Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36674655

RESUMEN

Mycobacterium tuberculosis is able to establish a chronic colonization of lung macrophages in a controlled replication manner, giving rise to a so-called latent infection. Conversely, when intracellular bacteria undergo actively uncontrolled replication rates, they provide the switch for the active infection called tuberculosis to occur. Our group found that the pathogen is able to manipulate the activity of endolysosomal enzymes, cathepsins, directly at the level of gene expression or indirectly by regulating their natural inhibitors, cystatins. To provide evidence for the crucial role of cathepsin manipulation for the success of tuberculosis bacilli in their intracellular survival, we used liposomal delivery of saquinavir. This protease inhibitor was previously found to be able to increase cathepsin proteolytic activity, overcoming the pathogen induced blockade. In this study, we demonstrate that incorporation in liposomes was able to increase the efficiency of saquinavir internalization in macrophages, reducing cytotoxicity at higher concentrations. Consequently, our results show a significant impact on the intracellular killing not only to reference and clinical strains susceptible to current antibiotic therapy but also to multidrug- and extensively drug-resistant (XDR) Mtb strains. Altogether, this indicates the manipulation of cathepsins as a fine-tuning strategy used by the pathogen to survive and replicate in host cells.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Mycobacterium tuberculosis/metabolismo , Catepsinas/metabolismo , Saquinavir/farmacología , Saquinavir/metabolismo , Liposomas/metabolismo , Macrófagos/metabolismo , Tuberculosis/microbiología , Interacciones Huésped-Patógeno/fisiología
2.
Int J Mol Sci ; 23(20)2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36293006

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) has continued to be a global concern. With the new HIV incidence, the emergence of multi-drug resistance and the untoward side effects of currently used anti-HIV drugs, there is an urgent need to discover more efficient anti-HIV drugs. Modern computational tools have played vital roles in facilitating the drug discovery process. This research focuses on a pharmacophore-based similarity search to screen 111,566,735 unique compounds in the PubChem database to discover novel HIV-1 protease inhibitors (PIs). We used an in silico approach involving a 3D-similarity search, physicochemical and ADMET evaluations, HIV protease-inhibitor prediction (IC50/percent inhibition), rigid receptor-molecular docking studies, binding free energy calculations and molecular dynamics (MD) simulations. The 10 FDA-approved HIV PIs (saquinavir, lopinavir, ritonavir, amprenavir, fosamprenavir, atazanavir, nelfinavir, darunavir, tipranavir and indinavir) were used as reference. The in silico analysis revealed that fourteen out of the twenty-eight selected optimized hit molecules were within the acceptable range of all the parameters investigated. The hit molecules demonstrated significant binding affinity to the HIV protease (PR) when compared to the reference drugs. The important amino acid residues involved in hydrogen bonding and п-п stacked interactions include ASP25, GLY27, ASP29, ASP30 and ILE50. These interactions help to stabilize the optimized hit molecules in the active binding site of the HIV-1 PR (PDB ID: 2Q5K). HPS/002 and HPS/004 have been found to be most promising in terms of IC50/percent inhibition (90.15%) of HIV-1 PR, in addition to their drug metabolism and safety profile. These hit candidates should be investigated further as possible HIV-1 PIs with improved efficacy and low toxicity through in vitro experiments and clinical trial investigations.


Asunto(s)
Fármacos Anti-VIH , Inhibidores de la Proteasa del VIH , VIH-1 , Humanos , Inhibidores de la Proteasa del VIH/química , Proteasa del VIH/química , Darunavir/farmacología , Indinavir/química , Indinavir/metabolismo , Indinavir/farmacología , Nelfinavir/química , Nelfinavir/metabolismo , Nelfinavir/farmacología , Ritonavir/química , Saquinavir/metabolismo , Saquinavir/farmacología , Lopinavir/farmacología , Sulfato de Atazanavir/farmacología , Simulación del Acoplamiento Molecular , Fármacos Anti-VIH/farmacología , Aminoácidos/farmacología
3.
Plast Reconstr Surg ; 150(6): 1264e-1274e, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36112847

RESUMEN

BACKGROUND: Using immunomodulatory methods to address the challenging issue of craniofacial bone repair may be a potentially effective approach. The protease inhibitor saquinavir has been shown to inhibit the inflammatory response by targeting the toll-like receptor 4/myeloid differentiation primary response complex. Independently, inhibition of toll-like receptor 4 or myeloid differentiation primary response led to enhanced skull bone repair. Therefore, the authors aimed to investigate the effects of saquinavir on skull bone healing. METHODS: The effects of saquinavir on skull bone healing were assessed by means of gene expression, histology, immunohistochemistry, and tomography in a mouse calvarial defect model. Subsequently, the role of saquinavir in cell viability, migration, and osteogenic and osteoclastogenic differentiation was also evaluated in vitro. RESULTS: One-week saquinavir administration improved skull bone healing based on micro-computed tomographic and histomorphometric analyses. Compared to the vehicle control, 1-week saquinavir treatment (1) enhanced osteoclast infiltration (tartrate-resistant acid phosphatase staining) at day 7, but not at days 14 and 28; (2) induced more CD206 + M2 macrophage infiltration, but not F4/80 + M0 macrophages at days 7, 14, and 28; and (3) elevated osteoclastogenic gene RANKL (quantitative polymerase chain reaction) expression and other osteogenic and cytokine expression. Furthermore, in vitro data showed that saquinavir administration did not influence MC3T3-E1 cell migration or mineralization, whereas higher concentrations of saquinavir inhibited cell viability. Saquinavir treatment also enhanced the osteoclastic differentiation of bone marrow-derived precursors, and partially reversed high-mobility group box 1-driven osteoclastogenesis inhibition and elevated proinflammatory cytokine expression. CONCLUSION: The improved skull bone repair following short-term saquinavir treatment may involve enhanced osteoclastogenesis and modulated inflammatory response following skull injury. CLINICAL RELEVANCE STATEMENT: The authors' work demonstrates improved skull bone healing by short-term application of saquinavir, a drug traditionally used in the treatment of acquired immunodeficiency syndrome. As such, saquinavir may be repurposed for skeletal repair.


Asunto(s)
Inhibidores de la Proteasa del VIH , Saquinavir , Ratones , Animales , Saquinavir/farmacología , Saquinavir/metabolismo , Saquinavir/uso terapéutico , Inhibidores de la Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Receptor Toll-Like 4/fisiología , Osteogénesis , Cráneo/lesiones
4.
Viruses ; 14(4)2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35458427

RESUMEN

Drug-resistance-associated mutation in essential proteins of the viral life cycle is a major concern in anti-retroviral therapy. M46I, a non-active site mutation in HIV-1 protease has been clinically associated with saquinavir resistance in HIV patients. A 100 ns molecular dynamics (MD) simulation and MM-PBSA calculations were performed to study the molecular mechanism of M46I-mutation-based saquinavir resistance. In order to acquire deeper insight into the drug-resistance mechanism, the flap curling, closed/semi-open/open conformations, and active site compactness were studied. The M46I mutation significantly affects the energetics and conformational stability of HIV-1 protease in terms of RMSD, RMSF, Rg, SASA, and hydrogen formation potential. This mutation significantly decreased van der Waals interaction and binding free energy (∆G) in the M46I-saquinavir complex and induced inward flap curling and a wider opening of the flaps for most of the MD simulation period. The predominant open conformation was reduced, but inward flap curling/active site compactness was increased in the presence of saquinavir in M46I HIV-1 protease. In conclusion, the M46I mutation induced structural dynamics changes that weaken the protease grip on saquinavir without distorting the active site of the protein. The produced information may be utilized for the discovery of inhibitor(s) against drug-resistant HIV-1 protease.


Asunto(s)
Infecciones por VIH , Inhibidores de la Proteasa del VIH , VIH-1 , Sitios de Unión , Dominio Catalítico , Farmacorresistencia Viral/genética , Infecciones por VIH/tratamiento farmacológico , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/genética , VIH-1/metabolismo , Humanos , Simulación de Dinámica Molecular , Mutación , Saquinavir/química , Saquinavir/metabolismo , Saquinavir/farmacología
5.
J Biomol Struct Dyn ; 40(14): 6439-6449, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-33663345

RESUMEN

Developing novel drug molecules against HIV is a scientific quest necessitated by development of drug resistance against used drugs. We report comparative results of molecular dynamics simulation studies on 11 structural analogues of Saquinavir (SQV) - against HIV-protease that were earlier examined for pharmacodynamic and pharmacokinetic properties. We reported analogues S1, S5 and S8 to qualify the ADMET criterion and may be considered as potential lead molecules. In this study the designed molecules were successively docked with native HIV-protease at AutoDock. Docking scores established relative goodness of the 11 analogues against the benchmark for Saquinavir. The docked complexes were subjected to molecular dynamics simulation studies using GROMACS 4.6.2. Four parameters viz. H-bonding, RMSD, Binding energy and Protein-Ligand Distance were used for comparative analyses of the analogues relative to Saquinavir. The comparison and analysis of the results are indicative that analogues S8, S9 and S1 are promising candidates among all the analogues studied. From our earlier work and present study it is evident that among the three S8 and S1 qualify the ADMET criterion and between S1 and S8, the analogue S8 shows more target efficacy and specificity over S1 and have better molecular dynamics simulation results. Thus, of the 11 de novo Saquinavir analogues, the S8 appears to be the most promising candidate as lead molecule for HIV-protease inhibitor and is best suited for testing under biological system. Further validation of the proposed lead molecules through wet lab studies involving antiviral assays however is required.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Infecciones por VIH , Inhibidores de la Proteasa del VIH , Infecciones por VIH/tratamiento farmacológico , Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/química , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Saquinavir/química , Saquinavir/metabolismo , Saquinavir/farmacología
6.
Sci Rep ; 10(1): 16986, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046764

RESUMEN

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.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Cisteína Endopeptidasas/metabolismo , Reposicionamiento de Medicamentos/métodos , Inhibidores de la Proteasa del VIH/farmacología , Neumonía Viral/tratamiento farmacológico , Proteínas no Estructurales Virales/metabolismo , Betacoronavirus/metabolismo , Sitios de Unión/efectos de los fármacos , Fenómenos Biofísicos , COVID-19 , Dominio Catalítico/efectos de los fármacos , Biología Computacional , Proteasas 3C de Coronavirus , Darunavir/metabolismo , Darunavir/farmacología , Inhibidores de la Proteasa del VIH/metabolismo , Humanos , Indinavir/metabolismo , Indinavir/farmacología , Lopinavir/metabolismo , Lopinavir/farmacología , Simulación de Dinámica Molecular , Nelfinavir/metabolismo , Nelfinavir/farmacología , Pandemias , Ritonavir/metabolismo , Ritonavir/farmacología , SARS-CoV-2 , Saquinavir/metabolismo , Saquinavir/farmacología
7.
Int J Nanomedicine ; 15: 5113-5129, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32764940

RESUMEN

BACKGROUND: Low bioavailability and poor permeability of the blood-brain barrier are problematic when delivering therapeutic agents and particularly anti-human immunodeficiency virus therapy to the central nervous system. The intranasal route offers an alternative for central nervous system delivery. Cubosomes have been reported as helpful vehicles for intranasal delivery of therapeutics to enable brain targeting. OBJECTIVE: In this study, we aimed to develop the intranasal cubosomal thermogelling dispersion of saquinavir mesylate for central nervous system delivery. METHODS: The Box-Behnken design was applied to study the effect of monoolein, Poloxamer 407, and polyvinyl alcohol as independent factors and the particle size, entrapment efficiency, gelation temperature, and stability index as responses. The optimized cubosomes were evaluated using transmission electron microscopy, ex vivo permeation, and in vivo pharmacokinetics. RESULTS: The optimized formula consisting of monoolein (8.96%), Poloxamer 407 (17.45%), and polyvinyl alcohol (7.5%) was prepared and evaluated. Higher values for the steady-state flux, permeability coefficient, and enhancement factor were observed for the cubosomal thermogelling dispersion of saquinavir during ex vivo permeation in comparison with an aqueous suspension of saquinavir. From the pharmacokinetic profile, the relative bioavailability for the intranasal optimized formula was approximately 12-fold higher when compared with oral aqueous suspension and 2.5-fold greater when compared to the intranasal aqueous suspension of saquinavir. CONCLUSION: Overall, the saquinavir-loaded cubosomal thermogelling formulation is promising for central nervous system delivery by intranasal administration.


Asunto(s)
Portadores de Fármacos/química , Cristales Líquidos/química , Nanoestructuras/química , Saquinavir/farmacología , Temperatura , Administración Intranasal , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Geles , Glicéridos/química , Masculino , Tamaño de la Partícula , Permeabilidad , Poloxámero/química , Alcohol Polivinílico/química , Saquinavir/administración & dosificación , Saquinavir/metabolismo
8.
Drug Metab Dispos ; 46(3): 268-278, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29233818

RESUMEN

Prediction of clearance-a vital component of drug discovery-remains in need of improvement and, in particular, requires more incisive assessment of mechanistic methodology in vitro, according to a number of recent reports. Although isolated hepatocytes have become an irreplaceable standard system for the measurement of intrinsic hepatic clearance mediated by active uptake transport and metabolism, the lack of prediction reliability appears to reflect a lack of methodological validation, especially for highly cleared drugs, as we have previously shown. Here, novel approaches were employed to explore fundamental experimental processes and associated potential limitations of in vitro predictions of clearance. Rat hepatocytes deemed nonviable by trypan blue staining showed undiminished metabolic activity for probe cytochrome P450 (P450) substrates midazolam and propranolol; supplementation with NADPH enhanced these activities. Extensive permeabilization of the plasma membrane using saponin showed either full or minimal P450 activity, depending on the presence or absence of 1 mM NADPH, respectively. The shaking of incubations facilitated P450 metabolic rates up to 5-fold greater than static incubation, depending on intrinsic clearance, indicating the critical influence of the unstirred water layer (UWL). Permeabilization allowed static incubation metabolic rates to approach those of shaking for intact cells, indicating an artificially induced breakdown of the UWL. Permeabilization combined with shaking allowed an increased metabolic rate for saquinavir, resolving the membrane permeability limitation for this drug. These findings advance the interpretation of the rate-limiting processes involved in intrinsic clearance measurements and could be critical for successful in vitro prediction.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Hepatocitos/metabolismo , Hígado/metabolismo , Tasa de Depuración Metabólica/fisiología , Agua/metabolismo , Animales , Transporte Biológico/fisiología , Sistema Enzimático del Citocromo P-450/metabolismo , Cinética , Masculino , Midazolam/metabolismo , NADP/metabolismo , Propranolol/metabolismo , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Saquinavir/metabolismo
9.
Drug Des Devel Ther ; 10: 3699-3706, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27895462

RESUMEN

BACKGROUND: The intestinal cytochrome P450 3A (CYP 3A) and P-glycoprotein (P-gp) present a barrier to the oral absorption of saquinavir (SQV). Resveratrol (RESV) has been indicated to have modulatory effects on P-gp and CYP 3A. Therefore, this study was to investigate the effects of RESV on P-gp and CYP 3A activities in vitro and in vivo on oral SQV pharmacokinetics in rats. METHODS: In vitro, intestinal microsomes were used to evaluate RESV effect on CYP 3A-mediated metabolism of SQV; MDR1-expressing Madin-Darby canine kidney (MDCKII-MDR1) cells were employed to assess the impact of RESV on P-gp-mediated efflux of SQV. In vivo effects were studied using 10 rats randomly assigned to receive oral SQV (30 mg/kg) with or without RESV (20 mg/kg). Serial blood samples were obtained over the following 24 h. Concentrations of SQV in samples were ascertained using high-performance liquid chromatography-tandem mass spectrometry analysis. RESULTS: RESV (1-100 µM) enhanced residual SQV (% of control) in a dose-dependent manner after incubation with intestinal microsomes. RESV (1-100 µM) reduced the accumulation of SQV in MDCKII-MDR1 cells in a concentration-dependent manner. A double peaking phenomenon was observed in the plasma SQV profiles in rats. The first peak of plasma SQV concentration was increased, but the second peak was reduced by coadministration with RESV. The mean AUC0-∞ of SQV was slightly decreased, with no statistical significance probably due to the high individual variation. CONCLUSION: RESV can alter the plasma SQV concentration profiles, shorten the Tmax of SQV. RESV might also cause a slight decrease tendency in the SQV bioavailability in rats.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inhibidores Enzimáticos/farmacología , Saquinavir/metabolismo , Saquinavir/farmacocinética , Estilbenos/farmacología , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Línea Celular , Cromatografía Líquida de Alta Presión , Perros , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacocinética , Absorción Intestinal , Masculino , Microsomas , Ratas , Ratas Sprague-Dawley , Resveratrol , Saquinavir/administración & dosificación , Estilbenos/farmacocinética , Espectrometría de Masas en Tándem
10.
J Lipid Res ; 56(8): 1543-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26026069

RESUMEN

α-Tocopherol (vitamin E) has attracted considerable attention as a potential protective or palliative agent. In vitro, its free radical-scavenging antioxidant action has been widely demonstrated. In vivo, however, vitamin E treatment exhibits negligible benefits against oxidative stress. α-Tocopherol influences lipid ordering within biological membranes and its derivatives have been suggested to inhibit the multi-drug efflux pump, P-glycoprotein (P-gp). This study employs the fluorescent membrane probe, 1-(3-sulfonatopropyl)-4-[ß[2-(di-n-octylamino)-6-naphthyl]vinyl] pyridinium betaine, to investigate whether these effects are connected via influences on the membrane dipole potential (MDP), an intrinsic property of biological membranes previously demonstrated to modulate P-gp activity. α-Tocopherol and its non-free radical-scavenging succinate analog induced similar decreases in the MDP of phosphatidylcholine vesicles. α-Tocopherol succinate also reduced the MDP of T-lymphocytes, subsequently decreasing the binding affinity of saquinavir for P-gp. Additionally, α-tocopherol succinate demonstrated a preference for cholesterol-treated (membrane microdomain enriched) cells over membrane cholesterol-depleted cells. Microdomain disruption via cholesterol depletion decreased saquinavir's affinity for P-gp, potentially implicating these structures in the influence of α-tocopherol succinate on P-gp. This study provides evidence of a microdomain dipole potential-dependent mechanism by which α-tocopherol analogs influence P-gp activity. These findings have implications for the use of α-tocopherol derivatives for drug delivery across biological barriers.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Saquinavir/metabolismo , alfa-Tocoferol/farmacología , Membrana Celular/química , Fluoresceínas/química , Colorantes Fluorescentes/química , Humanos , Células Jurkat , Cetocolesteroles/farmacología , Ligandos , Modelos Moleculares , Conformación Molecular , Fosfatidiletanolaminas/química , Unión Proteica/efectos de los fármacos , Compuestos de Piridinio/química , Liposomas Unilamelares/química , Liposomas Unilamelares/metabolismo
11.
Eur J Med Chem ; 93: 338-48, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25707014

RESUMEN

In spite of remarkable advances in the knowledge on Trypanosoma cruzi biology, no medications to treat Chagas disease have been approved in the last 40 years and almost 8 million people remain infected. Since the public sector and non-profit organizations play a significant role in the research efforts on Chagas disease, it is important to implement research strategies that promote translation of basic research into the clinical practice. Recent international public-private initiatives address the potential of drug repositioning (i.e. finding second or further medical uses for known-medications) which can substantially improve the success at clinical trials and the innovation in the pharmaceutical field. In this work, we present the computer-aided identification of approved drugs clofazimine, benidipine and saquinavir as potential trypanocidal compounds and test their effects at biochemical as much as cellular level on different parasite stages. According to the obtained results, we discuss biopharmaceutical, toxicological and physiopathological criteria applied to decide to move clofazimine and benidipine into preclinical phase, in an acute model of infection. The article illustrates the potential of computer-guided drug repositioning to integrate and optimize drug discovery and preclinical development; it also proposes rational rules to select which among repositioned candidates should advance to investigational drug status and offers a new insight on clofazimine and benidipine as candidate treatments for Chagas disease. One Sentence Summary: We present the computer-guided drug repositioning of three approved drugs as potential new treatments for Chagas disease, integrating computer-aided drug screening and biochemical, cellular and preclinical tests.


Asunto(s)
Reposicionamiento de Medicamentos/métodos , Tripanocidas/farmacología , Animales , Clofazimina/metabolismo , Clofazimina/farmacología , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Dihidropiridinas/metabolismo , Dihidropiridinas/farmacología , Femenino , Masculino , Ratones , Simulación del Acoplamiento Molecular , Conformación Proteica , Proteínas Protozoarias , Saquinavir/metabolismo , Saquinavir/farmacología , Tripanocidas/metabolismo , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/enzimología
12.
Int J Pharm ; 475(1-2): 191-7, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25178831

RESUMEN

Saquinavir mesylate (SM) is a protease inhibitor with activity against human immunodeficiency virus type 1 (HIV-1) and is available in tablet form, which has three major problems. First, the drug undergoes extensive first pass metabolism. Second, the drug has a poor aqueous solubility. And third, it has low GIT permeability and absorption. These constrains lead to decrease oral bioavailability (4% only) and administration of large doses which increase the incidence of occurrence of the side effects. The aim of this research was to utilize nanotechnology to formulate (SM) into a nasal in situ nanosized microemulsion gel (NEG) to provide a solution for the previously mentioned problems. The solubility of (SM) in various oils, surfactants, and cosurfactants was estimated. Pseudo-ternary phase diagrams were developed and various nanosized microemulsion (NE) were prepared, and subjected to characterization, stability study, and droplet size measurements. Gellan gum was used as an in situ gelling agent. The gel strength, critical ionic concentration, gelation characteristics, in vitro release, and ex vivo nasal permeation were determined. The pharmacokinetic study was carried out in rabbits. Stable NEs were successfully developed with a droplet size range of 25-61 nm. A NEG composed of 17.5% Labrafac PG, 33% Labrasol, and 11% Transcutol HP successfully provided the maximum in vitro and ex vivo permeation, and enhanced the bioavailability in the rabbits by 12-fold when compared with the marketed tablets. It can be concluded that the nasal NEG is a promising novel formula for (SM) that has higher nasal tissue permeability and enhanced systemic bioavailability.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Excipientes/química , Inhibidores de la Proteasa del VIH/administración & dosificación , Nanoestructuras/química , Absorción Nasal , Saquinavir/administración & dosificación , Administración Intranasal , Animales , Disponibilidad Biológica , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Composición de Medicamentos , Estabilidad de Medicamentos , Emulsiones , Glicoles de Etileno/química , Geles , Glicéridos/química , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacocinética , Masculino , Polietilenglicoles/química , Polisacáridos Bacterianos/química , Conejos , Saquinavir/química , Saquinavir/metabolismo , Saquinavir/farmacocinética , Solubilidad , Tensoactivos/química
13.
Biomed Res Int ; 2014: 358425, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24982867

RESUMEN

P-glycoprotein (P-gp) is involved in the transport of xenobiotic compounds and responsible for the decrease of the drug accumulation in multi-drug-resistant cells. In this investigation we compare several docking algorithms in order to find the conditions that are able to discriminate between P-gp binders and nonbinders. We built a comprehensive dataset of binders and nonbinders based on a careful analysis of the experimental data available in the literature, trying to overcome the discrepancy noticeable in the experimental results. We found that Autodock Vina flexible docking is the best choice for the tested options. The results will be useful to filter virtual screening results in the rational design of new drugs that are not expected to be expelled by P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Simulación del Acoplamiento Molecular , Algoritmos , Animales , Cristalografía por Rayos X , Humanos , Ratones , Curva ROC , Saquinavir/química , Saquinavir/metabolismo , Homología Estructural de Proteína
14.
Int J Pharm ; 468(1-2): 105-11, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24746410

RESUMEN

The main objectives of the present study were (i) to evaluate the effect of the mucus layer on saquinavir-loaded nanostructured lipid carriers (SQV-NLCs) uptake and (ii) to evaluate the mucopenetrating properties of dextran-protamine (Dex-Prot) coating on NLCs as per SQV permeability enhancement. Three different NLC formulations differing on particle size and surfactant content were obtained and coated with Dex-Prot complexes. SQV permeability was then evaluated across Caco-2 cell monolayers (enterocyte-like model) and Caco-2/HT29-MTX cell monolayers (mucus model). In the Caco-2 monolayers, Dex-Prot-NLCs increased up to 9-fold SQV permeability in comparison to uncoated nanoparticles. In the Caco-2/HT29-MTX monolayers, Dex-Prot-NLCs presenting a surface charge close to neutrality significantly increased SQV permeability. Hence, Dex-Prot complex coating is a promising strategy to ensure successful nanoparticle mucus-penetration, and thus, an efficient nanoparticle oral delivery. To our knowledge, this is the first time that Dex-Prot coating has been described as a nanoparticle muco-penetration enhancer across the intestinal mucus barrier.


Asunto(s)
Dextranos/química , Portadores de Fármacos , Enterocitos/metabolismo , Moco/metabolismo , Protaminas/química , Saquinavir/metabolismo , Células CACO-2 , Química Farmacéutica , Células HT29 , Humanos , Absorción Intestinal , Nanoestructuras , Nanotecnología , Tamaño de la Partícula , Permeabilidad , Saquinavir/química , Propiedades de Superficie , Tensoactivos/química , Tecnología Farmacéutica/métodos
15.
Drug Deliv ; 21(2): 148-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24128122

RESUMEN

The central nervous system (CNS) is an immunological privileged sanctuary site-providing reservoir for HIV-1 virus. Current anti-HIV drugs, although effective in reducing plasma viral levels, cannot eradicate the virus completely from the body. The low permeability of anti-HIV drugs across the blood-brain barrier (BBB) leads to insufficient delivery. Therefore, developing a novel approaches enhancing the CNS delivery of anti-HIV drugs are required for the treatment of neuro-AIDS. The aim of this study was to develop intranasal nanoemulsion (NE) for enhanced bioavailability and CNS targeting of saquinavir mesylate (SQVM). SQVM is a protease inhibitor which is a poorly soluble drug widely used as antiretroviral drug, with oral bioavailability is about 4%. The spontaneous emulsification method was used to prepare drug-loaded o/w nanoemulsion, which was characterized by droplet size, zeta potential, pH, drug content. Moreover, ex-vivo permeation studies were performed using sheep nasal mucosa. The optimized NE showed a significant increase in drug permeation rate compared to the plain drug suspension (PDS). Cilia toxicity study on sheep nasal mucosa showed no significant adverse effect of SQVM-loaded NE. Results of in vivo biodistribution studies show higher drug concentration in brain after intranasal administration of NE than intravenous delivered PDS. The higher percentage of drug targeting efficiency (% DTE) and nose-to-brain drug direct transport percentage (% DTP) for optimized NE indicated effective CNS targeting of SQVM via intranasal route. Gamma scintigraphy imaging of the rat brain conclusively demonstrated transport of drug in the CNS at larger extent after intranasal administration as NE.


Asunto(s)
Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/metabolismo , Mucosa Nasal/metabolismo , Saquinavir/metabolismo , Administración Intranasal , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Emulsiones , Masculino , Nanopartículas/administración & dosificación , Mucosa Nasal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Saquinavir/administración & dosificación , Ovinos
16.
Drug Metab Dispos ; 42(2): 213-20, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24212380

RESUMEN

Saquinavir (SQV) is a protease inhibitor widely used for the treatment of human immunodeficiency virus (HIV) infection. We profiled SQV metabolism in mice using a metabolomic approach. Thirty SQV metabolites were identified in mouse feces and urine, of which 20 are novel. Most metabolites observed in mice were recapitulated in human liver microsomes. Among these novel metabolites, one α-hydroxyaldehyde produced from SQV N-dealkylation was noted and verified for the first time. Meanwhile, the corresponding product (3S)-N-tert-butyldecahydro-isoquinoline-3-carboxamide and its further metabolites were identified in mouse urine. The α-hydroxyaldehyde pathway was confirmed by using semicarbazide as a trapping reagent as well. Using recombinant cytochrome P450 (CYP450) isoenzymes and Cyp3a-null mice, CYP3A was identified as the dominant enzyme contributing to the formation of α-hydroxyaldehyde. This study enhances our knowledge of SQV metabolism, which can be used for predicting drug-drug interactions and further understanding the mechanism of adverse effects associated with SQV.


Asunto(s)
Aldehídos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , Hígado/enzimología , Saquinavir/metabolismo , Aldehídos/orina , Animales , Biotransformación , Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/genética , Remoción de Radical Alquila , Heces/química , Inhibidores de la Proteasa del VIH/orina , Humanos , Masculino , Metabolómica , Ratones , Ratones Noqueados , Proteínas Recombinantes/metabolismo , Saquinavir/orina
17.
PLoS One ; 8(10): e75374, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098380

RESUMEN

Persistence of HIV-1 reservoirs within the Central Nervous System (CNS) remains a significant challenge to the efficacy of potent anti-HIV-1 drugs. The primary human Brain Microvascular Endothelial Cells (HBMVEC) constitutes the Blood Brain Barrier (BBB) which interferes with anti-HIV drug delivery into the CNS. The ATP binding cassette (ABC) transporters expressed on HBMVEC can efflux HIV-1 protease inhibitors (HPI), enabling the persistence of HIV-1 in CNS. Constitutive low level expression of several ABC-transporters, such as MDR1 (a.k.a. P-gp) and MRPs are documented in HBMVEC. Although it is recognized that inflammatory cytokines and exposure to xenobiotic drug substrates (e.g HPI) can augment the expression of these transporters, it is not known whether concomitant exposure to virus and anti-retroviral drugs can increase drug-efflux functions in HBMVEC. Our in vitro studies showed that exposure of HBMVEC to HIV-1 significantly up-regulates both MDR1 gene expression and protein levels; however, no significant increases in either MRP-1 or MRP-2 were observed. Furthermore, calcein-AM dye-efflux assays using HBMVEC showed that, compared to virus exposure alone, the MDR1 mediated drug-efflux function was significantly induced following concomitant exposure to both HIV-1 and saquinavir (SQV). This increase in MDR1 mediated drug-efflux was further substantiated via increased intracellular retention of radiolabeled [(3)H-] SQV. The crucial role of MDR1 in (3)H-SQV efflux from HBMVEC was further confirmed by using both a MDR1 specific blocker (PSC-833) and MDR1 specific siRNAs. Therefore, MDR1 specific drug-efflux function increases in HBMVEC following co-exposure to HIV-1 and SQV which can reduce the penetration of HPIs into the infected brain reservoirs of HIV-1. A targeted suppression of MDR1 in the BBB may thus provide a novel strategy to suppress residual viral replication in the CNS, by augmenting the therapeutic efficacy of HAART drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/virología , VIH-1/fisiología , Saquinavir/metabolismo , Saquinavir/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Transporte Biológico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/virología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , VIH-1/efectos de los fármacos , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , ARN Interferente Pequeño/genética
18.
J Chem Inf Model ; 53(8): 2141-53, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23834142

RESUMEN

The emergence of HIV-1 drug-resistant mutations is the major problem against AIDS treatment. We employed molecular dynamics (MD) calculations and free energy (MM-PBSA and thermodynamic integration) analyses on wild-type (WT) and mutated HIV-1 protease (HIV-1 PR) complexes with darunavir, amprenavir, indinavir, and saquinavir to clarify the mechanism of resistance due to the I50V flap mutation. Conformational analysis showed that the protease flaps are increasingly flexible in the I50V complexes. In the WT, stabilization of the HIV-1 PR structure is achieved via interflap and water-mediated hydrogen-bonding interactions between the flaps. Furthermore, hydrogen bonds between drugs and binding cavity residues (Asp29/29'/30/30') are crucial for effective inhibition. All these interactions were significantly diminished (or absent) in the mutated forms, thus denoting their importance toward binding. Thermodynamic integration calculations reproduced the experimental data to within ≈1 kcal mol⁻¹ and showed that the I50V mutation results in weaker binding free energies for all analyzed complexes with respect to the WT. It was observed that the loss in binding energy upon mutation was mostly enthalpically driven in all complexes, with the greatest effect coming from the reduction of van der Waals interactions. Our results motivated us to test two novel compounds that have been synthesized to maximize interactions with HIV-1 PR. MM-PBSA and TI calculations showed that compound 3c (Ghosh et al. Bioorg. Med. Chem. Lett. 2012, 22, 2308) is a promising protease inhibitor, which presents very effective binding to the WT PR (ΔG(MM-PBSA) = -17.2 kcal mol⁻¹, ΔG(exp) = -16.1 kcal mol⁻¹). Upon I50V mutation, the complex binding free energy was weakened by a ΔΔG(TI) of 1.8 kcal mol⁻¹, comparable to the marketed inhibitors. This predicts that I50V may confer low resistance to 3c. This computational comparative study contributes toward elucidation of the I50V drug-resistance mechanism in HIV-1 PR.


Asunto(s)
Fármacos Anti-VIH/metabolismo , Farmacorresistencia Viral/genética , Inhibidores de la Proteasa del VIH/metabolismo , Proteasa del VIH/metabolismo , VIH-1/efectos de los fármacos , Simulación de Dinámica Molecular , Mutación , Fármacos Anti-VIH/farmacología , Carbamatos/metabolismo , Carbamatos/farmacología , Análisis por Conglomerados , Darunavir , Furanos , Proteasa del VIH/química , Proteasa del VIH/genética , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/enzimología , VIH-1/genética , Enlace de Hidrógeno , Indinavir/metabolismo , Indinavir/farmacología , Conformación Proteica , Saquinavir/metabolismo , Saquinavir/farmacología , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Termodinámica
19.
J Neuroinflammation ; 10: 58, 2013 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-23642074

RESUMEN

BACKGROUND: Active HIV infection within the central nervous system (CNS) is confined primarily to microglia. The glial cell compartment acts as a viral reservoir behind the blood-brain barrier. It provides an additional roadblock to effective pharmacological treatment via expression of multiple drug efflux transporters, including P-glycoprotein. HIV/AIDS patients frequently suffer bacterial and viral co-infections, leading to deregulation of glial cell function and release of pro-inflammatory mediators including cytokines, chemokines, and nitric oxide. METHODS: To better define the role of inflammation in decreased HIV drug accumulation into CNS targets, accumulation of the antiretroviral saquinavir was examined in purified cultures of rodent microglia exposed to the prototypical inflammatory mediator lipopolysaccharide (LPS). RESULTS: [(3)H]-Saquinavir accumulation by microglia was rapid, and was increased up to two-fold in the presence of the specific P-glycoprotein inhibitor, PSC833. After six or 24 hours of exposure to 10 ng/ml LPS, saquinavir accumulation was decreased by up to 45%. LPS did not directly inhibit saquinavir transport, and did not affect P-glycoprotein protein expression. LPS exposure did not alter RNA and/or protein expression of other transporters including multidrug resistance-associated protein 1 and several solute carrier uptake transporters. CONCLUSIONS: The decrease in saquinavir accumulation in microglia following treatment with LPS is likely multi-factorial, since drug accumulation was attenuated by inhibitors of NF-κß and the MEK1/2 pathway in the microglia cell line HAPI, and in primary microglia cultures from toll-like receptor 4 deficient mice. These data provide new pharmacological insights into why microglia act as a difficult-to-treat viral sanctuary site.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacología , Activación de Macrófagos/efectos de los fármacos , Microglía/fisiología , Saquinavir/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Western Blotting , Línea Celular , Interpretación Estadística de Datos , Femenino , Infecciones por VIH/virología , Inhibidores de la Proteasa del VIH/metabolismo , Lipopolisacáridos/farmacología , Nitritos/metabolismo , Embarazo , Cultivo Primario de Células , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Saquinavir/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
20.
J Pharm Sci ; 101(9): 3199-213, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22611042

RESUMEN

In an approach to overcome biological barriers mediated by P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A), a series of stereoisomeric valine-valine prodrugs of saquinavir (SQV) were synthesized and investigated with respect to affinity for efflux pump P-gp, and resistance to oxidative and hydrolytic enzymes. Cellular uptake and bidirectional transport in Caco-2 cells indicated that all peptide SQV conjugates can bypass P-gp-mediated efflux significantly, regardless of stereochemistry in promoieties. In comparison with D-configuration, L-configuration was favored for the interaction between prodrugs and rat hepatic CYP3A enzymes, and resulted in a relatively rapid clearance by CYP3A. Elimination half-life of SQV in rat liver microsomes was prolonged dramatically by sevenfold to 40-fold because of the prodrug modification with the rank order of D-valine-D-valine-SQV > D-valine-L-valine-SQV > L-valine-D-valine-SQV > L-valine-L-valine-SQV > D-valine-SQV > L-valine-SQV > SQV. Results of hydrolysis studies performed in rat intestinal homogenates and plasma indicated that prodrugs attached with D-valine exhibited significantly reduced biodegradation. In conclusion, the enhanced transepithelial accumulation and enzymatic stability observed by SQV peptide prodrug modification are found to be stereoselective. Specific stereoisomeric dipeptide prodrugs with optimized metabolic stability can be employed to improve oral bioavailability of SQV.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , Hidrolasas/metabolismo , Intestinos/enzimología , Hígado/enzimología , Profármacos/metabolismo , Saquinavir/metabolismo , Valina/metabolismo , Animales , Transporte Biológico , Biotransformación , Células CACO-2 , Química Farmacéutica , Inhibidores del Citocromo P-450 CYP3A , Diseño de Fármacos , Estabilidad de Medicamentos , Inhibidores Enzimáticos/farmacología , Inhibidores de la Proteasa del VIH/química , Semivida , Humanos , Hidrolasas/antagonistas & inhibidores , Cinética , Hígado/efectos de los fármacos , Masculino , Microsomas Hepáticos/enzimología , Modelos Biológicos , Estructura Molecular , Profármacos/química , Ratas , Ratas Sprague-Dawley , Saquinavir/análogos & derivados , Saquinavir/química , Especificidad por Sustrato , Tecnología Farmacéutica/métodos , Valina/análogos & derivados , Valina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...