Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Int J Mol Sci ; 23(20)2022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36293006

RESUMO

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.


Assuntos
Fármacos Anti-HIV , Inibidores da Protease de HIV , HIV-1 , Humanos , Inibidores da Protease de HIV/química , Protease de HIV/química , Darunavir/farmacologia , Indinavir/química , Indinavir/metabolismo , Indinavir/farmacologia , Nelfinavir/química , Nelfinavir/metabolismo , Nelfinavir/farmacologia , Ritonavir/química , Saquinavir/metabolismo , Saquinavir/farmacologia , Lopinavir/farmacologia , Sulfato de Atazanavir/farmacologia , Simulação de Acoplamento Molecular , Fármacos Anti-HIV/farmacologia , Aminoácidos/farmacologia
2.
J Chem Inf Model ; 62(5): 1328-1344, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35212226

RESUMO

A human immunodeficiency virus-1 (HIV-1) protease is a homodimeric aspartic protease essential for the replication of HIV. The HIV-1 protease is a target protein in drug discovery for antiretroviral therapy, and various inhibitor molecules of transition state analogues have been developed. However, serious drug-resistant mutants have emerged. For understanding the molecular mechanism of the drug resistance, an accurate examination of the impacts of the mutations on ligand binding and enzymatic activity is necessary. Here, we present a molecular simulation study on the ligand binding of indinavir, a potent transition state analogue inhibitor, to the wild-type protein and a V82T/I84V drug-resistant mutant of the HIV-1 protease. We employed a hybrid ab initio quantum mechanical/molecular mechanical (QM/MM) free-energy optimization technique which combines a highly accurate QM description of the ligand molecule and its interaction with statistically ample conformational sampling of the MM protein environment by long-time molecular dynamics simulations. Through the free-energy calculations of protonation states of catalytic groups at the binding pocket and of the ligand-binding affinity changes upon the mutations, we successfully reproduced the experimentally observed significant reduction of the binding affinity upon the drug-resistant mutations and elucidated the underlying molecular mechanism. The present study opens the way for understanding the molecular mechanism of drug resistance through the direct quantitative comparison of ligand binding and enzymatic reaction with the same accuracy.


Assuntos
Inibidores da Protease de HIV , Indinavir , Sítios de Ligação , Farmacorresistência Viral , Protease de HIV/metabolismo , Inibidores da Protease de HIV/química , Humanos , Indinavir/química , Indinavir/metabolismo , Indinavir/farmacologia , Simulação de Dinâmica Molecular , Mutação
3.
PLoS One ; 16(9): e0257916, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34570822

RESUMO

Determining the unbinding pathways of potential small molecule compounds from their target proteins is of great significance for designing efficacious treatment solutions. One of these potential compounds is the approved HIV-1 protease inhibitor, Indinavir, which has a weak effect on the HTLV-1 protease. In this work, by employing the SuMD method, we reconstructed the unbinding pathways of Indinavir from HIV and HTLV-1 proteases to compare and understand the mechanism of the unbinding and to discover the reasons for the lack of inhibitory activity of Indinavir against the HTLV-1 protease. We achieved multiple unbinding events from both HIV and HTLV-1 proteases in which the RMSD values of Indinavir reached over 40 Å. Also, we found that the mobility and fluctuations of the flap region are higher in the HTLV-1 protease, making the drug less stable. We realized that critically positioned aromatic residues such as Trp98/Trp98' and Phe67/Phe67' in the HTLV-1 protease could make strong π-Stacking interactions with Indinavir in the unbinding pathway, which are unfavorable for the stability of Indinavir in the active site. The details found in this study can make a reasonable explanation for the lack of inhibitory activity of this drug against HTLV-1 protease. We believe the details discovered in this work can help design more effective and selective inhibitors for the HTLV-1 protease.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Inibidores da Protease de HIV/metabolismo , Protease de HIV/metabolismo , Indinavir/metabolismo , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica
4.
Sci Rep ; 10(1): 16986, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046764

RESUMO

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.


Assuntos
Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Cisteína Endopeptidases/metabolismo , Reposicionamento de Medicamentos/métodos , Inibidores da Protease de HIV/farmacologia , Pneumonia Viral/tratamento farmacológico , Proteínas não Estruturais Virais/metabolismo , Betacoronavirus/metabolismo , Sítios de Ligação/efeitos dos fármacos , Fenômenos Biofísicos , COVID-19 , Domínio Catalítico/efeitos dos fármacos , Biologia Computacional , Proteases 3C de Coronavírus , Darunavir/metabolismo , Darunavir/farmacologia , Inibidores da Protease de HIV/metabolismo , Humanos , Indinavir/metabolismo , Indinavir/farmacologia , Lopinavir/metabolismo , Lopinavir/farmacologia , Simulação de Dinâmica Molecular , Nelfinavir/metabolismo , Nelfinavir/farmacologia , Pandemias , Ritonavir/metabolismo , Ritonavir/farmacologia , SARS-CoV-2 , Saquinavir/metabolismo , Saquinavir/farmacologia
5.
Phys Chem Chem Phys ; 22(8): 4464-4480, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32057044

RESUMO

Infection by human immunodeficiency virus type 1 (HIV-1) not only destroys the immune system bringing about acquired immune deficiency syndrome (AIDS), but also induces serious neurological diseases including behavioral abnormalities, motor dysfunction, toxoplasmosis, and HIV-1 associated dementia. The emergence of HIV-1 multidrug-resistant mutants has become a major problem in the therapy of patients with HIV-1 infection. Focusing on the wild type (WT) and G48T/L89M mutated forms of HIV-1 protease (HIV-1 PR) in complex with amprenavir (APV), indinavir (IDV), ritonavir (RTV), and nelfinavir (NFV), we have investigated the conformational dynamics and the resistance mechanism due to the G48T/L89M mutations by conducting a series of molecular dynamics (MD) simulations and free energy (MM-PBSA and solvated interaction energy (SIE)) analyses. The simulation results indicate that alterations in the side-chains of G48T/L89M mutated residues cause the inner active site to increase in volume and induce more curling of the flap tips, which provide the main contributions to weaker binding of inhibitors to the HIV-1 PR. The results of energy analysis reveal that the decrease in van der Waals interactions of inhibitors with the mutated PR relative to the wild-type (WT) PR mostly drives the drug resistance of mutations toward these four inhibitors. The energy decomposition analysis further indicates that the drug resistance of mutations can be mainly attributed to the change in van der Waals and electrostatic energy of some key residues (around Ala28/Ala28' and Ile50/Ile50'). Our work can give significant guidance to design a new generation of anti-AIDS inhibitors targeting PR in the therapy of patients with HIV-1 infection.


Assuntos
Protease de HIV/metabolismo , Simulação de Dinâmica Molecular , Fármacos Anti-HIV/química , Fármacos Anti-HIV/metabolismo , Carbamatos/química , Carbamatos/metabolismo , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Furanos , Protease de HIV/genética , Indinavir/química , Indinavir/metabolismo , Conformação Molecular , Mutação , Nelfinavir/química , Nelfinavir/metabolismo , Ligação Proteica , Ritonavir/química , Ritonavir/metabolismo , Sulfonamidas/química , Sulfonamidas/metabolismo
6.
Artif Cells Nanomed Biotechnol ; 47(1): 2123-2133, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31155961

RESUMO

Introduction: Indinavir (IDV) is a potent HIV protease inhibitor used in the treatment of human immunodeficiency virus (HIV). IDV is a weak base with limited aqueous solubility in its unprotonated form; therefore, solubility of IDV in the gastrointestinal tract fluids is the rate-limiting step of its absorption and onset of action. However, in many cases, drugs are not absorbed well in the gastrointestinal tract; polymer nanoparticles were recognized as an effective carrier system for drug encapsulation and are now studied as a vehicle for oral delivery of insoluble compounds. Preparation of methoxy poly (ethylene glycol)-poly (e-caprolactone) (mPEG-PCL) nanoparticles is among the strategies to overcome low bioavailability of drugs with poor aqueous solubility. Materials and method: The structure of the copolymers was characterized using 1H NMR, FTIR, DSC and GPC techniques. IDV loaded mPEG- PCL nanoparticles prepared by emulsification solvent evaporation method were optimized using D-optimal experimental design and were characterized by various techniques such as DLS, DSC, XRD, AFM and SEM. Using Caco-2 cells as a cellular model, we studied the cellular uptake and transport. Results: In vivo pharmacokinetic studies were performed in rats. The plasma AUC (0-t), t1/2 and Cmax of IDV-mPEG-PCL NPs were increased by 5.30, 5.57 and 1.37 fold compared to the IDV solution, respectively. Conclusion: The results of this study are promising for the use of biodegradable polymeric nanoparticles to improve oral drug delivery.


Assuntos
Portadores de Fármacos/química , Indinavir/administração & dosagem , Indinavir/farmacocinética , Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Administração Oral , Animais , Disponibilidade Biológica , Transporte Biológico , Células CACO-2 , Liberação Controlada de Fármacos , Humanos , Indinavir/química , Indinavir/metabolismo , Masculino , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley , Solubilidade , Distribuição Tecidual
7.
Chem Res Toxicol ; 31(2): 68-80, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29355304

RESUMO

Cytochromes P450 (CYPs) oxidize alkylated amines commonly found in drugs and other biologically active molecules, cleaving them into an amine and an aldehyde. Metabolic studies usually neglect to report or investigate aldehydes, even though they can be toxic. It is assumed that they are efficiently detoxified into carboxylic acids and alcohols. Nevertheless, some aldehydes are reactive and escape detoxification pathways to cause adverse events by forming DNA and protein adducts. Herein, we modeled N-dealkylations that produce both amine and aldehyde metabolites and then predicted the reactivity of the aldehyde. This model used a deep learning approach previously developed by our group to predict other types of drug metabolism. In this study, we trained the model to predict N-dealkylation by human liver microsomes (HLM), finding that including isozyme-specific metabolism data alongside HLM data significantly improved results. The final HLM model accurately predicted the site of N-dealkylation within metabolized substrates (97% top-two and 94% area under the ROC curve). Next, we combined the metabolism, metabolite structure prediction, and previously published reactivity models into a bioactivation model. This combined model predicted the structure of the most likely reactive metabolite of a small validation set of drug-like molecules known to be bioactivated by N-dealkylation. Applying this model to approved and withdrawn medicines, we found that aldehyde metabolites produced from N-dealkylation may explain the hepatotoxicity of several drugs: indinavir, piperacillin, verapamil, and ziprasidone. Our results suggest that N-dealkylation may be an under-appreciated bioactivation pathway, especially in clinical contexts where aldehyde detoxification pathways are inhibited. Moreover, this is the first report of a bioactivation model constructed by combining a metabolism and reactivity model. These results raise hope that more comprehensive models of bioactivation are possible. The model developed in this study is available at http://swami.wustl.edu/xenosite/ .


Assuntos
Indinavir/metabolismo , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Piperacilina/metabolismo , Piperazinas/metabolismo , Tiazóis/metabolismo , Verapamil/metabolismo , Aldeídos/química , Aldeídos/metabolismo , Aminas/química , Aminas/metabolismo , Remoção de Radical Alquila , Humanos , Indinavir/farmacologia , Fígado/efeitos dos fármacos , Microssomos Hepáticos/química , Microssomos Hepáticos/efeitos dos fármacos , Modelos Moleculares , Estrutura Molecular , Piperacilina/farmacologia , Piperazinas/farmacologia , Tiazóis/farmacologia , Verapamil/farmacologia
8.
ChemMedChem ; 10(2): 238-44, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25491400

RESUMO

Selectivity is a central aspect of lead optimization in the drug discovery process. Medicinal chemists often try to decrease molecular flexibility to improve selectivity, given the common belief that the two are interdependent. To investigate the relationship between polypharmacology and conformational flexibility, we mined the Protein Data Bank and constructed a dataset of pharmaceutically relevant ligands that crystallized in more than one protein target while binding to each co-crystallized receptor with similar in vitro affinities. After analyzing the molecular conformations of these 100 ligands, we found that 59 ligands bound to different protein targets without significantly changing conformation, suggesting that there is no distinct correlation between conformational flexibility and polypharmacology within our dataset. Ligands crystallized in similar proteins and highly ligand-efficient compounds with five or fewer rotatable bonds were less likely to adjust conformation when binding.


Assuntos
Proteínas/metabolismo , Benzamidas/química , Benzamidas/metabolismo , Bases de Dados de Proteínas , Desenho de Fármacos , Protease de HIV/química , Protease de HIV/metabolismo , Mesilato de Imatinib , Indinavir/química , Indinavir/metabolismo , Indóis/química , Indóis/metabolismo , Cinética , Ligantes , Piperazinas/química , Piperazinas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Proteínas Proto-Oncogênicas c-kit/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Ácidos Sulfônicos/química , Ácidos Sulfônicos/metabolismo
9.
J Chem Phys ; 141(23): 234111, 2014 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-25527923

RESUMO

A systematic examination of noncovalent interactions as modeled by wavefunction theory is presented in comparison to gold-standard quality benchmarks available for 345 interaction energies of 49 bimolecular complexes. Quantum chemical techniques examined include spin-component-scaling (SCS) variations on second-order perturbation theory (MP2) [SCS, SCS(N), SCS(MI)] and coupled cluster singles and doubles (CCSD) [SCS, SCS(MI)]; also, method combinations designed to improve dispersion contacts [DW-MP2, MP2C, MP2.5, DW-CCSD(T)-F12]; where available, explicitly correlated (F12) counterparts are also considered. Dunning basis sets augmented by diffuse functions are employed for all accessible ζ-levels; truncations of the diffuse space are also considered. After examination of both accuracy and performance for 394 model chemistries, SCS(MI)-MP2/cc-pVQZ can be recommended for general use, having good accuracy at low cost and no ill-effects such as imbalance between hydrogen-bonding and dispersion-dominated systems or non-parallelity across dissociation curves. Moreover, when benchmarking accuracy is desirable but gold-standard computations are unaffordable, this work recommends silver-standard [DW-CCSD(T**)-F12/aug-cc-pVDZ] and bronze-standard [MP2C-F12/aug-cc-pVDZ] model chemistries, which support accuracies of 0.05 and 0.16 kcal/mol and efficiencies of 97.3 and 5.5 h for adenine·thymine, respectively. Choice comparisons of wavefunction results with the best symmetry-adapted perturbation theory [T. M. Parker, L. A. Burns, R. M. Parrish, A. G. Ryno, and C. D. Sherrill, J. Chem. Phys. 140, 094106 (2014)] and density functional theory [L. A. Burns, Á. Vázquez-Mayagoitia, B. G. Sumpter, and C. D. Sherrill, J. Chem. Phys. 134, 084107 (2011)] methods previously studied for these databases are provided for readers' guidance.


Assuntos
Teoria Quântica , Protease de HIV/química , Protease de HIV/metabolismo , HIV-2/enzimologia , Ligação de Hidrogênio , Indinavir/química , Indinavir/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , Compostos Orgânicos/química , Padrões de Referência , Eletricidade Estática , Termodinâmica
10.
J Biol Chem ; 289(23): 16100-13, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24706759

RESUMO

Pharmacologic HIV protease inhibitors (PIs) and structurally related oligopeptides are known to reversibly bind and inactivate the insulin-responsive facilitative glucose transporter 4 (GLUT4). Several PIs exhibit isoform selectivity with little effect on GLUT1. The ability to target individual GLUT isoforms in an acute and reversible manner provides novel means both to investigate the contribution of individual GLUTs to health and disease and to develop targeted treatment of glucose-dependent diseases. To determine the molecular basis of transport inhibition, a series of chimeric proteins containing transmembrane and cytosolic domains from GLUT1 and GLUT4 and/or point mutations were generated and expressed in HEK293 cells. Structural integrity was confirmed via measurement of N-[2-[2-[2-[(N-biotinylcaproylamino)ethoxy)ethoxyl]-4-[2-(trifluoromethyl)-3H-diazirin-3-yl]benzoyl]-1,3-bis(mannopyranosyl-4-yloxy)-2-propylamine (ATB-BMPA) labeling of the chimeric proteins in low density microsome fractions isolated from stably transfected 293 cells. Functional integrity was assessed via measurement of zero-trans 2-deoxyglucose (2-DOG) uptake. ATB-BMPA labeling studies and 2-DOG uptake revealed that transmembrane helices 1 and 5 contain amino acid residues that influence inhibitor access to the transporter binding domain. Substitution of Thr-30 and His-160 in GLUT1 to the corresponding positions in GLUT4 is sufficient to completely transform GLUT1 into GLUT4 with respect to indinavir inhibition of 2-DOG uptake and ATB-BMPA binding. These data provide a structural basis for the selectivity of PIs toward GLUT4 over GLUT1 that can be used in ongoing novel drug design.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Inibidores da Protease de HIV/farmacologia , Isoformas de Proteínas/antagonistas & inibidores , Sequência de Aminoácidos , Linhagem Celular , Clonagem Molecular , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Inibidores da Protease de HIV/metabolismo , Humanos , Indinavir/metabolismo , Indinavir/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homologia de Sequência de Aminoácidos
11.
J Chem Inf Model ; 53(8): 2141-53, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23834142

RESUMO

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.


Assuntos
Fármacos Anti-HIV/metabolismo , Farmacorresistência Viral/genética , Inibidores da Protease de HIV/metabolismo , Protease de HIV/metabolismo , HIV-1/efeitos dos fármacos , Simulação de Dinâmica Molecular , Mutação , Fármacos Anti-HIV/farmacologia , Carbamatos/metabolismo , Carbamatos/farmacologia , Análise por Conglomerados , Darunavir , Furanos , Protease de HIV/química , Protease de HIV/genética , Inibidores da Protease de HIV/farmacologia , HIV-1/enzimologia , HIV-1/genética , Ligação de Hidrogênio , Indinavir/metabolismo , Indinavir/farmacologia , Conformação Proteica , Saquinavir/metabolismo , Saquinavir/farmacologia , Sulfonamidas/metabolismo , Sulfonamidas/farmacologia , Termodinâmica
12.
Anesthesiology ; 116(2): 432-47, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22273859

RESUMO

BACKGROUND: Methadone disposition and pharmacodynamics are highly susceptible to interactions with antiretroviral drugs. Methadone clearance and drug interactions have been attributed to cytochrome P4503A4 (CYP3A4), but actual mechanisms are unknown. Drug interactions can be clinically and mechanistically informative. This investigation assessed effects of the protease inhibitor indinavir on methadone pharmacokinetics and pharmacodynamics, hepatic and intestinal CYP3A4/5 activity (using alfentanil), and intestinal transporter activity (using fexofenadine). METHODS: Twelve healthy volunteers underwent a sequential crossover. On three consecutive days they received oral alfentanil plus fexofenadine, intravenous alfentanil, and intravenous plus oral (deuterium-labeled) methadone. This was repeated after 2 weeks of indinavir. Plasma and urine analytes were measured by mass spectrometry. Opioid effects were measured by miosis. RESULTS: Indinavir significantly inhibited hepatic and first-pass CYP3A activity. Intravenous alfentanil systemic clearance and hepatic extraction were reduced to 40-50% of control, apparent oral clearance to 30% of control, and intestinal extraction decreased by half, indicating 50% and 70% inhibition of hepatic and first-pass CYP3A activity. Indinavir increased fexofenadine area under the plasma concentration-time curve 3-fold, suggesting significant P-glycoprotein inhibition. Indinavir had no significant effects on methadone plasma concentrations, methadone N-demethylation, systemic or apparent oral clearance, renal clearance, hepatic extraction or clearance, or bioavailability. Methadone plasma concentration-effect relationships were unaffected by indinavir. CONCLUSIONS: Despite significant inhibition of hepatic and intestinal CYP3A activity, indinavir had no effect on methadone N-demethylation and clearance, suggesting little or no role for CYP3A in clinical disposition of single-dose methadone. Inhibition of gastrointestinal transporter activity had no influence of methadone bioavailability.


Assuntos
Inibidores do Citocromo P-450 CYP3A , Citocromo P-450 CYP3A/metabolismo , Indinavir/metabolismo , Intestinos/enzimologia , Fígado/enzimologia , Metadona/metabolismo , Adolescente , Adulto , Estudos Cross-Over , Interações Medicamentosas/fisiologia , Feminino , Humanos , Indinavir/farmacologia , Intestinos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Taxa de Depuração Metabólica/efeitos dos fármacos , Taxa de Depuração Metabólica/fisiologia , Metadona/farmacologia , Adulto Jovem
13.
J Chem Phys ; 135(8): 085101, 2011 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-21895219

RESUMO

An energy expansion (binding energy decomposition into n-body interaction terms for n ≥ 2) to express the receptor-ligand binding energy for the fragmented HIV II protease-Indinavir system is described to address the role of cooperativity in ligand binding. The outcome of this energy expansion is compared to the total receptor-ligand binding energy at the Hartree-Fock, density functional theory, and semiempirical levels of theory. We find that the sum of the pairwise interaction energies approximates the total binding energy to ∼82% for HF and to >95% for both the M06-L density functional and PM6-DH2 semiempirical method. The contribution of the three-body interactions amounts to 18.7%, 3.8%, and 1.4% for HF, M06-L, and PM6-DH2, respectively. We find that the expansion can be safely truncated after n=3. That is, the contribution of the interactions involving more than three parties to the total binding energy of Indinavir to the HIV II protease receptor is negligible. Overall, we find that the two-body terms represent a good approximation to the total binding energy of the system, which points to pairwise additivity in the present case. This basic principle of pairwise additivity is utilized in fragment-based drug design approaches and our results support its continued use. The present results can also aid in the validation of non-bonded terms contained within common force fields and in the correction of systematic errors in physics-based score functions.


Assuntos
Inibidores da Protease de HIV/metabolismo , Protease de HIV/metabolismo , HIV-2/enzimologia , Indinavir/metabolismo , Proteínas/metabolismo , Ligantes , Modelos Moleculares
14.
J Control Release ; 150(2): 204-11, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21108978

RESUMO

Long-term antiretroviral therapy (ART) for human immunodeficiency virus type one (HIV-1) infection shows limitations in pharmacokinetics and biodistribution while inducing metabolic and cytotoxic aberrations. In turn, ART commonly requires complex dosing schedules and leads to the emergence of viral resistance and treatment failures. We posit that the development of nanoformulated ART could preclude such limitations and affect improved clinical outcomes. To this end, we wet-milled 20 nanoparticle formulations of crystalline indinavir, ritonavir, atazanavir, and efavirenz, collectively referred to as "nanoART," then assessed their performance using a range of physicochemical and biological tests. These tests were based on cell-nanoparticle interactions using monocyte-derived macrophages and their abilities to uptake and release nanoformulated drugs and affect viral replication. We demonstrate that physical characteristics such as particle size, surfactant coating, surface charge, and most importantly shape are predictors of cell uptake and antiretroviral efficacy. These studies bring this line of research a step closer to developing nanoART that can be used in the clinic to affect the course of HIV-1 infection.


Assuntos
Antirretrovirais/administração & dosagem , Antirretrovirais/farmacologia , HIV-1/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Nanopartículas/química , Alcinos , Antirretrovirais/metabolismo , Sulfato de Atazanavir , Benzoxazinas/administração & dosagem , Benzoxazinas/metabolismo , Benzoxazinas/farmacologia , Ciclopropanos , Humanos , Indinavir/administração & dosagem , Indinavir/metabolismo , Indinavir/farmacologia , Macrófagos/metabolismo , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Oligopeptídeos/administração & dosagem , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Tamanho da Partícula , Piridinas/administração & dosagem , Piridinas/metabolismo , Piridinas/farmacologia , Ritonavir/administração & dosagem , Ritonavir/metabolismo , Ritonavir/farmacologia , Eletricidade Estática , Tensoativos/química , Replicação Viral/efeitos dos fármacos
15.
Retrovirology ; 7: 78, 2010 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-20868521

RESUMO

BACKGROUND: HIV reservoirs are rapidly established after infection, and the effect of HAART initiated very early during acute infection on HIV reservoirs remains poorly documented, particularly in tissue known to actively replicate the virus. In this context, we used the model of experimental infection of macaques with pathogenic SIV to assess in different tissues: (i) the effect of a short term HAART initiated at different stages during acute infection on viral dissemination and replication, and (ii) the local concentration of antiviral drugs. RESULTS: Here, we show that early treatment with AZT/3TC/IDV initiated either within 4 hours after intravenous infection of macaques with SIVmac251 (as a post exposure prophylaxis) or before viremia peak (7 days post-infection [pi]), had a strong impact on SIV production and dissemination in all tissues but did not prevent infection. When treatment was initiated after the viremia peak (14 days pi) or during early chronic infection (150 days pi), significant viral replication persists in the peripheral lymph nodes and the spleen of treated macaques despite a strong effect of treatment on viremia and gut associated lymphoid tissues. In these animals, the level of virus persistence in tissues was inversely correlated with local concentrations of 3TC: high concentrations of 3TC were measured in the gut whereas low concentrations were observed in the secondary lymphoid tissues. IDV, like 3TC, showed much higher concentration in the colon than in the spleen. AZT concentration was below the quantification threshold in all tissues studied. CONCLUSIONS: Our results suggest that limited antiviral drug diffusion in secondary lymphoid tissues may allow persistent viral replication in these tissues and could represent an obstacle to HIV prevention and eradication.


Assuntos
Inibidores da Protease de HIV/uso terapêutico , Indinavir/uso terapêutico , Lamivudina/uso terapêutico , Inibidores da Transcriptase Reversa/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia , Zidovudina/uso terapêutico , Administração Cutânea , Administração Oral , Animais , Terapia Antirretroviral de Alta Atividade , Esquema de Medicação , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/metabolismo , Indinavir/administração & dosagem , Indinavir/metabolismo , Lamivudina/administração & dosagem , Lamivudina/metabolismo , Macaca fascicularis , Masculino , Inibidores da Transcriptase Reversa/administração & dosagem , Inibidores da Transcriptase Reversa/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Fatores de Tempo , Distribuição Tecidual , Viremia/tratamento farmacológico , Viremia/metabolismo , Viremia/virologia , Zidovudina/administração & dosagem , Zidovudina/metabolismo
16.
Vopr Virusol ; 55(1): 10-5, 2010.
Artigo em Russo | MEDLINE | ID: mdl-20364665

RESUMO

HIV-1 virions are as immature noninfectious particles lacking a central core. Shortly after budding, virions temporally mature and acquire cores and infectious activity. The cause of maturation remains poorly studied. We have revealed that the virions produced early after infection following 24-36 hours, never mature and remain noninfectious, and only virions produced 48-72 hours after infection mature. The mature virions contain 3 times more genomic viral RNA than "early" virus. The "early" virions contain the same proteolytically cleaved Gag proteins as mature virions in contrast to the accepted version. The virus protease inhibitor Indinavir sulfate (IS) fully blocks infectivity when added early after infection. The early proteolysis of Gag precursor in the infected cells and inclusion into the virions of cellularly cleaved matrix protein (cMA) are shown in the IS-treated cells. cMA is associated with genomic viral RNA.


Assuntos
Infecções por HIV/virologia , HIV-1/crescimento & desenvolvimento , HIV-1/metabolismo , Precursores de Proteínas/metabolismo , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacologia , HIV-1/patogenicidade , Humanos , Indinavir/metabolismo , Indinavir/farmacologia , Virulência/efeitos dos fármacos , Eliminação de Partículas Virais
17.
Drug Metab Dispos ; 38(5): 841-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20100816

RESUMO

The 2-methyl substituted indole, 2MI [2-(4-(4-(2,4-dichlorophenylsulfonamido)-2-methyl-1H-indol-5-yloxy)-3-methoxyphenyl)acetic acid] is a potent dual inhibitor of 1) chemoattractant receptor-homologous molecule expressed on T-helper type-2 cells and 2) d-prostanoid receptor. During evaluation as a potential treatment for asthma and allergic rhinitis, 2MI was identified as a mechanism-based inactivator of CYP3A4 in vitro. The inactivation was shown to be irreversible by dialysis and accompanied by an NADPH-dependent increase in 2MI covalent binding to a 55- to 60-kDa microsomal protein, consistent with irreversible binding to CYP3A4. Two glutathione (GSH) adducts, G1 and G2, were identified in vitro, and the more abundant adduct (G1) was unambiguously determined via NMR to be GSH adducted to the 3-position of the 2-methylindole moiety. The potential for a clinical drug-drug interaction arising from mechanism-based inactivation of CYP3A4 by 2MI was predicted using a steady-state model, and a 4.3- to 7.5-fold increase in the exposure of midazolam was predicted at anticipated therapeutic concentrations. To better assess the potential for in vivo drug-drug interactions, the Sprague-Dawley rat was used as an in vivo model. An excellent in vitro-in vivo correlation was observed for the reduction in enzyme steady-state concentration (E'(ss/Ess)) as well as the change in the exposure of a prototypical CYP3A substrate, indinavir (area under the curve (AUC) for indinavir/AUC). In summary, 2MI was identified as a potent mechanism-based inactivator of CYP3A and was predicted to elicit a clinically relevant drug-drug interaction in humans at an anticipated therapeutic concentration.


Assuntos
Citocromo P-450 CYP3A/efeitos dos fármacos , Citocromo P-450 CYP3A/metabolismo , Glutationa/metabolismo , Indóis/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores , Animais , Área Sob a Curva , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/efeitos dos fármacos , Hidrocarboneto de Aril Hidroxilases/metabolismo , Biocatálise/efeitos dos fármacos , Inibidores do Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Indinavir/metabolismo , Indinavir/farmacocinética , Indóis/farmacologia , Cinética , Espectroscopia de Ressonância Magnética , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Midazolam/metabolismo , Midazolam/farmacocinética , Modelos Biológicos , Estrutura Molecular , NADP/metabolismo , Farmacocinética , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
18.
J Pharm Sci ; 99(6): 2886-98, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20014428

RESUMO

Using sandwich-cultured hepatocytes from rat, dog, pig, and human, we investigated the species-specificity of interaction of HIV protease inhibitors (PI) with in vitro hepatic accumulation of the bile salt analogue cholyl-glycylamido-fluorescein (CGamF). Extracellular sodium depletion or coincubation with the OATP/Oatp inhibitors rifampicin and digoxin revealed that about 35% of active CGamF accumulation was mediated by Ntcp/NTCP in rat and human hepatocytes, while the contribution of this sodium-dependent transporter reached 50-60% in dog and pig hepatocytes. One or more sodium-independent transporters, likely belonging to the Oatp/OATP family, constitute a major transport mechanism for CGamF accumulation. Various HIV PI (0.5, 5, 25 microM) exhibited pronounced species differences in their interaction with active CGamF accumulation (1 microM), although some similarity was observed between the dog and human interaction profiles when HIV PI were tested at 0.5 microM. Atazanavir, indinavir, and darunavir were the most potent inhibitors of CGamF accumulation in human hepatocytes. Potent inhibition of CGamF accumulation by ritonavir in rat hepatocytes contrasted with a weak effect in human hepatocytes. Thorough characterization of in vitro disposition of probe substrates in preclinical species compared to human hepatocytes will ultimately support a better insight in species-specific mechanisms underlying drug interactions and drug-mediated toxicity.


Assuntos
Inibidores da Protease de HIV/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/farmacologia , Transporte Biológico/efeitos dos fármacos , Darunavir , Cães , Formas de Dosagem , Feminino , Fluoresceínas , Inibidores da Protease de HIV/metabolismo , Humanos , Indinavir/metabolismo , Indinavir/farmacologia , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Wistar , Ritonavir/metabolismo , Ritonavir/farmacologia , Especificidade da Espécie , Sulfonamidas/metabolismo , Sulfonamidas/farmacologia , Sus scrofa
19.
Rapid Commun Mass Spectrom ; 23(11): 1683-93, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19418486

RESUMO

Multiple ion monitoring (MIM)-dependent acquisition with a triple quadrupole-linear ion trap mass spectrometer (Q-trap) was previously developed for drug metabolite profiling. In the analysis, multiple predicted metabolite ions are monitored in both Q1 and Q3 regardless of their fragmentations. The collision energy in Q2 is set to a low value to minimize fragmentation. Once an expected metabolite is detected by MIM, enhanced product ion (EPI) spectral acquisition of the metabolite is triggered. To analyze in vitro metabolites, MIM-EPI retains the sensitivity and selectivity similar to that of multiple reaction monitoring (MRM)-EPI in the analysis of in vitro metabolites. Here we present an improved approach utilizing MIM-EPI for data acquisition and multiple data mining techniques for detection of metabolite ions and recovery of their MS/MS spectra. The postacquisition data processing tools included extracted ion chromatographic analysis, product ion filtering and neutral loss filtering. The effectiveness of this approach was evaluated by analyzing oxidative metabolites of indinavir and glutathione (GSH) conjugates of clozapine and 4-ethylphenol in liver microsome incubations. Results showed that the MIM-EPI-based data mining approach allowed for comprehensive detection of metabolites based on predicted protonated molecules, product ions or neutral losses without predetermination of the parent drug MS/MS spectra. Additionally, it enabled metabolite detection and MS/MS acquisition in a single injection. This approach is potentially useful in high-throughout screening of metabolic soft spots and reactive metabolites at the drug discovery stage.


Assuntos
Clozapina/metabolismo , Indinavir/metabolismo , Microssomos Hepáticos/química , Fenóis/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos , Animais , Clozapina/química , Haplorrinos , Indinavir/química , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Fenóis/química , Espectrometria de Massas por Ionização por Electrospray/instrumentação
20.
Anesthesiology ; 110(3): 660-72, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19225389

RESUMO

BACKGROUND: Methadone clearance is highly variable, and drug interactions are problematic. Both have been attributed to CYP3A, but actual mechanisms are unknown. Drug interactions can provide such mechanistic information. Ritonavir/indinavir, one of the earliest protease inhibitor combinations, may inhibit CYP3A. We assessed ritonavir/indinavir effects on methadone pharmacokinetics and pharmacodynamics, intestinal and hepatic CYP3A activity, and intestinal transporters (P-glycoprotein) activity. CYP3A and transporters were assessed with alfentanil and fexofenadine, respectively. METHODS: Twelve healthy human immunodeficiency virus-negative volunteers underwent a sequential three-part crossover. On three consecutive days, they received oral alfentanil/fexofenadine, intravenous alfentanil, and intravenous plus oral (deuterium-labeled) methadone, repeated after acute (3 days) and steady-state (2 weeks) ritonavir/indinavir. Plasma and urine analytes were measured by mass spectrometry. Opioid effects were assessed by miosis. RESULTS: Alfentanil apparent oral clearance was inhibited more than 97% by both acute and steady-state ritonavir/indinavir, and systemic clearance was inhibited more than 90% due to diminished hepatic and intestinal extraction. Ritonavir/indinavir increased fexofenadine area under the plasma concentration-time curve four- to five-fold, suggesting significant inhibition of gastrointestinal P-glycoprotein. Ritonavir/indinavir slightly increased methadone N-demethylation, but it had no significant effects on methadone plasma concentrations or on systemic or apparent oral clearance, renal clearance, hepatic extraction or clearance, or bioavailability. Ritonavir/indinavir had no significant effects on methadone plasma concentration-effect relationships. CONCLUSIONS: Inhibition of both hepatic and intestinal CYP3A activity is responsible for ritonavir/indinavir drug interactions. Methadone disposition was unchanged, despite profound inhibition of CYP3A activity, suggesting little or no role for CYP3A in clinical methadone metabolism and clearance. Methadone bioavailability was unchanged, despite inhibition of gastrointestinal P-glycoprotein activity, suggesting that this transporter does not limit methadone intestinal absorption.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Trato Gastrointestinal/metabolismo , Indinavir/metabolismo , Absorção Intestinal/fisiologia , Metadona/farmacocinética , Ritonavir/metabolismo , Adolescente , Adulto , Estudos Cross-Over , Interações Medicamentosas/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Humanos , Absorção Intestinal/efeitos dos fármacos , Masculino , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...