Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
J Med Chem ; 64(3): 1481-1496, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33497209

RESUMO

CD4 mimics are small molecules that inhibit the interaction of gp120 with CD4. We have developed several CD4 mimics. Herein, hybrid molecules consisting of CD4 mimics with a long alkyl chain or a PEG unit attached through a self-cleavable linker were synthesized. In anti-HIV activity, modification with a PEG unit appeared to be more suitable than modification with a long alkyl chain. Thus, hybrid molecules of CD4 mimics, with PEG units attached through an uncleavable linker, were developed and showed high anti-HIV activity and low cytotoxicity. In investigation of pharmacokinetics in a rhesus macaque, a hybrid compound had a more effective PK profile than that of the parent compound, and intramuscular injection was a more useful administration route to maintain the high blood concentration of the CD4 mimic than intravenous injection. The presented hybrid molecules of CD4 mimics with a PEG unit would be practically useful when combined with a neutralizing antibody.


Assuntos
Antígenos CD4/química , Inibidores da Fusão de HIV/síntese química , Inibidores da Fusão de HIV/farmacologia , Polietilenoglicóis/química , Animais , Anticorpos Neutralizantes/química , Proteína gp120 do Envelope de HIV/efeitos dos fármacos , Inibidores da Fusão de HIV/farmacocinética , HIV-1/efeitos dos fármacos , Humanos , Injeções Intramusculares , Macaca mulatta , Modelos Moleculares , Mimetismo Molecular , Relação Estrutura-Atividade
2.
Viruses ; 14(1)2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-35062273

RESUMO

We report the discovery of several highly potent small molecules with low-nM potency against severe acute respiratory syndrome coronavirus (SARS-CoV; lowest half-maximal inhibitory concentration (IC50: 13 nM), SARS-CoV-2 (IC50: 23 nM), and Middle East respiratory syndrome coronavirus (MERS-CoV; IC50: 76 nM) in pseudovirus-based assays with excellent selectivity index (SI) values (>5000), demonstrating potential pan-coronavirus inhibitory activities. Some compounds showed 100% inhibition against the cytopathic effects (CPE; IC100) of an authentic SARS-CoV-2 (US_WA-1/2020) variant at 1.25 µM. The most active inhibitors also potently inhibited variants of concern (VOCs), including the UK (B.1.1.7) and South African (B.1.351) variants and the Delta variant (B.1.617.2) originally identified in India in pseudovirus-based assay. Surface plasmon resonance (SPR) analysis with one potent inhibitor confirmed that it binds to the prefusion SARS-CoV-2 spike protein trimer. These small-molecule inhibitors prevented virus-mediated cell-cell fusion. The absorption, distribution, metabolism, and excretion (ADME) data for one of the most active inhibitors, NBCoV1, demonstrated drug-like properties. An in vivo pharmacokinetics (PK) study of NBCoV1 in rats demonstrated an excellent half-life (t1/2) of 11.3 h, a mean resident time (MRT) of 14.2 h, and oral bioavailability. We expect these lead inhibitors to facilitate the further development of preclinical and clinical candidates.


Assuntos
Antivirais/farmacologia , SARS-CoV-2/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/farmacocinética , Disponibilidade Biológica , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Coronavirus/classificação , Coronavirus/efeitos dos fármacos , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/farmacologia , Humanos , Ligação Proteica , Ratos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacocinética , Bibliotecas de Moléculas Pequenas/farmacologia , Glicoproteína da Espícula de Coronavírus/antagonistas & inibidores
3.
J Virol ; 94(14)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32404523

RESUMO

Anti-human immunodeficiency virus type 1 (anti-HIV-1) fusion peptides have been studied for nearly 2 decades, but few candidates have found useful clinical applications. One factor underlying the failure of such agents to reach the clinic is their poor pharmacokinetic properties, and many efforts have been made to overcome this problem. In this study, we modified C34, a peptide inhibitor of HIV-1 fusion, at its conserved glycosylation site using polyethylene glycols (PEGs) of different molecular weights. PEG40-NC, a conjugate of C34 and branched PEG 40 kDa (PEG40), which has been previously shown to improve the pharmacokinetic profiles of proteins, showed a significantly extended half-life (t1/2; 10.39 h in rats), which compensated for decreased in vitro activity (50% effective concentration [EC50] of 18.51 nM). PEG40-NC also showed a mechanism of action similar to that of C34. PEG40-NC monotherapy in acutely simian-human immunodeficiency virus (SHIV)-infected rhesus monkeys significantly suppressed viral load compared with a control treatment. Efficacy was linked to the extended half-life and lymphatic exposure conferred by attached PEG40. These results highlight the potential of further clinical investigations of PEG40-NC in combination with antiretroviral therapy or other anti-HIV agents.IMPORTANCE Poor pharmacokinetics have severely hindered the clinical use of anti-HIV peptides. Different small molecules, such as lipid, cholesterol, and small PEG, were designed to modify peptides to improve their pharmacokinetics. In this study, we incorporated large branched PEG to anti-HIV peptide and obtained a conjugate with extended half-life and improved in vivo efficacy. The strategy we developed in this study can also be applicable for the development of other peptide candidates.


Assuntos
Proteína gp41 do Envelope de HIV , Inibidores da Fusão de HIV , Infecções por HIV , HIV-1/metabolismo , Fragmentos de Peptídeos , Polietilenoglicóis/química , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia/metabolismo , Animais , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/farmacocinética , Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/farmacologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Humanos , Macaca mulatta , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacocinética , Fragmentos de Peptídeos/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/patologia
4.
PLoS One ; 14(10): e0223969, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31647836

RESUMO

Effects of steady-state rifabutin on the pharmacokinetics of steady-state maraviroc were investigated in fourteen healthy adult female and male volunteers. Maraviroc 300 mg twice daily (BID) was given orally with food for fifteen days. On day six, rifabutin 300 mg once daily (QD, P.O.) was added to the regimen. Formal pharmacokinetic (PK) sampling was performed on days five and fifteen. Individual plasma drug concentration-time data for maraviroc, and rifabutin on day fifteen, were obtained using validated High Performance Liquid Chromatography (HPLC) tandem Mass Spectrometry (MS/MS). Rifabutin steady state exposure was comparable to data in the literature. Maraviroc area under the curve (AUC) and minimum plasma concentration (Clast or Cmin) were reduced by 17% and 30% respectively when co-administered with rifabutin. No unexpected or serious adverse eventsoccurred. Based on the reduced exposure of maraviroc observed in this study, increasing the dose of maraviroc may be studied to normalize its moderately reduced exposure following rifabutin co-administration, a moderate inducer of CYP3A4.


Assuntos
Antibacterianos/farmacocinética , Interações Medicamentosas , Inibidores da Fusão de HIV/farmacocinética , Maraviroc/farmacocinética , Rifabutina/farmacocinética , Adolescente , Adulto , Idoso , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Distribuição Tecidual , Adulto Jovem
5.
Viruses ; 11(9)2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31480738

RESUMO

The clinical application of HIV fusion inhibitor, enfuvirtide (T20), was limited mainly because of its short half-life. Here we designed and synthesized two PEGylated C34 peptides, PEG2kC34 and PEG5kC34, with the PEG chain length of 2 and 5 kDa, respectively, and evaluated their anti-HIV-1 activity and mechanisms of action. We found that these two PEGylated peptides could bind to the HIV-1 peptide N36 to form high affinity complexes with high α-helicity. The peptides PEG2kC34 and PEG5kC34 effectively inhibited HIV-1 Env-mediated cell-cell fusion with an effective concentration for 50% inhibition (EC50) of about 36 nM. They also inhibited infection of the laboratory-adapted HIV-1 strain NL4-3 with EC50 of about 4-5 nM, and against 47 HIV-1 clinical isolates circulating in China with mean EC50 of PEG2kC34 and PEG5kC34 of about 26 nM and 32 nM, respectively. The plasma half-life (t1/2) of PEG2kC34 and PEG5kC34 was 2.6 h and 5.1 h, respectively, and the t1/2 of PEGylated C34 was about 2.4-fold and 4.6-fold longer than C34 (~1.1 h), respectively. These findings suggest that PEGylated C34 with broad-spectrum anti-HIV-1 activity and prolonged half-life can be further developed as a peptide fusion inhibitor-based long-acting anti-HIV drug for clinical use to treat HIV-infected patients who have failed to respond to current anti-retrovirus drugs.


Assuntos
Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular , Dicroísmo Circular , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Enfuvirtida/farmacologia , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV/síntese química , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , HIV-1/genética , Humanos , Fusão de Membrana/efeitos dos fármacos , Mutação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Polietilenoglicóis/química , Ligação Proteica , Conformação Proteica em alfa-Hélice , Ratos , Replicação Viral/efeitos dos fármacos
6.
Eur J Pharm Biopharm ; 137: 218-226, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30851352

RESUMO

HIV therapy with anti-retroviral drugs is limited by the poor exposure of viral reservoirs, such as lymphoid tissue, to these small molecule drugs. We therefore investigated the effect of PEGylation on the anti-retroviral activity and subcutaneous lymphatic pharmacokinetics of the peptide-based fusion inhibitor enfuvirtide in thoracic lymph duct cannulated rats. Both the peptide and the PEG were quantified in plasma and lymph via ELISA. Conjugation to a single 5 kDa linear PEG decreased anti-HIV activity three-fold compared to enfuvirtide. Whilst plasma and lymphatic exposure to peptide mass was moderately increased, the loss of anti-viral activity led to an overall decrease in exposure to enfuvirtide activity. A 20 kDa 4-arm branched PEG conjugated with an average of two enfuvirtide peptides decreased peptide activity by six-fold. Plasma and lymph exposure to enfuvirtide, however, increased significantly such that anti-viral activity was increased two- and six-fold respectively. The results suggest that a multi-enfuvirtide-PEG complex may optimally enhance the anti-retroviral activity of the peptide in plasma and lymph.


Assuntos
Enfuvirtida/administração & dosagem , Inibidores da Fusão de HIV/administração & dosagem , HIV/efeitos dos fármacos , Polietilenoglicóis/química , Animais , Linhagem Celular , Enfuvirtida/farmacocinética , Enfuvirtida/farmacologia , Ensaio de Imunoadsorção Enzimática , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/farmacologia , Infecções por HIV/tratamento farmacológico , Humanos , Linfa/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
8.
J Clin Pharmacol ; 59(1): 139-152, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30192390

RESUMO

Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. Previous studies show that cytochrome P450 3A5 (CYP3A5) plays a role in maraviroc metabolism. CYP3A5 is subject to a genetic polymorphism. The presence of 2 functional alleles (CYP3A5*1/*1) confers the extensive metabolism phenotype, which is rare in whites but common in blacks. The effect of CYP3A5 genotype on maraviroc and/or metabolite pharmacokinetics was evaluated in 2 clinical studies: a post hoc analysis from a phase 2b/3 study (NCT00098293) conducted in 494 HIV-1-infected subjects (study 1) in which the impact on maraviroc efficacy in 303 subjects was also assessed, and a study conducted in 47 healthy volunteers (study 2). In study 2 (NCT02625207), extensive metabolizers had 26% to 37% lower mean area under the concentration-time curve compared with poor metabolizers (no CYP3A5*1 alleles). This effect diminished to 17% in the presence of potent CYP3A inhibition. The effect of CYP3A5 genotype was greatest in the formation of the metabolite (1S,2S)-2-hydroxymaraviroc. In study 1, the CYP3A5*1/*1 genotype unexpectedly had higher maraviroc area under the curve predictions (20%) compared with those with no CYP3A5*1 alleles. The reason for this disparity remains unclear. The proportions of subjects with viral loads <50 and <400 copies/mL for maraviroc were comparable among all 3 CYP3A5 genotypes. In both studies maraviroc exposures were in the range of near-maximal viral inhibition in the majority of subjects. These results demonstrate that although CYP3A5 contributes to the metabolism of maraviroc, CYP3A5 genotype does not affect the clinical response to maraviroc in combination treatment of HIV-1 infection at approved doses.


Assuntos
Citocromo P-450 CYP3A/genética , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV , HIV-1 , Maraviroc/farmacocinética , Maraviroc/uso terapêutico , Adulto , Método Duplo-Cego , Feminino , Genótipo , Inibidores da Fusão de HIV/sangue , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Infecções por HIV/metabolismo , Voluntários Saudáveis , Humanos , Masculino , Maraviroc/sangue , Pessoa de Meia-Idade , Polimorfismo Genético , Resultado do Tratamento , Adulto Jovem
9.
Eur J Pharm Sci ; 125: 244-253, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30292749

RESUMO

DNA helix-based HIV-1 fusion inhibitors have been discovered as potent drug candidates, but further research is required to enhance their efficiency. The trimeric structure of the HIV-1 envelope glycoprotein provides a structural basis for multivalent drug design. In this work, a "multi-domain" strategy was adopted for design of an oligodeoxynucleotide with assembly, linkage, and activity domains. Built on the self-assembly of higher-order nucleic acid structure, a novel category of multivalent DNA helix-based HIV-1 fusion inhibitor could be easily obtained by a simple annealing course in solution buffer, with no other chemical synthesis for multivalent connection. An optimized multivalent molecule, M4, showed significantly higher anti-HIV-1 fusion activity than did corresponding monovalent inhibitors. Examination of the underlying mechanism indicated that M4 could interact with HIV-1 glycoproteins gp120 and gp41, thereby inhibiting 6HB formation in the fusion course. M4 also showed anti-RDDP and anti-RNase H activity of reverse transcriptase. Besides, these assembled molecules showed improved in vitro metabolic stability in liver homogenate, kidney homogenate, and rat plasma. Moreover, little acute toxicity was observed. Our findings aid in the structural design and understanding of the mechanisms of DNA helix-based HIV-1 inhibitors. This study also provides a general strategy based on a new structural paradigm for the design of other multivalent nucleic acid drugs.


Assuntos
Inibidores da Fusão de HIV/administração & dosagem , HIV-1/efeitos dos fármacos , Oligodesoxirribonucleotídeos/administração & dosagem , Animais , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/toxicidade , HIV-1/fisiologia , Rim/metabolismo , Fígado/metabolismo , Masculino , Camundongos Endogâmicos ICR , Oligodesoxirribonucleotídeos/farmacocinética , Oligodesoxirribonucleotídeos/toxicidade , Ratos Sprague-Dawley , Internalização do Vírus/efeitos dos fármacos
10.
Mol Pharm ; 15(3): 1169-1179, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29436835

RESUMO

Peptides often suffer from short in vivo half-lives due to proteolysis and renal clearance that limit their therapeutic potential in many indications, necessitating pharmacokinetic (PK) enhancement. d-Peptides, composed of mirror-image d-amino acids, overcome proteolytic degradation but are still vulnerable to renal filtration due to their small size. If renal filtration could be slowed, d-peptides would be promising therapeutic agents for infrequent dosing, such as in extended-release depots. Here, we tether a diverse set of PK-enhancing cargoes to our potent, protease-resistant d-peptide HIV entry inhibitor, PIE12-trimer. This inhibitor panel provides an opportunity to evaluate the PK impact of the cargoes independently of proteolysis. While all the PK-enhancing strategies (PEGylation, acylation, alkylation, and cholesterol conjugation) improved in vivo half-life, cholesterol conjugation of PIE12-trimer dramatically improves both antiviral potency and half-life in rats, making it our lead anti-HIV drug candidate. We designed its chemical synthesis for large-scale production (CPT31) and demonstrated that the PK profile in cynomolgous monkeys supports future development of monthly or less frequent depot dosing in humans. CPT31 could address an urgent need in both HIV prevention and treatment.


Assuntos
Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Fragmentos de Peptídeos/farmacocinética , Animais , Colesterol/química , Preparações de Ação Retardada , Portadores de Fármacos/química , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores da Fusão de HIV/administração & dosagem , Inibidores da Fusão de HIV/síntese química , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , HIV-1/fisiologia , Meia-Vida , Macaca fascicularis , Masculino , Modelos Animais , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/síntese química , Ratos , Ratos Sprague-Dawley , Estereoisomerismo
11.
Pediatr Infect Dis J ; 37(5): 459-465, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29023357

RESUMO

BACKGROUND: Maraviroc is a CC-chemokine receptor 5 antagonist approved to treat adults infected with CC-chemokine receptor 5-tropic (R5) HIV-1. Study A4001031 was conducted to evaluate the pharmacokinetics, safety and efficacy of maraviroc in combination with optimized background therapy in treatment-experienced pediatric patients infected with R5 HIV-1 and support registration of maraviroc for pediatric use. METHODS: This is an open-label, 2-stage, age-stratified, noncomparative multicenter study. One-hundred and three participants were enrolled into 4 age/formulation cohorts and dosed twice daily. Initial doses were determined by body surface area and optimized background therapy, based on drug interactions with maraviroc in adults. Dose adjustment and pharmacokinetic reevaluation occurred if the average concentrations (Cavg) at Week 2 were <100 ng/mL (Stage 1-dose finding). RESULTS: Data from the Week 48 analysis demonstrated that 49/50 Stage 1 participants rolling over into Stage 2 (safety and efficacy) achieved Cavg ≥100 ng/mL. Doses were identified that achieved similar concentration ranges to those seen in adults. The majority (90/103) received optimized background therapy containing potent cytochrome P450 3A inhibitors. Maraviroc was well tolerated and the safety and efficacy were comparable to those of adults. All cohorts had a mean decrease from baseline in HIV-1 RNA of >1 log10. Increases from baseline in the median CD4+ cell count and percentage were seen for all age groups. CONCLUSIONS: The maraviroc dosing strategy resulted in participants achieving the target Cavg, with exposure ranges similar to those observed in adults on approved doses. The safety and efficacy of maraviroc in this pediatric population were comparable to those seen in adults.


Assuntos
Antagonistas dos Receptores CCR5/farmacocinética , Antagonistas dos Receptores CCR5/uso terapêutico , Inibidores da Fusão de HIV/farmacocinética , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Maraviroc/farmacocinética , Maraviroc/uso terapêutico , Adolescente , Antagonistas dos Receptores CCR5/efeitos adversos , Contagem de Linfócito CD4 , Criança , Pré-Escolar , Feminino , Inibidores da Fusão de HIV/efeitos adversos , HIV-1/efeitos dos fármacos , Humanos , Masculino , Maraviroc/efeitos adversos , Receptores CCR5 , Carga Viral/efeitos dos fármacos , Tropismo Viral
12.
J Acquir Immune Defic Syndr ; 73(3): 252-257, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27727157

RESUMO

To investigate the pharmacokinetics/pharmacodynamics of single-dose maraviroc 300 mg in HIV-1 exposure compartments. Maraviroc concentrations in blood, secretions (vaginal, urethral, oral, and rectal), and tissue (vaginal and rectal) were measured, and ex vivo challenge was performed in 54 healthy volunteers to study protection from HIV infection. Maraviroc Cmax occurred within 4 hours in most compartments. Concentrations from 4 to 72 hours were above intracellular (IC) IC90 in all compartments, range 15-8095 ng/mL. Mean AUC0-72 compartment-to-plasma ratios were highest in the rectum (45-819) and urethra (144) compared with the female genital tract (1.6-4.8) and saliva (0.2). No sex differences in AUC0-72 or Cmax were observed. No ex vivo protection from HIV-1BaL occurred in rectal or vaginal tissue. Despite high and sustained concentrations, single-dose maraviroc was not protective against ex vivo challenge of vaginal/rectal tissue.


Assuntos
Antagonistas dos Receptores CCR5/farmacocinética , Cicloexanos/farmacocinética , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , Profilaxia Pré-Exposição , Triazóis/farmacocinética , Administração Oral , Adulto , Antagonistas dos Receptores CCR5/administração & dosagem , Antagonistas dos Receptores CCR5/farmacologia , Cicloexanos/administração & dosagem , Cicloexanos/farmacologia , Feminino , Inibidores da Fusão de HIV/administração & dosagem , Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/virologia , Masculino , Maraviroc , Modelos Biológicos , Ensaios Clínicos Controlados Aleatórios como Assunto , Reto/efeitos dos fármacos , Reto/virologia , Saliva/efeitos dos fármacos , Saliva/virologia , Resultado do Tratamento , Triazóis/administração & dosagem , Triazóis/farmacologia , Uretra/efeitos dos fármacos , Uretra/virologia , Vagina/efeitos dos fármacos , Vagina/virologia
13.
Medicine (Baltimore) ; 95(28): e4174, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27428211

RESUMO

BACKGROUND: The ex vivo challenge assay is a bio-indicator of drug efficacy and was utilized in this randomized, placebo controlled trial as one of the exploratory endpoints. Fresh and cryopreserved tissues were evaluated for human immunodeficiency virus (HIV) infection and pharmacokinetic (PK)/pharmacodynamic (PD) relationships. METHODS: HIV-negative women used vaginal rings containing 25 mg dapivirine (DPV)/100 mg maraviroc (MVC) (n = 12), DPV only (n = 12), MVC only (n = 12), or placebo (n = 12) for 28 days. Blood plasma, cervicovaginal fluid (CVF), and cervical biopsies were collected for drug quantification and the ex vivo challenge assay; half (fresh) were exposed immediately to HIV while the other half were cryopreserved, thawed, then exposed to HIV. HIV replication was monitored by p24 enzyme-linked immunosorbent assay from culture supernatant. Data were log-transformed and analyzed by linear least squared regression, nonlinear Emax dose-response model and Satterthwaite t test. RESULTS: HIV replication was greater in fresh compared to cryopreserved tissue (P = 0.04). DPV was detected in all compartments, while MVC was consistently detected only in CVF. Significant negative correlations between p24 and DPV levels were observed in fresh cervical tissue (P = 0.01) and CVF (P = 0.03), but not plasma. CVF MVC levels showed a significant negative correlation with p24 levels (P = 0.03); drug levels in plasma and tissue were not correlated with HIV suppression. p24 levels from cryopreserved tissue did not correlate to either drug from any compartment. CONCLUSION: Fresh tissue replicated HIV to greater levels and defined PK/PD relationships while cryopreserved tissue did not. The ex vivo challenge assay using fresh tissue could prioritize drugs being considered for HIV prevention.


Assuntos
Dispositivos Anticoncepcionais Femininos , Cicloexanos/farmacologia , Inibidores da Fusão de HIV/farmacologia , Infecções por HIV/prevenção & controle , HIV-1/efeitos dos fármacos , Pirimidinas/farmacologia , Doenças Virais Sexualmente Transmissíveis/prevenção & controle , Triazóis/farmacologia , Administração Intravaginal , Adulto , Biópsia , Colo do Útero/virologia , Criopreservação , Cicloexanos/farmacocinética , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , Feminino , Inibidores da Fusão de HIV/farmacocinética , Humanos , Técnicas In Vitro , Maraviroc , Pirimidinas/farmacocinética , Triazóis/farmacocinética , Estados Unidos
14.
Eur J Med Chem ; 121: 232-237, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27240277

RESUMO

Enfuvirtide (ENF) is a clinically used peptide drug for the treatment of HIV infections, but its poor pharmacokinetic profile (T1/2 = 1.5 h in rats) and low aqueous solubility make the therapy expensive and inconvenience. In this study, we present a simple and practical strategy to address these problems by conjugating ENF with polyethylene glycol (PEG). Site-specific attachment of a 2 kDa PEG at the N-terminus of ENF resulted in an ENF-PEG (EP) conjugate with high solubility (≥3 mg/mL) and long half-life in rats (T1/2 = 16.1 h). This conjugate showed similar antiviral activity to ENF against various primary HIV-1 isolates (EC50 = 6-91 nM). Mechanistic studies suggested the sources of the antiviral potency. The conjugate bound to a functional domain of the HIV gp41 protein in a helical conformation with high affinity (Kd = 307 nM), thereby inhibiting the gp41-mediated fusion of viral and host-cell membranes. As PEG conjugation has advanced many bioactive proteins and peptides into clinical applications, the EP conjugate described here represents a potential new treatment for HIV infections that may address the unmet medical needs associated with the current ENF therapy.


Assuntos
Proteína gp41 do Envelope de HIV/farmacocinética , Inibidores da Fusão de HIV/farmacocinética , Fragmentos de Peptídeos/farmacocinética , Animais , Antivirais/química , Antivirais/farmacocinética , Enfuvirtida , Proteína gp41 do Envelope de HIV/química , Inibidores da Fusão de HIV/química , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Meia-Vida , Fragmentos de Peptídeos/química , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Ratos , Solubilidade
15.
Bioorg Med Chem Lett ; 26(2): 397-400, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26706175

RESUMO

Several CD4 mimics have been reported as HIV-1 entry inhibitors which can block the interaction between the viral envelope glycoprotein gp120 and the cell surface protein CD4. We previously found a lead compound 2 (YYA-021) with high anti-HIV activity and low cytotoxicity. Pharmacokinetic analysis however showed compound 2 to have wide tissue distribution and relatively high distribution volumes in rats and rhesus macaques. In the present study we searched for more hydrophilic CD4 mimics with a view to reducing tissue distribution. A new compound (5) with a 1,3-benzodioxolyl moiety was found to have relatively high anti-HIV activity and no significant cytotoxicity. Compound 5 is more hydrophilic than compound 2 and the pharmacokinetics of the intravenous administration of compound 5 in a rhesus macaque showed that compound 5 has lower tissue distribution than compound 2, suggesting that compound 5 possesses a better profile.


Assuntos
Antígenos CD4/química , Antígenos CD4/farmacologia , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , Animais , Proteína gp120 do Envelope de HIV/metabolismo , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/tratamento farmacológico , Macaca mulatta , Simulação de Acoplamento Molecular , Ácido Oxâmico/análogos & derivados , Ácido Oxâmico/química , Ácido Oxâmico/farmacocinética , Ácido Oxâmico/farmacologia , Piperidinas/química , Piperidinas/farmacocinética , Piperidinas/farmacologia , Ratos
16.
Drug Des Devel Ther ; 9: 5447-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26491256

RESUMO

The human immunodeficiency virus-1 (HIV-1) enters target cells by binding its envelope glycoprotein gp120 to the CD4 receptor and/or coreceptors such as C-C chemokine receptor type 5 (CCR5; R5) and C-X-C chemokine receptor type 4 (CXCR4; X4), and R5-tropic viruses predominate during the early stages of infection. CCR5 antagonists bind to CCR5 to prevent viral entry. Maraviroc (MVC) is the only CCR5 antagonist currently approved by the United States Food and Drug Administration, the European Commission, Health Canada, and several other countries for the treatment of patients infected with R5-tropic HIV-1. MVC has been shown to be effective at inhibiting HIV-1 entry into cells and is well tolerated. With expanding MVC use by HIV-1-infected humans, different clinical outcomes post-approval have been observed with MVC monotherapy or combination therapy with other antiretroviral drugs, with MVC use in humans infected with dual-R5- and X4-tropic HIV-1, infected with different HIV-1 genotype or infected with HIV-2. This review discuss the role of CCR5 in HIV-1 infection, the development of the CCR5 antagonist MVC, its pharmacokinetics, pharmacodynamics, drug-drug interactions, and the implications of these interactions on treatment outcomes, including viral mutations and drug resistance, and the mechanisms associated with the development of resistance to MVC. This review also discusses available studies investigating the use of MVC in the treatment of other diseases such as cancer, graft-versus-host disease, and inflammatory diseases.


Assuntos
Antagonistas dos Receptores CCR5/uso terapêutico , Cicloexanos/uso terapêutico , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Triazóis/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/uso terapêutico , Antagonistas dos Receptores CCR5/efeitos adversos , Antagonistas dos Receptores CCR5/farmacocinética , Cicloexanos/efeitos adversos , Cicloexanos/farmacocinética , Interações Medicamentosas , Farmacorresistência Viral/genética , Genótipo , Inibidores da Fusão de HIV/efeitos adversos , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/diagnóstico , Infecções por HIV/virologia , HIV-1/genética , HIV-1/metabolismo , Humanos , Maraviroc , Mutação , Fenótipo , Polimedicação , Resultado do Tratamento , Triazóis/efeitos adversos , Triazóis/farmacocinética
17.
Sci Rep ; 5: 13028, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26286358

RESUMO

Enfuvirtide (T20), is the first HIV fusion inhibitor approved for treatment of HIV/AIDS patients who fail to respond to the current antiretroviral drugs. However, its clinical application is limited because of short half-life, drug resistance and cross-reactivity with the preexisting antibodies in HIV-infected patients. Using an artificial peptide strategy, we designed a peptide with non-native protein sequence, AP3, which exhibited potent antiviral activity against a broad spectrum of HIV-1 strains, including those resistant to T20, and had remarkably longer in vivo half-life than T20. While the preexisting antibodies in HIV-infected patients significantly suppressed T20's antiviral activity, these antibodies neither recognized AP3, nor attenuated its anti-HIV-1 activity. Structurally different from T20, AP3 could fold into single-helix and interact with gp41 NHR. The two residues, Met and Thr, at the N-terminus of AP3 form a hook-like structure to stabilize interaction between AP3 and NHR helices. Therefore, AP3 has potential for further development as a new HIV fusion inhibitor with improved antiviral efficacy, resistance profile and pharmacological properties over enfuvirtide. Meanwhile, this study highlighted the advantages of artificially designed peptides, and confirmed that this strategy could be used in developing artificial peptide-based viral fusion inhibitors against HIV and other enveloped viruses.


Assuntos
Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Fenômenos Biofísicos/efeitos dos fármacos , Cristalografia por Raios X , Endopeptidase K/metabolismo , Enfuvirtida , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/metabolismo , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/imunologia , HIV-1/isolamento & purificação , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Estrutura Secundária de Proteína , Proteólise/efeitos dos fármacos , Ratos Sprague-Dawley
18.
Eur J Pharm Biopharm ; 93: 254-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25900863

RESUMO

We have previously developed a linker technology for half-life extension of peptides, proteins and small molecule drugs (1). The linkers undergo ß-elimination reactions with predictable cleavage rates to release the native drug. Here we utilize this technology for half-life extension of the 38 amino acid HIV-1 fusion inhibitor TRI-1144. Conjugation of TRI-1144 to 40 kDa PEG by an appropriate ß-eliminative linker and i.v. administration of the conjugate increased the in vivo half-life of the released peptide from 4 to 34 h in the rat, and the pharmacokinetic parameters were in excellent accord with a one-compartment model. From these data we simulated the pharmacokinetics of the PEG-TRI-1144 conjugate in humans, predicting a t1/2,ß of 70 h for the released peptide, and that a serum concentration of 25 nM could be maintained by weekly doses of 8 µmol of the conjugate. Using a non-circulating carrier (2) similar simulations indicated a t1/2,ß of 150 h for the peptide released from the conjugate and that dosing of only 1.8 µmol/week could maintain serum concentrations of TRI-1144 above 25 nM. Hence, releasable ß-eliminative linkers provide significant half-life extension to TRI-1144 and would be expected to do likewise for related peptides.


Assuntos
Proteína gp41 do Envelope de HIV/farmacocinética , Inibidores da Fusão de HIV/farmacocinética , Fragmentos de Peptídeos/farmacocinética , Polietilenoglicóis/farmacocinética , Pró-Fármacos/farmacocinética , Administração Intravenosa , Animais , Química Farmacêutica , Simulação por Computador , Proteína gp41 do Envelope de HIV/administração & dosagem , Proteína gp41 do Envelope de HIV/sangue , Proteína gp41 do Envelope de HIV/síntese química , Inibidores da Fusão de HIV/administração & dosagem , Inibidores da Fusão de HIV/sangue , Inibidores da Fusão de HIV/síntese química , Meia-Vida , Hidrólise , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/síntese química , Peptídeos , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/síntese química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/síntese química , Ratos Sprague-Dawley , Tecnologia Farmacêutica/métodos
19.
Antimicrob Agents Chemother ; 59(7): 3816-22, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25870057

RESUMO

BMS-663068 is a prodrug of BMS-626529, a first-in-class attachment inhibitor that binds directly to HIV-1 gp120, preventing initial viral attachment and entry into host CD4(+) T cells. This open-label, multiple-dose, four-sequence, crossover study addressed potential two-way drug-drug interactions following coadministration of BMS-663068 (BMS-626529 is a CYP3A4 substrate), atazanavir (ATV), and ritonavir (RTV) (ATV and RTV are CYP3A4 inhibitors). Thirty-six healthy subjects were randomized 1:1:1:1 to receive one of four treatment sequences with three consecutive treatments: BMS-663068 at 600 mg twice daily (BID), BMS-663068 at 600 mg BID plus RTV at 100 mg once daily (QD), ATV at 300 mg QD plus RTV at 100 mg QD (RTV-boosted ATV [ATV/r]), or BMS-663068 at 600 mg BID plus ATV at 300 mg QD plus RTV at 100 mg QD. Compared with the results obtained by administration of BMS-663068 alone, coadministration of BMS-663068 with ATV/r increased the BMS-626529 maximum concentration in plasma (Cmax) and the area under the concentration-time curve in one dosing interval (AUCtau) by 68% and 54%, respectively. Similarly, coadministration of BMS-663068 with RTV increased the BMS-626529 Cmax and AUCtau by 53% and 45%, respectively. Compared with the results obtained by administration of ATV/r alone, ATV and RTV systemic exposures remained similar following coadministration of BMS-663068 with ATV/r. BMS-663068 was generally well tolerated, and there were no adverse events (AEs) leading to discontinuation, serious AEs, or deaths. Moderate increases in BMS-626529 systemic exposure were observed following coadministration of BMS-663068 with ATV/r or RTV. However, the addition of ATV to BMS-663068 plus RTV did not further increase BMS-626529 systemic exposure. ATV and RTV exposures remained similar following coadministration of BMS-663068 with either ATV/r or RTV. BMS-663068 was generally well tolerated alone or in combination with either RTV or ATV/r.


Assuntos
Fármacos Anti-HIV/farmacocinética , Sulfato de Atazanavir/uso terapêutico , Inibidores da Fusão de HIV/uso terapêutico , Piperazinas/uso terapêutico , Ritonavir/uso terapêutico , Triazóis/uso terapêutico , Adolescente , Adulto , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/uso terapêutico , Sulfato de Atazanavir/efeitos adversos , Sulfato de Atazanavir/farmacocinética , Estudos Cross-Over , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Proteína gp120 do Envelope de HIV/metabolismo , Inibidores da Fusão de HIV/efeitos adversos , Inibidores da Fusão de HIV/farmacocinética , HIV-1/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Organofosfatos/uso terapêutico , Piperazinas/efeitos adversos , Piperazinas/farmacocinética , Ritonavir/efeitos adversos , Ritonavir/farmacocinética , Triazóis/efeitos adversos , Triazóis/farmacocinética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...