Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 68(4): e0137323, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38380945

RESUMO

Protease inhibitors (PIs) remain an important component of antiretroviral therapy for the treatment of HIV-1 infection due to their high genetic barrier to resistance development. Nevertheless, the two most commonly prescribed HIV PIs, atazanavir and darunavir, still require co-administration with a pharmacokinetic boosting agent to maintain sufficient drug plasma levels which can lead to undesirable drug-drug interactions. Herein, we describe GS-9770, a novel investigational non-peptidomimetic HIV PI with unboosted once-daily oral dosing potential due to improvements in its metabolic stability and its pharmacokinetic properties in preclinical animal species. This compound demonstrates potent inhibitory activity and high on-target selectivity for recombinant HIV-1 protease versus other aspartic proteases tested. In cell culture, GS-9770 inhibits Gag polyprotein cleavage and shows nanomolar anti-HIV-1 potency in primary human cells permissive to HIV-1 infection and against a broad range of HIV subtypes. GS-9770 demonstrates an improved resistance profile against a panel of patient-derived HIV-1 isolates with resistance to atazanavir and darunavir. In resistance selection experiments, GS-9770 prevented the emergence of breakthrough HIV-1 variants at all fixed drug concentrations tested and required multiple protease substitutions to enable outgrowth of virus exposed to escalating concentrations of GS-9770. This compound also remained fully active against viruses resistant to drugs from other antiviral classes and showed no in vitro antagonism when combined pairwise with drugs from other antiretroviral classes. Collectively, these preclinical data identify GS-9770 as a potent, non-peptidomimetic once-daily oral HIV PI with potential to overcome the persistent requirement for pharmacological boosting with this class of antiretroviral agents.


Assuntos
Infecções por HIV , Inibidores da Protease de HIV , HIV-1 , Humanos , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/uso terapêutico , Darunavir/farmacologia , Darunavir/uso terapêutico , Sulfato de Atazanavir/farmacologia , Sulfato de Atazanavir/uso terapêutico , Farmacorresistência Viral , HIV-1/genética , Antirretrovirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Protease de HIV/genética , Protease de HIV/metabolismo
2.
Antimicrob Agents Chemother ; 64(10)2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32759267

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is already responsible for far more deaths than previous pathogenic coronaviruses (CoVs) from 2002 and 2012. The identification of clinically approved drugs to be repurposed to combat 2019 CoV disease (COVID-19) would allow the rapid implementation of potentially life-saving procedures. The major protease (Mpro) of SARS-CoV-2 is considered a promising target, based on previous results from related CoVs with lopinavir (LPV), an HIV protease inhibitor. However, limited evidence exists for other clinically approved antiretroviral protease inhibitors. Extensive use of atazanavir (ATV) as antiretroviral and previous evidence suggesting its bioavailability within the respiratory tract prompted us to study this molecule against SARS-CoV-2. Our results show that ATV docks in the active site of SARS-CoV-2 Mpro with greater strength than LPV, blocking Mpro activity. We confirmed that ATV inhibits SARS-CoV-2 replication, alone or in combination with ritonavir (RTV) in Vero cells and a human pulmonary epithelial cell line. ATV/RTV also impaired virus-induced enhancement of interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) levels. Together, our data strongly suggest that ATV and ATV/RTV should be considered among the candidate repurposed drugs undergoing clinical trials in the fight against COVID-19.


Assuntos
Antivirais/farmacologia , Sulfato de Atazanavir/farmacologia , Betacoronavirus/efeitos dos fármacos , Citocinas/metabolismo , Ritonavir/farmacologia , Animais , Sulfato de Atazanavir/química , Betacoronavirus/patogenicidade , Betacoronavirus/fisiologia , COVID-19 , Morte Celular/efeitos dos fármacos , Chlorocebus aethiops , Proteases 3C de Coronavírus , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/patologia , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Quimioterapia Combinada , Humanos , Inflamação/metabolismo , Inflamação/virologia , Lopinavir/farmacologia , Simulação de Acoplamento Molecular , Monócitos/virologia , Pandemias , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/metabolismo , Pneumonia Viral/patologia , Inibidores de Proteases/farmacologia , SARS-CoV-2 , Células Vero , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
3.
Nanomedicine ; 25: 102172, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32061722

RESUMO

We introduce the use of laser ablation to develop a multi-drug encapsulating theranostic nanoformulation for HIV-1 antiretroviral therapy. Laser ablated nanoformulations of ritonavir, atazanavir, and curcumin, a natural product that has both optical imaging and pharmacologic properties, were produced in an aqueous media containing Pluronic® F127. Cellular uptake was confirmed with the curcumin fluorescence signal localized in the cytoplasm. Formulations produced with F127 had improved water dispersibility, are ultrasmall in size (20-25 nm), exhibit enhanced cellular uptake in microglia, improve blood-brain barrier (BBB) crossing in an in vitro BBB model, and reduce viral p24 by 36 fold compared to formulations made without F127. This work demonstrates that these ultrasmall femtosecond laser-ablated nanoparticles are effective in delivering drugs across the BBB for brain therapy and show promise as an effective method to formulate nanoparticles for brain theranostics, reducing the need for organic solvents during preparation.


Assuntos
Composição de Medicamentos , Infecções por HIV/tratamento farmacológico , Nanopartículas/química , Nanomedicina Teranóstica/tendências , Sulfato de Atazanavir/síntese química , Sulfato de Atazanavir/química , Sulfato de Atazanavir/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Curcumina/síntese química , Curcumina/química , Curcumina/farmacologia , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Terapia a Laser , Nanopartículas/uso terapêutico , Medicina de Precisão , Ritonavir/síntese química , Ritonavir/química , Ritonavir/farmacologia
4.
BMC Res Notes ; 11(1): 825, 2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463602

RESUMO

OBJECTIVE: The low investment in research, diagnosis and treatment are factors that contribute to the continuity of Chagas' disease as a neglected tropical diseases (NTDs). In this context, the repositioning of drugs represents a useful strategy, in the search for new chemotherapeutic approaches for NTDs. HIV aspartic peptidase inhibitors (HIV IPs) are good candidates for drug repurposing. Here, we modeled the three dimensional structure of an aspartyl peptidase of Trypanosoma cruzi, the causative agent of Chagas' disease, aligned it to the HIV aspartyl peptidase and performed docking binding assays with the HIV PIs. RESULTS: The 3D structure confirmed the presence of acid aspartic residues, which are critical to enzyme activity. The docking experiment revealed that HIV IPs bind to the active site of the enzyme, being ritonavir and lopinavir the ones with greater affinity. Benznidazole presented the worst binding affinity, this drug is currently used in Chagas' disease treatment and was included as negative control. These results together with previous data on the trypanocidal effect of the HIV PIs support the hypothesis that a T. cruzi aspartyl peptidase can be the intracellular target of these inhibitors. However, the direct demonstration of the inhibition of T. cruzi aspartyl peptidase activity by HIV PIs is still a goal to be persuaded.


Assuntos
Fármacos Anti-HIV/farmacologia , Ácido Aspártico Proteases/química , Reposicionamento de Medicamentos , Simulação de Acoplamento Molecular , Peptídeo Hidrolases/química , Inibidores de Proteases/farmacologia , Trypanosoma cruzi/enzimologia , Sulfato de Atazanavir/farmacologia , Cristalografia por Raios X , Bases de Dados de Proteínas , HIV/efeitos dos fármacos , Nelfinavir/farmacologia , Conformação Proteica , Proteínas de Saccharomyces cerevisiae/química , Saquinavir/farmacologia
5.
Nanomedicine (Lond) ; 13(17): 2139-2154, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30129397

RESUMO

AIM: Pharmacologic agents that affect autophagy were tested for their abilities to enhance macrophage nanoformulated antiretroviral drug (ARV) depots and its slow release. METHODS: These agents included URMC-099, rapamycin, metformin, desmethylclomipramine, 2-hydroxy-ß-cyclodextrin (HBC) and clonidine. Each was administered with nanoformulated atazanavir (ATV) nanoparticles to human monocyte-derived macrophages. ARV retention, antiretroviral activity and nanocrystal autophagosomal formation were evaluated. RESULTS: URMC-099, HBC and clonidine retained ATV. HBC, URMC-099 and rapamycin improved intracellular ATV retention. URMC-099 proved superior among the group in affecting antiretroviral activities. CONCLUSION: Autophagy inducing agents, notably URMC-099, facilitate nanoformulated ARV depots and lead to sustained release and improved antiretroviral responses. As such, they may be considered for development as part of long acting antiretroviral treatment regimens.


Assuntos
Fármacos Anti-HIV/química , Sulfato de Atazanavir/farmacologia , Autofagia/efeitos dos fármacos , Portadores de Fármacos/química , Nanopartículas/química , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacologia , Sulfato de Atazanavir/administração & dosagem , Sulfato de Atazanavir/química , Sobrevivência Celular/efeitos dos fármacos , Clomipramina/administração & dosagem , Clomipramina/análogos & derivados , Clomipramina/química , Clomipramina/farmacologia , Clonidina/administração & dosagem , Clonidina/química , Clonidina/farmacologia , Interações Medicamentosas , Liberação Controlada de Fármacos , HIV-1/efeitos dos fármacos , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Metformina/administração & dosagem , Metformina/química , Metformina/farmacologia , Tamanho da Partícula , Piridinas/administração & dosagem , Piridinas/química , Piridinas/farmacologia , Pirróis/administração & dosagem , Pirróis/química , Pirróis/farmacologia , Sirolimo/administração & dosagem , Sirolimo/química , Sirolimo/farmacologia , Distribuição Tecidual , beta-Ciclodextrinas/administração & dosagem , beta-Ciclodextrinas/química , beta-Ciclodextrinas/farmacologia
6.
Reprod Toxicol ; 79: 57-65, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29859254

RESUMO

Atazanavir and ritonavir are preferred protease inhibitors frequently used in combination antiretroviral therapy for prevention of HIV mother-to-child transmission. Although their use is associated with higher risk of congenital anomalies, factors affecting atazanavir and ritonavir placental transfer are not known. This study is the first attempt to evaluate whether the placental drug efflux ATP-binding cassette (ABC) transporters, p-glycoprotein (ABCB1), breast cancer resistance protein (ABCG2), and/or multidrug resistance-associated proteins 2 (ABCC2), affect placental pharmacokinetics of atazanavir or ritonavir. Transport experiments across MDCKII cells expressing respective human ABC carrier showed that atazanavir is a substrate of ABCB1 and dual perfusion studies in a rat placenta confirmed this finding. In conclusion, we suggest that placental ABCB1 might reduce ATV maternal-to-fetal transfer and therefore represent a site for pharmacokinetic drug-drug interactions of ATV. Further studies in human placenta models are necessary to provide additional data closer to clinical environment.


Assuntos
Sulfato de Atazanavir/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Placenta/metabolismo , Ritonavir/farmacocinética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Sulfato de Atazanavir/farmacologia , Cães , Interações Medicamentosas , Feminino , Inibidores da Protease de HIV/farmacologia , Células Madin Darby de Rim Canino , Troca Materno-Fetal , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Gravidez , Ratos Wistar , Ritonavir/farmacologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-29463535

RESUMO

We identified four novel nonpeptidic human immunodeficiency virus type 1 (HIV-1) protease inhibitors (PIs), GRL-078, -079, -077, and -058, containing an alkylamine at the C-5 position of P2 tetrahydropyrano-tetrahydrofuran (Tp-THF) and a P2' cyclopropyl (Cp) (or isopropyl)-aminobenzothiazole (Abt) moiety. Their 50% effective concentrations (EC50s) were 2.5 to 30 nM against wild-type HIV-1NL4-3, 0.3 to 6.7 nM against HIV-2EHO, and 0.9 to 90 nM against laboratory-selected PI-resistant HIV-1 and clinical HIV-1 variants resistant to multiple FDA-approved PIs (HIVMDR). GRL-078, -079, -077, and -058 also effectively blocked the replication of HIV-1 variants highly resistant to darunavir (DRV) (HIVDRVrp51), with EC50s of 38, 62, 61, and 90 nM, respectively, while four FDA-approved PIs examined (amprenavir, atazanavir, lopinavir [LPV], and DRV) had virtually no activity (EC50s of >1,000 nM) against HIVDRVrp51 Structurally, GRL-078, -079, and -058 form strong hydrogen bond interactions between Tp-THF modified at C-5 and Asp29/Asp30/Gly48 of wild-type protease, while the P2' Cp-Abt group forms strong hydrogen bonds with Asp30'. The Tp-THF and Cp-Abt moieties also have good nonpolar interactions with protease residues located in the flap region. For selection with LPV and DRV by use of a mixture of 11 HIVMDR strains (HIV11MIX), HIV11MIX became highly resistant to LPV and DRV over 13 to 32 and 32 to 41 weeks, respectively. However, for selection with GRL-079 and GRL-058, HIV11MIX failed to replicate at >0.08 µM and >0.2 µM, respectively. Thermal stability results supported the highly favorable anti-HIV-1 potency of GRL-079 as well as other PIs. The present data strongly suggest that the P2 Tp-THF group modified at C-5 and the P2' Abt group contribute to the potent anti-HIV-1 profiles of the four PIs against HIV-1NL4-3 and a wide spectrum of HIVMDR strains.


Assuntos
Inibidores da Protease de HIV/farmacologia , Protease de HIV/metabolismo , HIV-1/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Sequência de Aminoácidos/genética , Sulfato de Atazanavir/farmacologia , Carbamatos/farmacologia , Linhagem Celular Tumoral , Darunavir/farmacologia , Farmacorresistência Viral Múltipla/genética , Furanos , Protease de HIV/genética , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Humanos , Lopinavir/farmacologia , Testes de Sensibilidade Microbiana , Sulfonamidas/farmacologia
8.
Basic Clin Pharmacol Toxicol ; 122(2): 199-207, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28816009

RESUMO

Atazanavir sulphate, an antiretroviral protease inhibitor, has been used to treat HIV/AIDS, but its ability to serve as an antipulmonary fibrosis (PF) agent remains unknown. In this study, the effects of atazanavir sulphate on various aspects of PF were examined and CoCl2 was used to induce the hypoxia-mimicking condition in vitro, including epithelial-mesenchymal transition (EMT) in A549 cells, endothelial-mesenchymal transition (EndMT) in human pulmonary microvascular endothelial cells (HPMECs), proliferation in human lung fibroblasts (HLF-1) and potential protective effects in human type I alveolar epithelial cells (AT I). Additionally, the effects of atazanavir sulphate were examined using a bleomycin (BLM)-induced pulmonary fibrosis model. After atazanavir sulphate treatment, in A549 cells and HPMECs, the expression of vimentin, HMGB1, Toll-like receptor 4 (TLR-4) and p-NF-κB decreased, while the expression of E-cadherin and VE-cadherin increased. In AT I cells, the expression of aquaporin 5 and RAGE were increased after atazanavir treatment. Proliferation of HLF-1 was reduced after atazanavir treatment, meanwhile the expression of hypoxia-inducible factor-1α (HIF-1α), prolyl hydroxylase domain protein 2 (PHD-2), HMGB1, TLR-9, p-NF-κB, collagen I and collagen III was decreased. In the BLM-induced pulmonary fibrosis rat model, atazanavir sulphate ameliorated PF by reducing pathological score, collagen deposition and the expression of α-SMA, HIF-1α, PHD-2, HMGB1, TLR-4, TLR-9 and p-NF-κB. In summary, our study supports the proposal that atazanavir sulphate may have a therapeutic potential in reducing the progression of pulmonary fibrosis by suppressing HMGB1/TLR signalling.


Assuntos
Sulfato de Atazanavir/farmacologia , Pulmão/efeitos dos fármacos , Fibrose Pulmonar/prevenção & controle , Células A549 , Animais , Bleomicina , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Citoproteção , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteína HMGB1/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/metabolismo
9.
J Clin Invest ; 127(3): 857-873, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28134625

RESUMO

Long-acting anti-HIV products can substantively change the standard of care for patients with HIV/AIDS. To this end, hydrophobic antiretroviral drugs (ARVs) were recently developed for parenteral administration at monthly or longer intervals. While shorter-acting hydrophilic drugs can be made into nanocarrier-encased prodrugs, the nanocarrier encasement must be boosted to establish long-acting ARV depots. The mixed-lineage kinase 3 (MLK-3) inhibitor URMC-099 provides this function by affecting autophagy. Here, we have shown that URMC-099 facilitates ARV sequestration and its antiretroviral responses by promoting the nuclear translocation of the transcription factor EB (TFEB). In monocyte-derived macrophages, URMC-099 induction of autophagy led to retention of nanoparticles containing the antiretroviral protease inhibitor atazanavir. These nanoparticles were localized within macrophage autophagosomes, leading to a 4-fold enhancement of mitochondrial and cell vitality. In rodents, URMC-099 activation of autophagy led to 50-fold increases in the plasma drug concentration of the viral integrase inhibitor dolutegravir. These data paralleled URMC-099-mediated induction of autophagy and the previously reported antiretroviral responses in HIV-1-infected humanized mice. We conclude that pharmacologic induction of autophagy provides a means to extend the action of a long-acting, slow, effective release of antiretroviral therapy.


Assuntos
Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Antirretrovirais/farmacologia , Autofagia/efeitos dos fármacos , HIV-1/metabolismo , Macrófagos/metabolismo , Nanopartículas , Síndrome da Imunodeficiência Adquirida/metabolismo , Animais , Sulfato de Atazanavir/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Feminino , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Masculino , Camundongos , Oxazinas , Piperazinas , Piridinas/farmacologia , Piridonas , Pirróis/farmacologia
10.
Parasitol Res ; 115(10): 3881-7, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27249967

RESUMO

Co-infection of Leishmaniasis, a neglected tropical disease, with human immunodeficiency virus (HIV) has hindered treatment efficacy. In this study, we aim to evaluate the antileishmanial activity of two protease inhibitors (darunavir and atazanavir) and four reverse transcriptase inhibitors (tenofovir, efavirenz, neviraprine, and delavirdine mesylate) on Leishmania infantum. The activity of different antiretrovirals combinations and of antiretroviral with miltefosine, a drug used on leishmaniasis treatment, was also evaluated. Only two non-nucleoside reverse transcriptase inhibitors (NNRTIs) were active on L. infantum. Efavirenz showed the best antileishmanial activity on promastigotes cells with IC50 value of 26.1 µM followed by delavirdine mesylate with an IC50 value of 136.2 µM. Neviraprine, tenofovir, atazanavir, and darunavir were not active at the concentrations tested (IC50 > 200 µM). The efavirenz also showed high antileishmanial activity on intramacrophage amastigotes with IC50 of 12.59 µM. The interaction of efavirenz with miltefosine improved antileishmanial activity on promastigotes and intracellular amastigotes (IC50 values of 11. 8 µM and 8.89 µM, respectively). These results suggest that combined-therapy including efavirenz and miltefosine could be alternative options for treating Leishmaniasis and Leishmania/HIV co-infections.


Assuntos
Antirretrovirais/farmacologia , Antiprotozoários/farmacologia , Infecções por HIV/tratamento farmacológico , Leishmania infantum/efeitos dos fármacos , Leishmaniose Visceral/tratamento farmacológico , Fosforilcolina/análogos & derivados , Alcinos , Animais , Sulfato de Atazanavir/farmacologia , Benzoxazinas/farmacologia , Coinfecção , Ciclopropanos , Darunavir/farmacologia , Delavirdina/farmacologia , Quimioterapia Combinada , Infecções por HIV/complicações , Humanos , Leishmaniose Visceral/complicações , Leishmaniose Visceral/parasitologia , Macrófagos/parasitologia , Macrófagos/virologia , Masculino , Camundongos Endogâmicos BALB C , Nevirapina/farmacologia , Fosforilcolina/farmacologia , Inibidores de Proteases/farmacologia , Inibidores da Transcriptase Reversa/farmacologia , Tenofovir/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA