Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ther Adv Infect Dis ; 10: 20499361231198335, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37720384

RESUMO

Background: A growing interest in long-term sequelae of COVID-19 has prompted several systematic literature reviews (SLRs) to evaluate long-COVID-19 effects. However, many of these reviews lack in-depth information on the timing, duration, and severity of these conditions. Objectives: Our aim was to synthesize both qualitative and quantitative evidence on prevalence and outcomes of long-term effect of COVID-19 through an umbrella review. Design: Umbrella review of relevant SLRs on long-COVID-19 in terms of prolonged symptoms and clinical conditions, and comprehensively synthesized the latest existing evidence. Data Sources and Methods: We systematically identified and appraised prior systematic reviews/meta-analyses using MEDLINE, Embase, and Cochrane database of systematic review from 2020 to 2021 following the preferred reporting items for systematic reviews and meta-analyses guidance. We summarized and categorized all relevant clinical symptoms and outcomes in adults with COVID-19 using the Medical Dictionary for Regulatory Activities System Organ Class (MedDRA SOC). Results: We identified 967 systematic reviews/meta-analyses; 36 were retained for final data extraction. The most prevalent SOC were social circumstances (40%), blood and lymphatic system disorders (39%), and metabolism and nutrition disorder (38%). The most frequently reported SOC outcomes within each MedDRA category were poor quality of life (59%), wheezing and dyspnea (19-49%), fatigue (30-64%), chest pain (16%), decreased or loss of appetite (14-17%), abdominal discomfort or digestive disorder (12-18%), arthralgia with or without myalgia (16-24%), paresthesia (27%) and hair loss (14-25%), and hearing loss or tinnitus (15%). Conclusion: This study confirmed a high prevalence of several long COVID-19 outcomes according to the MedDRA categories and indicated that the majority of evidence was rated as moderate to low. Registration: The review was registered at PROSPERO (https://www.crd.york.ac.uk/prospero/) (CRD42022303557).

2.
ERJ Open Res ; 8(4)2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36411792

RESUMO

Owing to condensed development processes, expanding evidence and differences in healthcare system characteristics, many COVID-19 guidelines differ in their quality and treatment recommendations, which has consequences for clinical practice. This review aimed to identify COVID-19 treatment guidelines, assess their quality and summarise their recommendations. Guidelines were identified for five therapies most commonly used among inpatients with COVID-19 (remdesivir, dexamethasone, tocilizumab, baricitinib and casirivimab/imdevimab) from 11 countries. Guideline quality was assessed using the Appraisal of Guidelines for Research and Evaluation II (AGREE-II) tool. Full details of recommendations and supporting evidence were analysed for high-quality guidelines, defined as those scoring ≥50% in Domain 3 (Rigour of Development) of AGREE-II. Overall, guidelines differed substantially in their quality and, even among high-quality guidelines using the same evidence, recommendations regarding specific therapeutics varied. Potential reasons for this heterogeneity, including the availability and consistency of clinical data, visibility of trial end-points and context-specific factors, are discussed.

3.
Nature ; 584(7822): 614-618, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32612233

RESUMO

Oral antiretroviral agents provide life-saving treatments for millions of people living with HIV, and can prevent new infections via pre-exposure prophylaxis1-5. However, some people living with HIV who are heavily treatment-experienced have limited or no treatment options, owing to multidrug resistance6. In addition, suboptimal adherence to oral daily regimens can negatively affect the outcome of treatment-which contributes to virologic failure, resistance generation and viral transmission-as well as of pre-exposure prophylaxis, leading to new infections1,2,4,7-9. Long-acting agents from new antiretroviral classes can provide much-needed treatment options for people living with HIV who are heavily treatment-experienced, and additionally can improve adherence10. Here we describe GS-6207, a small molecule that disrupts the functions of HIV capsid protein and is amenable to long-acting therapy owing to its high potency, low in vivo systemic clearance and slow release kinetics from the subcutaneous injection site. Drawing on X-ray crystallographic information, we designed GS-6207 to bind tightly at a conserved interface between capsid protein monomers, where it interferes with capsid-protein-mediated interactions between proteins that are essential for multiple phases of the viral replication cycle. GS-6207 exhibits antiviral activity at picomolar concentrations against all subtypes of HIV-1 that we tested, and shows high synergy and no cross-resistance with approved antiretroviral drugs. In phase-1 clinical studies, monotherapy with a single subcutaneous dose of GS-6207 (450 mg) resulted in a mean log10-transformed reduction of plasma viral load of 2.2 after 9 days, and showed sustained plasma exposure at antivirally active concentrations for more than 6 months. These results provide clinical validation for therapies that target the functions of HIV capsid protein, and demonstrate the potential of GS-6207 as a long-acting agent to treat or prevent infection with HIV.


Assuntos
Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Proteínas do Capsídeo/antagonistas & inibidores , HIV-1/efeitos dos fármacos , Adolescente , Adulto , Fármacos Anti-HIV/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Células Cultivadas , Farmacorresistência Viral/genética , Feminino , HIV-1/crescimento & desenvolvimento , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Replicação Viral/efeitos dos fármacos , Adulto Jovem
4.
Artigo em Inglês | MEDLINE | ID: mdl-27799218

RESUMO

GS-9620 is a potent and selective oral Toll-like receptor 7 (TLR7) agonist that directly activates plasmacytoid dendritic cells (pDCs). GS-9620 suppressed hepatitis B virus (HBV) in animal models of chronic infection and transiently activated HIV expression ex vivo in latently infected peripheral blood mononuclear cells (PBMCs) from virally suppressed patients. Currently, GS-9620 is under clinical evaluation for treating chronic HBV infection and for reducing latent reservoirs in virally suppressed HIV-infected patients. Here, we investigated the in vitro anti-HIV-1 activity of GS-9620. GS-9620 potently inhibited viral replication in PBMCs, particularly when it was added 24 to 48 h prior to HIV infection (50% effective concentration = 27 nM). Depletion of pDCs but not other immune cell subsets from PBMC cultures suppressed GS-9620 antiviral activity. Although GS-9620 was inactive against HIV in purified CD4+ T cells and macrophages, HIV replication was potently inhibited by conditioned medium derived from GS-9620-treated pDC cultures when added to CD4+ T cells prior to infection. This suggests that GS-9620-mediated stimulation of PBMCs induced the production of a soluble factor(s) inhibiting HIV replication in trans GS-9620-treated PBMCs primarily showed increased production of interferon alpha (IFN-α), and cotreatment with IFN-α-blocking antibodies reversed the HIV-1-inhibitory effect of GS-9620. Additional studies demonstrated that GS-9620 inhibited a postentry event in HIV replication at a step coincident with or prior to reverse transcription. The simultaneous activation of HIV-1 expression and inhibition of HIV-1 replication are important considerations for the clinical evaluation of GS-9620 since these antiviral effects may help restrict potential local HIV spread upon in vivo latency reversal.


Assuntos
Antivirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Leucócitos Mononucleares/virologia , Pteridinas/uso terapêutico , Receptor 7 Toll-Like/antagonistas & inibidores , Anticorpos/uso terapêutico , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Interferon-alfa/antagonistas & inibidores , Interferons/metabolismo , Interleucina-6/metabolismo , Leucócitos Mononucleares/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Replicação Viral/efeitos dos fármacos
5.
Antimicrob Agents Chemother ; 60(12): 7086-7097, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27645238

RESUMO

Bictegravir (BIC; GS-9883), a novel, potent, once-daily, unboosted inhibitor of HIV-1 integrase (IN), specifically targets IN strand transfer activity (50% inhibitory concentration [IC50] of 7.5 ± 0.3 nM) and HIV-1 integration in cells. BIC exhibits potent and selective in vitro antiretroviral activity in both T-cell lines and primary human T lymphocytes, with 50% effective concentrations ranging from 1.5 to 2.4 nM and selectivity indices up to 8,700 relative to cytotoxicity. BIC exhibits synergistic in vitro antiviral effects in pairwise combinations with tenofovir alafenamide, emtricitabine, or darunavir and maintains potent antiviral activity against HIV-1 variants resistant to other classes of antiretrovirals. BIC displayed an in vitro resistance profile that was markedly improved compared to the integrase strand transfer inhibitors (INSTIs) raltegravir (RAL) and elvitegravir (EVG), and comparable to that of dolutegravir (DTG), against nine INSTI-resistant site-directed HIV-1 mutants. BIC displayed statistically improved antiviral activity relative to EVG, RAL, and DTG against a panel of 47 patient-derived HIV-1 isolates with high-level INSTI resistance; 13 of 47 tested isolates exhibited >2-fold lower resistance to BIC than DTG. In dose-escalation experiments conducted in vitro, BIC and DTG exhibited higher barriers to resistance than EVG, selecting for HIV-1 variants with reduced phenotypic susceptibility at days 71, 87, and 20, respectively. A recombinant virus with the BIC-selected M50I/R263K dual mutations in IN exhibited only 2.8-fold reduced susceptibility to BIC compared to wild-type virus. All BIC-selected variants exhibited low to intermediate levels of cross-resistance to RAL, DTG, and EVG (<8-fold) but remained susceptible to other classes of antiretrovirals. A high barrier to in vitro resistance emergence for both BIC and DTG was also observed in viral breakthrough studies in the presence of constant clinically relevant drug concentrations. The overall virologic profile of BIC supports its ongoing clinical investigation in combination with other antiretroviral agents for both treatment-naive and -experienced HIV-infected patients.


Assuntos
Farmacorresistência Viral/efeitos dos fármacos , Inibidores de Integrase de HIV/farmacologia , Integrase de HIV/metabolismo , HIV-1/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Amidas , Fármacos Anti-HIV/farmacologia , Linhagem Celular , Sinergismo Farmacológico , Integrase de HIV/genética , HIV-1/genética , HIV-1/isolamento & purificação , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Mutação , Oxazinas , Piperazinas , Piridonas , Raltegravir Potássico/farmacologia
6.
Protein Eng Des Sel ; 29(5): 159-67, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26968452

RESUMO

The CTLA4-Ig therapeutics abatacept and belatacept inhibit CD28-mediated T cell activation by binding CD80 (B7-1) and CD86 (B7-2) co-stimulatory ligands. Both compounds preferentially bind CD80, yet CD86 has been implicated as the dominant co-stimulatory ligand. Using directed evolution methods, novel CTLA4-Ig variants were created with selective CD86 binding affinity, a property that confers increased immunosuppressive potency and potentially improved efficacy and safety profiles. Relative to abatacept (wild-type CTLA4-Ig), ASP2408 and ASP2409 have 83-fold and 220-fold enhanced binding affinity to CD86 while retaining 1.5-fold and 5.6-fold enhanced binding affinity to CD80, respectively. Improvements in CD86 binding affinity correlates with increased immunosuppressive potencyin vitroandin vivo Our results highlight the power of directed evolution methods to obtain non-intuitive protein engineering solutions and represent the first examples of CD86-selective CTLA4-Ig compounds that have entered clinical trials.


Assuntos
Abatacepte/genética , Abatacepte/farmacologia , Antígeno B7-2/metabolismo , Evolução Molecular Direcionada , Imunoconjugados/metabolismo , Imunoconjugados/farmacologia , Imunossupressores/metabolismo , Imunossupressores/farmacologia , Abatacepte/química , Abatacepte/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Feminino , Humanos , Imunoconjugados/química , Imunossupressores/química , Ligantes , Camundongos , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
7.
Antimicrob Agents Chemother ; 60(1): 316-22, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26503655

RESUMO

Tenofovir alafenamide fumarate (TAF) is an oral phosphonoamidate prodrug of the HIV reverse transcriptase nucleotide inhibitor tenofovir (TFV). Previous studies suggested a principal role for the lysosomal serine protease cathepsin A (CatA) in the intracellular activation of TAF. Here we further investigated the role of CatA and other human hydrolases in the metabolism of TAF. Overexpression of CatA or liver carboxylesterase 1 (Ces1) in HEK293T cells increased intracellular TAF hydrolysis 2- and 5-fold, respectively. Knockdown of CatA expression with RNA interference (RNAi) in HeLa cells reduced intracellular TAF metabolism 5-fold. Additionally, the anti-HIV activity and the rate of CatA hydrolysis showed good correlation within a large set of TFV phosphonoamidate prodrugs. The covalent hepatitis C virus (HCV) protease inhibitors (PIs) telaprevir and boceprevir potently inhibited CatA-mediated TAF activation (50% inhibitory concentration [IC50] = 0.27 and 0.16 µM, respectively) in vitro and also reduced its anti-HIV activity in primary human CD4(+) T lymphocytes (21- and 3-fold, respectively) at pharmacologically relevant concentrations. In contrast, there was no inhibition of CatA or any significant effect on anti-HIV activity of TAF observed with cobicistat, noncovalent HIV and HCV PIs, or various prescribed inhibitors of host serine proteases. Collectively, these studies confirm that CatA plays a pivotal role in the intracellular metabolism of TAF, whereas the liver esterase Ces1 likely contributes to the hepatic activation of TAF. Moreover, this work demonstrates that a wide range of viral and host PIs, with the exception of telaprevir and boceprevir, do not interfere with the antiretroviral activity of TAF.


Assuntos
Adenina/análogos & derivados , Fármacos Anti-HIV/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Pró-Fármacos/metabolismo , Tenofovir/metabolismo , Adenina/metabolismo , Adenina/farmacologia , Alanina , Fármacos Anti-HIV/farmacologia , Biotransformação , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/virologia , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Catepsina A/antagonistas & inibidores , Catepsina A/genética , Catepsina A/metabolismo , Cobicistat/farmacologia , Interações Medicamentosas , Expressão Gênica , Células HEK293 , HIV-1/efeitos dos fármacos , HIV-1/crescimento & desenvolvimento , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Oligopeptídeos/farmacologia , Cultura Primária de Células , Pró-Fármacos/farmacologia , Prolina/análogos & derivados , Prolina/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Inibidores de Serina Proteinase/farmacologia , Tenofovir/farmacologia
8.
Antivir Ther ; 19(7): 687-92, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24699134

RESUMO

BACKGROUND: Tenofovir alafenamide (TAF) is a novel prodrug of tenofovir that shows enhanced antiretroviral effect and reduced plasma tenofovir exposures at approximately one-tenth the clinically approved dose of tenofovir disoproxil fumarate (TDF). Tenofovir released from TDF undergoes active renal secretion via organic anion transporters (OAT1 and OAT3), leading to higher exposure of renal proximal tubules to tenofovir and a potential for renal adverse effects in a small subset of TDF-treated patients. Here, we evaluate the interaction of TAF with OAT1 and OAT3 to assess the potential for its active accumulation in proximal tubules. METHODS: OAT-mediated transport and cytotoxicity (CC50) of TAF and tenofovir were assessed in cells expressing OATs and compared with matched transporter-null cells. RESULTS: While OAT1 and OAT3 expression increased tenofovir cellular uptake by >70-fold and 8.2-fold, respectively, the expression of either OAT did not significantly change TAF intracellular accumulation under identical conditions. In addition, although tenofovir was significantly more cytotoxic in OAT1- and OAT3-expressing cells (>21 and >3.6 fold change in CC50 values, respectively), TAF in vitro cytotoxicity showed little to no change upon overexpression of either renal transporter (0.5-3.5 fold change in CC50). CONCLUSIONS: Unlike tenofovir, TAF does not interact with renal transporters OAT1 or OAT3 and exhibits no OAT-dependent cytotoxicity. TAF is thus unlikely to actively accumulate in renal proximal tubules in an OAT-dependent manner, supporting the potential for an improved renal safety profile.


Assuntos
Adenina/análogos & derivados , Túbulos Renais Proximais/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Adenina/química , Adenina/metabolismo , Adenina/farmacologia , Alanina , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Transporte Biológico , Linhagem Celular , Células Cultivadas , Expressão Gênica , Humanos , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Tenofovir/análogos & derivados
9.
Antivir Ther ; 19(7): 669-77, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24625459

RESUMO

BACKGROUND: Tenofovir alafenamide (TAF) is a novel investigational prodrug of tenofovir (TFV) that permits enhanced delivery of TFV into peripheral blood mononuclear cells (PBMCs) and lymphatic tissues. A critical step in the intracellular metabolic activation of TAF is mediated by the lysosomal protease cathepsin A (CatA). Here, we investigated CatA levels together with intracellular metabolism and antiretroviral activity of TAF in primary CD4+ T-lymphocytes (CD4s) and monocyte-derived macrophages (MDMs) isolated from a demographically diverse group of blood donors. METHODS: CD4s and MDMs were prepared from fresh PBMCs. CatA levels were quantified in cell extracts by monitoring TAF hydrolysis using HPLC. Intracellular TAF metabolites were quantified by HPLC combined with mass spectrometry. Antiviral activities in activated CD4s and MDMs were determined using HIV-1 single-cycle reporter and p24 antigen production assays, respectively. RESULTS: The levels of CatA and intracellular TAF metabolites differed minimally in CD4s and MDMs among 13 tested donors. TAF was >600-fold and 80-fold more potent than parent TFV in CD4s and MDMs, respectively, and its relative range of antiviral activity across all tested donors was comparable to that of other HIV-1 reverse transcriptase inhibitors, with mean ±sd (range) EC50 values of 11.0 ±3.4 (6.6-19.9) nM and 9.7 ±4.6 (2.5-15.7) nM in CD4s and MDMs, respectively. CONCLUSIONS: These results indicate consistent intracellular metabolism and antiretroviral potency of TAF in relevant target cells of HIV-1 infection across multiple donors of variable gender, age and ethnicity, supporting further clinical investigation of TAF.


Assuntos
Adenina/análogos & derivados , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Adenina/química , Adenina/metabolismo , Adenina/farmacologia , Alanina , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Catepsina A/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Tenofovir/análogos & derivados , Doadores de Tecidos
10.
PLoS One ; 8(9): e74163, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24040198

RESUMO

HIV-1 integrase (IN) is the target for two classes of antiretrovirals: i) the integrase strand-transfer inhibitors (INSTIs) and ii) the non-catalytic site integrase inhibitors (NCINIs). NCINIs bind at the IN dimer interface and are thought to interfere primarily with viral DNA (vDNA) integration in the target cell by blocking IN-vDNA assembly as well as the IN-LEDGF/p75 interaction. Herein we show that treatment of virus-producing cells, but not of mature virions or target cells, drives NCINI antiviral potency. NCINIs target an essential late-stage event in HIV replication that is insensitive to LEDGF levels in the producer cells. Virus particles produced in the presence of NCINIs displayed normal Gag-Pol processing and endogenous reverse transcriptase activity, but were defective at initiating vDNA synthesis following entry into the target cell. NCINI-resistant virus carrying a T174I mutation in the IN dimer interface was less sensitive to the compound-induced late-stage effects, including the reverse transcription block. Wild-type, but not T174I virus, produced in the presence of NCINIs exhibited striking defects in core morphology and an increased level of IN oligomers that was not observed upon treatment of mature cell-free particles. Collectively, these results reveal that NCINIs act through a novel mechanism that is unrelated to the previously observed inhibition of IN activity or IN-LEDGF interaction, and instead involves the disruption of an IN function during HIV-1 core maturation and assembly.


Assuntos
DNA Viral/antagonistas & inibidores , Inibidores de Integrase de HIV/farmacologia , Integrase de HIV/genética , Transcriptase Reversa do HIV/antagonistas & inibidores , HIV-1/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular Tumoral , DNA Viral/genética , Farmacorresistência Viral , Expressão Gênica , Genes Reporter , Vetores Genéticos , Integrase de HIV/metabolismo , Inibidores de Integrase de HIV/química , Transcriptase Reversa do HIV/genética , Transcriptase Reversa do HIV/metabolismo , HIV-1/genética , HIV-1/crescimento & desenvolvimento , HIV-1/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Luciferases/genética , Luciferases/metabolismo , Mutação , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Linfócitos T/virologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vírion/efeitos dos fármacos , Vírion/genética , Montagem de Vírus/efeitos dos fármacos , Integração Viral/efeitos dos fármacos
11.
Antimicrob Agents Chemother ; 57(10): 4982-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23896476

RESUMO

A once-daily single-tablet antiretroviral regimen containing tenofovir (TFV) disoproxil fumarate, emtricitabine (FTC), elvitegravir (EVG), and cobicistat (COBI) is an approved combination for the treatment of patients infected with HIV. COBI and TFV have been reported to interact with distinct transporters in renal proximal tubules; while TFV is renally eliminated by a combination of glomerular filtration and tubular secretion via anion transporters OAT1, OAT3, and MRP4, COBI inhibits renal cation transporters, particularly MATE1, resulting in a measurable decrease in the tubular secretion of creatinine. To investigate the potential for a renal drug-drug interaction between TFV and COBI in vitro, the uptake of TFV in the presence and absence of COBI was determined in fresh human renal cortex tissue and in cells expressing the relevant renal transporters. At concentrations exceeding clinical protein-unbound plasma levels, COBI did not significantly inhibit the transport of TFV by the anion transporters OAT1, OAT3, and MRP4 (50% inhibitory concentrations [IC50s] of >15, 6.6, and 8.5 µM, respectively). Conversely, TFV had little or no effect on the cation transporters OCT2 and MATE1 (IC50 > 100 µM). Consistent with studies using individual transporters, no increase in the accumulation of TFV in freshly isolated human renal cortex tissue or renal proximal tubule cells (RPTECs) was observed in the presence of COBI. Finally, COBI alone or in combination with FTC and EVG did not affect the sensitivity to TFV of cultured primary RPTECs or cells coexpressing OAT1 and MRP4. These results illustrate that COBI and TFV interact primarily with distinct renal transporters and indicate a low potential for pharmacokinetic renal drug-drug interaction.


Assuntos
Adenina/análogos & derivados , Fármacos Anti-HIV/farmacologia , Carbamatos/farmacologia , Organofosfonatos/farmacologia , Tiazóis/farmacologia , Adenina/farmacologia , Adulto , Linhagem Celular , Cobicistat , Interações Medicamentosas , Humanos , Técnicas In Vitro , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico , Tenofovir
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...