism , Indinavir/pharmacology , Lopinavir/metabolism , Lopinavir/pharmacology , Molecular Dynamics Simulation , Nelfinavir/metabolism , Nelfinavir/pharmacology , Pandemics , Ritonavir/metabolism , Ritonavir/pharmacology , SARS-CoV-2 , Saquinavir/metabolism , Saquinavir/pharmacology
10.
Pharm Res ; 37(7): 121, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32514792

ABSTRACT

PURPOSE: MDCK cells are commonly used to assess drug permeability, but the existence of various strains merits a comparative functional study. Since metformin absorption is largely mediated by transporters and paracellular diffusion, we used it to functionally compare MDCK-wt and MDCK-II. METHODS: Uptake, bidirectional transport and efflux experiments were performed using different buffers, pH, and a panel of transporter inhibitors. Relative contributions to total transport in both strains were estimated. RESULTS: Metformin uptake into MDCK-wt was linear but saturable in MDCK-II. Uptake into MDCK-wt or -II was promoted at pH 5.4 or 8.4, respectively. Quinidine and cimetidine similarly inhibited uptake in both strains. Lopinavir (PMAT specific) at pH 5.4 or pyrimethamine (MATE specific) at pH 8.4 differentially inhibited MDCK-wt or -II, respectively. Transport at pH 7.4 was absorptive regardless of strains, but secretory (MDCK-II) or absorptive (MDCK-wt) at pH 5.4. Efflux was largely basolateral in both strains. While paracellular permeability was similar between strains, total transport was dominated by transporters in MDCK-II or paracellular diffusion in MDCK-wt. CONCLUSIONS: Metformin transport revealed functional differences between MDCK strains. Apical uptake was governed by MATE in MDCK-II or PMAT in MDCK-wt, such that metformin transport was either secretory or absorptive, respectively.


Subject(s)
Metformin/metabolism , Animals , Biofilms , Biological Transport/drug effects , Cell Adhesion , Cells, Cultured , Cimetidine/metabolism , Diffusion , Dogs , Humans , Hydrogen-Ion Concentration , Lopinavir/metabolism , Madin Darby Canine Kidney Cells , Pyrimethamine/metabolism , Quinidine/metabolism
12.
J Chem Theory Comput ; 14(6): 3218-3227, 2018 Jun 12.
Article in English | MEDLINE | ID: mdl-29712434

ABSTRACT

We present a general method called atom-wise free energy perturbation (AFEP), which extends a conventional molecular dynamics free energy perturbation (FEP) simulation to give the contribution to a free energy change from each atom. AFEP is derived from an expansion of the Zwanzig equation used in the exponential averaging method by defining that the system total energy can be partitioned into contributions from each atom. A partitioning method is assumed and used to group terms in the expansion to correspond to individual atoms. AFEP is applied to six example free energy changes to demonstrate the method. Firstly, the hydration free energies of methane, methanol, methylamine, methanethiol, and caffeine in water. AFEP highlights the atoms in the molecules that interact favorably or unfavorably with water. Finally AFEP is applied to the binding free energy of human immunodeficiency virus type 1 protease to lopinavir, and AFEP reveals the contribution of each atom to the binding free energy, indicating candidate areas of the molecule to improve to produce a more strongly binding inhibitor. FEP gives a single value for the free energy change and is already a very useful method. AFEP gives a free energy change for each "part" of the system being simulated, where part can mean individual atoms, chemical groups, amino acids, or larger partitions depending on what the user is trying to measure. This method should have various applications in molecular dynamics studies of physical, chemical, or biochemical phenomena, specifically in the field of computational drug discovery.


Subject(s)
Molecular Dynamics Simulation , Water/chemistry , Caffeine/chemistry , HIV Protease/chemistry , HIV Protease/metabolism , HIV-1/enzymology , Humans , Lopinavir/chemistry , Lopinavir/metabolism , Methane/chemistry , Methylamines/chemistry , Protein Binding , Sulfhydryl Compounds/chemistry , Thermodynamics
13.
Pharm Res ; 33(11): 2713-21, 2016 11.
Article in English | MEDLINE | ID: mdl-27431863

ABSTRACT

PURPOSE: Macrophages are an important cellular reservoir in HIV, and exist in two phenotypically dissimilar subsets, the pro-inflammatory M1 phenotype, and the anti-inflammatory M2 phenotype. The role of these two subsets is uncertain. We hypothesized that differences in drug efflux transporters exist between the subsets, which would result in altered intracellular drug concentrations between these cells. METHODS: U937 monocytic cells were polarized to the M1 or M2 phenotype via treatment with interferon-gamma and LPS, or interleukins 4, 13, and LPS, respectively. PGP function was assessed with Hoechst 33342, and expression via western blotting. Intracellular lopinavir was assessed via LC-MS/MS. Data was confirmed with primary monocyte derived macrophages. RESULTS: We observed significant differences in intracellular concentrations of lopinavir, a PGP substrate, with higher concentrations in M1 cells. PGP function and expression was higher in the M2 macrophages. These results were confirmed with primary monocyte derived macrophages. CONCLUSIONS: This data shows that there are previously unreported differences in P-glycoprotein expression between macrophage subsets, and suggests that there may be differences for other transporters. These differences can play a role in intracellular drug concentrations in these cells, and may allow for low-level HIV replication.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Macrophages/metabolism , Anti-HIV Agents/metabolism , Arginase/metabolism , Benzimidazoles/chemistry , Cell Polarity , Cytokines/pharmacology , Fluorescent Dyes/chemistry , Humans , Intracellular Space/metabolism , Lopinavir/metabolism , Macrophages/cytology , Phenotype , U937 Cells
14.
Drug Deliv ; 23(9): 3492-3504, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27297453

ABSTRACT

Nanoparticles (NPs) can be absorbed via M cells of Peyer's patches after oral delivery leading to passive lymphatic targeting followed by systemic drug delivery. Hence, the study was aimed to formulate PLGA NPs of lopinavir. The NPs were prepared by nanoprecipitation, optimized by 33 factorial design and characterized by TEM, DSC, FTIR studies and safety was assessed by MTT assay. In vivo pharmacokinetic studies were performed in rats. The NPs were discrete spherical structures having particle size of 142.1 ± 2.13 nm and entrapment of 93.03 ± 1.27%. There was absence of drug-polymer interaction. Confocal images revealed the penetration and absorption of coumarin-loaded NPs in Caco-2 cells and intestine after oral delivery. There was 3.04 folds permeability and 13.9 folds bioavailability enhancement from NPs. The NPs can be promising delivery system for antiretroviral drug by delivering the drug to lymph (major HIV reservoir site) via direct absorption through intestine before reaching systemic circulation.


Subject(s)
Intestinal Mucosa/metabolism , Lactic Acid/chemistry , Lopinavir/administration & dosage , Lopinavir/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Polyglycolic Acid/chemistry , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Intestinal Absorption/physiology , Lopinavir/chemistry , Male , Nanoparticles/administration & dosage , Particle Size , Permeability , Peyer's Patches/chemistry , Peyer's Patches/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Wistar
15.
Drug Deliv ; 23(8): 3027-3042, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26882014

ABSTRACT

OBJECTIVE: Lopinavir (LPV), an antiretroviral protease inhibitor shows poor bioavailability because of poor aqueous solubility and extensive hepatic first-pass metabolism. The aim of the present work was to investigate the potential of the solid self-nanoemulsifying drug delivery system (S-SNEDDS) in improving dissolution rate and oral bioavailability of LPV. MATERIALS AND METHODS: Liquid SNEDDS (L-SNEDDS) of LPV were prepared using Capmul MCM C8, Cremophor RH 40 and propylene glycol and their amounts were optimized by Scheffe's mixture design. L-SNEDDS formulations were evaluated for different physicochemical and in vitro drug release parameters. S-SNEDDS were prepared by adsorbing L-SNEDDS on Neusilin US2 and characterized for solid-state properties. In vivo bioavailability of S-SNEDDS, marketed Lopinavir + Ritonavir (LPV/RTV) formulation and pure LPV was studied in Wistar rats. Stability study of S-SNEDDS was performed as per ICH guidelines. RESULTS AND DISCUSSION: Optimized L-SNEDDS obtained by Scheffe design had drug loading 160 ± 1.15 mg, globule size 32.9 ± 1.45 nm and drug release >95% within 15 min. Solid state studies suggested the transformation of the crystalline drug to amorphous drug. The size and zeta potential of globules obtained on dilution S-SNEDDS remained similar to L-SNEEDS. In vivo bioavailability study revealed that S-SNEDDS has 2.97 and 1.54-folds higher bioavailability than pure LPV and LPV/RTV formulation, respectively. The optimized S-SNEDDS was found to be stable and had a shelf life of 2.85 years. CONCLUSION: The significant increase in drug dissolution and bioavailability by prepared SNEDDS suggest that the developed S-SNEDDS is a useful solid platform for improving oral bioavailability of poorly soluble LPV.


Subject(s)
Emulsions/chemistry , Lopinavir/chemistry , Nanoparticles/chemistry , Administration, Oral , Animals , Biological Availability , Chemistry, Pharmaceutical/methods , Drug Carriers/chemistry , Drug Delivery Systems/methods , Drug Liberation , Drug Stability , Female , Lopinavir/metabolism , Particle Size , Polyethylene Glycols/chemistry , Rats , Rats, Wistar , Solubility
16.
PLoS One ; 11(1): e0146529, 2016.
Article in English | MEDLINE | ID: mdl-26741368

ABSTRACT

Cytochrome P450 3A4 (CYP3A4) is the major drug metabolic enzyme, and is involved in the metabolism of antiretroviral drugs, especially protease inhibitors (PIs). This study was undertaken to examine the effect of methamphetamine on the binding and metabolism of PIs with CYP3A4. We showed that methamphetamine exhibits a type I spectral change upon binding to CYP3A4 with δAmax and KD of 0.016±0.001 and 204±18 µM, respectively. Methamphetamine-CYP3A4 docking showed that methamphetamine binds to the heme of CYP3A4 in two modes, both leading to N-demethylation. We then studied the effect of methamphetamine binding on PIs with CYP3A4. Our results showed that methamphetamine alters spectral binding of nelfinavir but not the other type I PIs (lopinavir, atazanavir, tipranavir). The change in spectral binding for nelfinavir was observed at both δAmax (0.004±0.0003 vs. 0.0068±0.0001) and KD (1.42±0.36 vs.2.93±0.08 µM) levels. We further tested effect of methamphetamine on binding of 2 type II PIs; ritonavir and indinavir. Our results showed that methamphetamine alters the ritonavir binding to CYP3A4 by decreasing both the δAmax (0.0038±0.0003 vs. 0.0055±0.0003) and KD (0.043±0.0001 vs. 0.065±0.001 nM), while indinavir showed only reduced KD in presence of methamphetamine (0.086±0.01 vs. 0.174±0.03 nM). Furthermore, LC-MS/MS studies in high CYP3A4 human liver microsomes showed a decrease in the formation of hydroxy ritonavir in the presence of methamphetamine. Finally, CYP3A4 docking with lopinavir and ritonavir in the absence and presence of methamphetamine showed that methamphetamine alters the docking of ritonavir, which is consistent with the results obtained from spectral binding and metabolism studies. Overall, our results demonstrated differential effects of methamphetamine on the binding and metabolism of PIs with CYP3A4. These findings have clinical implication in terms of drug dose adjustment of antiretroviral medication, especially with ritonavir-boosted antiretroviral therapy, in HIV-1-infected individuals who abuse methamphetamine.


Subject(s)
Cytochrome P-450 CYP3A/chemistry , HIV Infections/drug therapy , HIV Protease Inhibitors/chemistry , Methamphetamine/chemistry , Amphetamine-Related Disorders/enzymology , Atazanavir Sulfate/chemistry , Atazanavir Sulfate/metabolism , Atazanavir Sulfate/pharmacology , Catalytic Domain , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , HIV Protease Inhibitors/pharmacology , Humans , Inactivation, Metabolic , Lopinavir/chemistry , Lopinavir/metabolism , Lopinavir/pharmacology , Methamphetamine/pharmacology , Microsomes, Liver/enzymology , Molecular Docking Simulation , Nelfinavir/chemistry , Nelfinavir/metabolism , Nelfinavir/pharmacology , Protein Binding , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology , Pyrones/chemistry , Pyrones/metabolism , Pyrones/pharmacology , Sulfonamides
17.
J Pharm Sci ; 104(9): 2977-85, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25989422

ABSTRACT

In vivo activities of absorption enhancers coencapsulated with poorly absorptive drugs in the same enteric-coated particles were evaluated. Lopinavir [a substrate of cytochrome P450 3A (CYP3A)] and ritonavir (an inhibitor of CYP3A-mediatd metabolism) were used as a model drug and a model absorption enhancer, respectively. Lopinavir and ritonavir were encapsulated into enteric-coated particles as amorphous forms using coaxial electrospray deposition. The electrospray treatment resulted in dramatic improvement of dissolution profiles of both compounds, probably because of complete amorphization and superior dispersion efficiency of the particles. Poor absorption of lopinavir in rats was observed after oral administration of enteric-coated particles containing lopinavir alone. When the particles were coadministered with enteric-coated particles containing ritonavir alone, lopinavir absorption was boosted. The boosting effect was further enhanced when ritonavir was coencapsulated with lopinavir into the same enteric-coated particles. A significant increase in area under the plasma concentration-time curve reflected an extension of mean residence time rather than an elevation of Cmax . Lopinavir absorption was improved presumably because lopinavir was always accompanied by a practical amount of ritonavir required for the boosting during the gastrointestinal transit of the particles. Not only did the electrospray coencapsulation technique improve drug absorption, but also increased trough concentration that might result in the reduction of the number of doses.


Subject(s)
Lopinavir/chemistry , Lopinavir/metabolism , Ritonavir/chemistry , Ritonavir/metabolism , Administration, Oral , Animals , Drug Therapy, Combination/methods , Gastrointestinal Transit/physiology , Male , Rats , Rats, Sprague-Dawley , Technology, Pharmaceutical/methods
18.
Drug Dev Ind Pharm ; 41(1): 131-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24180260

ABSTRACT

Lopinavir (LPV)-loaded poly-ε-caprolactone (PCL) nanoparticles (NPs) were prepared by emulsion solvent evaporation technique. Effects of various critical factors in preparation of loaded NPs were investigated. Box-Behnken design (BBD) was employed to optimize particle size and entrapment efficiency (EE) of loaded NPs. Optimized LPV NPs exhibited nanometeric size (195.3 nm) with high EE (93.9%). In vitro drug release study showed bi-phasic sustained release behavior of LPV from NPs. Pharmacokinetic study results in male Wistar rats indicated an increase in oral bioavailability of LPV by 4-folds after incorporation into PCL NPs. From tissue distribution studies, significant accumulation of loaded NPs in tissues like liver and spleen indicated possible involvement of lymphatic route in absorption of NPs. Mechanistic studies using rat everted gut sac model revealed endocytosis as a principal mechanism of NPs uptake. In vitro rat microsomal metabolism studies demonstrated noticeable advantage of LPV NPs by affording metabolic protection to LPV. These studies indicate usefulness of PCL NPs in enhancing oral bioavailability and improving pharmacokinetic profile of LPV.


Subject(s)
Caproates/administration & dosage , Drug Delivery Systems/methods , Drug Design , Lactones/administration & dosage , Lopinavir/administration & dosage , Nanoparticles/administration & dosage , Polymers/administration & dosage , Administration, Oral , Animals , Caproates/chemistry , Caproates/metabolism , Drug Evaluation, Preclinical/methods , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Lactones/chemistry , Lactones/metabolism , Lopinavir/chemistry , Lopinavir/metabolism , Male , Nanoparticles/chemistry , Nanoparticles/metabolism , Organ Culture Techniques , Polymers/chemistry , Polymers/metabolism , Rats , Rats, Wistar
19.
J Mol Graph Model ; 47: 18-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24291501

ABSTRACT

Crystal structure of multidrug-resistant (MDR) clinical isolate 769, human immunodeficiency virus type-1 (HIV-1) protease in complex with lopinavir (LPV) (PDB ID: 1RV7) showed altered binding orientation of LPV in the expanded active site cavity, causing loss of contacts and decrease in potency. In the current study, with a goal to restore the lost contacts, three libraries of LPV analogs containing extended P1 and/or P1' phenyl groups were designed and docked into the expanded active site cavity of the MDR769 HIV-1 protease. The compounds were then ranked based on three criteria: binding affinity, overall binding profile and predicted pharmacological properties. Among the twelve proposed extensions in different combinations, compound 14 (consists of para-fluoro phenyl group as both P1 and P1' moieties) was identified as a lead with improved binding profile, binding affinity against the MDR protease and favorable predicted pharmacological properties comparable to those of LPV. The binding affinity of 14 against wild type (NL4-3) HIV-1 protease was comparable to that of LPV and was better than LPV against an ensemble of MDR HIV-1 protease variants. Thus, 14 shows enhanced binding affinity by restoring lost contacts in the expanded active site cavity of MDR769 HIV-1 protease variants suggesting that it may have higher potency compared to that of LPV and hence should be further synthesized and evaluated against NL4-3 as well as MDR variants of HIV-1.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , Lopinavir/chemistry , Drug Resistance, Viral , HIV Protease/metabolism , HIV Protease Inhibitors/metabolism , Humans , Hydrogen Bonding , Lopinavir/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship
20.
J Antimicrob Chemother ; 69(4): 995-1004, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24275118

ABSTRACT

OBJECTIVES: The potential cardiovascular (CV) toxicity associated with combined antiretroviral therapy (cART) has been attributed mainly to the nucleoside reverse transcriptase inhibitors abacavir and didanosine. However, the other two components of cART--non-nucleoside reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs)--may also be implicated, either directly or by influencing the action of the other drugs. This study evaluates the acute direct effects of the NNRTIs efavirenz and nevirapine and one of the most widely employed PIs, lopinavir, on leucocyte-endothelium interactions, a hallmark of CV disease. METHODS: Drugs were analysed in vitro in human cells (interactions of peripheral blood polymorphonuclear or mononuclear cells with human umbilical vein endothelial cells) using a flow chamber system, and in vivo in rat mesenteric vessels by means of intravital microscopy. The expression of adhesion molecules in leucocytes and endothelial cells was studied by flow cytometry, and the role of these molecules in white cell recruitment was evaluated by pre-treating human cells or rats with blocking antibodies. RESULTS: Efavirenz and nevirapine, but not lopinavir, increased the rolling flux and adhesion of leucocytes in vitro and in vivo while inducing emigration in rat venules. Efavirenz, but not nevirapine, augmented the levels of CD11b, CD11c and CD18 in neutrophils and monocytes. The actions of efavirenz, but not of nevirapine, were reversed by antibodies against Mac-1 (CD11b/CD18), gp150,95 (CD11c/CD18) or ICAM-1 (CD54). CONCLUSIONS: NNRTIs, but not PIs, interfere with leucocyte-endothelial interactions. However, differences between efavirenz and nevirapine suggest a specific CV profile for each compound.


Subject(s)
Anti-HIV Agents/metabolism , Benzoxazines/metabolism , Cell Adhesion , Endothelium/physiology , Integrin alphaXbeta2/metabolism , Leukocytes/physiology , Macrophage-1 Antigen/metabolism , Alkynes , Animals , Cells, Cultured , Cyclopropanes , Endothelium/drug effects , Flow Cytometry , Gene Expression Profiling , Humans , Leukocytes/drug effects , Lopinavir/metabolism , Male , Nevirapine/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...