Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Antiviral Res ; 206: 105399, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36007601

RESUMO

Filoviruses enter cells through macropinocytosis and trafficking into the endosomes in which they bind to the receptor Niemann-Pick C1 protein (NPC1) for membrane fusion and entry into the cytoplasm. The endosomal receptor-binding is critical step for filovirus entry. Designing inhibitors to block receptor binding will prevent viral entry. Using available binding structural information from the co-crystal structures of the viral GP with the receptor NPC1 or with monoclonal antibodies, we have conducted structure-based design of peptide inhibitors to target the receptor binding site (RBS). The designed peptides were tested for their inhibition activity against pseudo-typed or replication-competent viruses in a cell-based assay. The results indicate that these peptides exhibited strong activities against both Ebola and Marburg virus infection. It is expected that these peptides can be further developed for therapeutic use to treat filovirus infection and combat the outbreaks.


Assuntos
Filoviridae , Receptores Virais , Inibidores de Proteínas Virais de Fusão , Sítios de Ligação , Proteínas de Transporte/metabolismo , Linhagem Celular , Ebolavirus/fisiologia , Endossomos/metabolismo , Filoviridae/química , Filoviridae/efeitos dos fármacos , Doença pelo Vírus Ebola , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Glicoproteínas de Membrana/metabolismo , Proteína C1 de Niemann-Pick/metabolismo , Receptores Virais/química , Receptores Virais/metabolismo , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacologia , Internalização do Vírus/efeitos dos fármacos
2.
Viruses ; 14(4)2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35458385

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic caused by infection of SARS-CoV-2 and its variants has posed serious threats to global public health, thus calling for the development of potent and broad-spectrum antivirals. We previously designed and developed a peptide-based pan-coronavirus (CoV) fusion inhibitor, EK1, which is effective against all human CoVs (HCoV) tested by targeting the HCoV S protein HR1 domain. However, its relatively short half-life may limit its clinical use. Therefore, we designed, constructed, and expressed a recombinant protein, FL-EK1, which consists of a modified fibronectin type III domain (FN3) with albumin-binding capacity, a flexible linker, and EK1. As with EK1, we found that FL-EK1 could also effectively inhibit infection of SARS-CoV-2 and its variants, as well as HCoV-OC43. Furthermore, it protected mice from infection by the SARS-CoV-2 Delta variant and HCoV-OC43. Importantly, the half-life of FL-EK1 (30 h) is about 15.7-fold longer than that of EK1 (1.8 h). These results suggest that FL-EK1 is a promising candidate for the development of a pan-CoV fusion inhibitor-based long-acting antiviral drug for preventing and treating infection by current and future SARS-CoV-2 variants, as well as other HCoVs.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Inibidores de Proteínas Virais de Fusão , Animais , Antivirais/química , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Domínio de Fibronectina Tipo III , Meia-Vida , Camundongos , Proteínas Recombinantes de Fusão , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacologia
3.
J Biomol Struct Dyn ; 40(12): 5566-5576, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-33438525

RESUMO

Regarding the urgency of therapeutic measures for coronavirus disease 2019 (COVID-19) pandemic, the use of available drugs with FDA approval is preferred because of the less time and cost required for their development. In silico drug repurposing is an accurate way to speed up the screening of the existing FDA-approved drugs to find a therapeutic option for COVID-19. The similarity in SARS-CoV-2 and HIV-1 fusion mechanism to host cells can be a key point for Inhibit SARS-CoV-2 entry into host cells by HIV fusion inhibitors. Accordingly, in this study, an HIV-1 fusion inhibitor called Enfuvirtide (Enf) was selected. The affinity and essential residues involving in the Enf binding to the S2 protein of SARS-CoV-2, HIV-1 gp41 protein and angiotensin-converting enzyme 2 (ACE-2) as a negative control, was evaluated using molecular docking. Eventually, Enf-S2 and Enf-gp41 protein complexes were simulated by molecular dynamics (MD) in terms of binding affinity and stability. Based on the most important criteria such as docking score, cluster size, energy and dissociation constant, the strongest interaction was observed between Enf with the S2 protein. In addition, MD results confirmed that Enf-S2 protein interaction was remarkably stable and caused the S2 protein residues to undergo the fewest fluctuations. In conclusion, it can be stated that Enf can act as a strong SARS-CoV-2 fusion inhibitor and demonstrates the potential to enter the clinical trial phase of COVID-19. Communicated by Ramaswamy H. Sarma.


Assuntos
Tratamento Farmacológico da COVID-19 , Reposicionamento de Medicamentos , Enfuvirtida , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2 , Reposicionamento de Medicamentos/métodos , Enfuvirtida/farmacologia , HIV-1 , Humanos , Simulação de Acoplamento Molecular , SARS-CoV-2/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão
4.
FEBS Open Bio ; 11(12): 3253-3261, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34710289

RESUMO

A key step in infections by enveloped viruses, such as influenza, is the fusion between the viral envelope and the host cell membrane, which allows the virus to insert its genetic material into the host cell and replicate. The influenza virus fusion process is promoted by hemagglutinin (HA), a glycoprotein that contains three identical monomers composed of two polypeptide chains (HA1 and HA2). Early studies on this protein revealed that HA-mediated fusion involves the insertion of the HA2 N-terminal segment into the host membrane and that this segment, known as the fusion peptide, is a key player in the fusion process. This mini-review highlights the main findings that have been obtained by experimental and computational studies on the HA fusion peptide, which give us a glimpse of its mode of action.


Assuntos
Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Proteínas Virais de Fusão/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Membranas , Modelos Moleculares , Peptídeos , Conformação Proteica , Inibidores de Proteínas Virais de Fusão/farmacologia , Proteínas Virais de Fusão/genética
5.
Viruses ; 13(9)2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34578344

RESUMO

Lassa virus (LASV)-a member of the family Arenaviridae-causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 µM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/efeitos dos fármacos , Arenaviridae/metabolismo , Isoquinolinas/farmacologia , Vírus Lassa/efeitos dos fármacos , Quinazolinas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Humanos , Febre Lassa/tratamento farmacológico , Febre Lassa/virologia , Vírus da Coriomeningite Linfocítica , Receptores Virais , Células Vero , Estomatite Vesicular/virologia , Inibidores de Proteínas Virais de Fusão/farmacologia
6.
Molecules ; 26(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946996

RESUMO

Respiratory syncytial virus (RSV) is a major pathogen that causes severe lower respiratory tract infection in infants, the elderly and the immunocompromised worldwide. At present no approved specific drugs or vaccines are available to treat this pathogen. Recently, several promising candidates targeting RSV entry and multiplication steps are under investigation. However, it is possible to lead to drug resistance under the long-term treatment. Therapeutic combinations constitute an alternative to prevent resistance and reduce antiviral doses. Therefore, we tested in vitro two-drug combinations of fusion inhibitors (GS5806, Ziresovir and BMS433771) and RNA-dependent RNA polymerase complex (RdRp) inhibitors (ALS8176, RSV604, and Cyclopamine). The statistical program MacSynergy II was employed to determine synergism, additivity or antagonism between drugs. From the result, we found that combinations of ALS8176 and Ziresovir or GS5806 exhibit additive effects against RSV in vitro, with interaction volume of 50 µM2% and 31 µM2% at 95% confidence interval, respectively. On the other hand, all combinations between fusion inhibitors showed antagonistic effects against RSV in vitro, with volume of antagonism ranging from -50 µM2 % to -176 µM2 % at 95% confidence interval. Over all, our results suggest the potentially therapeutic combinations in combating RSV in vitro could be considered for further animal and clinical evaluations.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Antivirais/química , Antivirais/uso terapêutico , Descoberta de Drogas/métodos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Quinazolinas/química , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/virologia , Bibliotecas de Moléculas Pequenas , Sulfonas , Tiazepinas/química , Tiazepinas/farmacologia , Tiazepinas/uso terapêutico , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacologia , Inibidores de Proteínas Virais de Fusão/uso terapêutico
7.
Viruses ; 13(4)2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33921515

RESUMO

Identification of Na+/taurocholate co-transporting polypeptide (NTCP) as high-affinity hepatic entry receptor for the Hepatitis B and D viruses (HBV/HDV) opened the field for target-based development of cell-entry inhibitors. However, most of the HBV/HDV entry inhibitors identified so far also interfere with the physiological bile acid transporter function of NTCP. The present study aimed to identify more virus-selective inhibitors of NTCP by screening of 87 propanolamine derivatives from the former development of intestinal bile acid reabsorption inhibitors (BARIs), which interact with the NTCP-homologous intestinal apical sodium-dependent bile acid transporter (ASBT). In NTCP-HEK293 cells, the ability of these compounds to block the HBV/HDV-derived preS1-peptide binding to NTCP (virus receptor function) as well as the taurocholic acid transport via NTCP (bile acid transporter function) were analyzed in parallel. Hits were subsequently validated by performing in vitro HDV infection experiments in NTCP-HepG2 cells. The most potent compounds S985852, A000295231, and S973509 showed in vitro anti-HDV activities with IC50 values of 15, 40, and 70 µM, respectively, while the taurocholic acid uptake inhibition occurred at much higher IC50 values of 24, 780, and 490 µM, respectively. In conclusion, repurposing of compounds from the BARI class as novel HBV/HDV entry inhibitors seems possible and even enables certain virus selectivity based on structure-activity relationships.


Assuntos
Reposicionamento de Medicamentos , Hepatite B , Hepatite D , Vírus Delta da Hepatite/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/administração & dosagem , Internalização do Vírus/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Hepatite B/tratamento farmacológico , Hepatite B/virologia , Vírus da Hepatite B/efeitos dos fármacos , Hepatite D/tratamento farmacológico , Hepatite D/virologia , Humanos , Ácido Taurocólico/antagonistas & inibidores
8.
Science ; 371(6536): 1379-1382, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33597220

RESUMO

Containment of the COVID-19 pandemic requires reducing viral transmission. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is initiated by membrane fusion between the viral and host cell membranes, which is mediated by the viral spike protein. We have designed lipopeptide fusion inhibitors that block this critical first step of infection and, on the basis of in vitro efficacy and in vivo biodistribution, selected a dimeric form for evaluation in an animal model. Daily intranasal administration to ferrets completely prevented SARS-CoV-2 direct-contact transmission during 24-hour cohousing with infected animals, under stringent conditions that resulted in infection of 100% of untreated animals. These lipopeptides are highly stable and thus may readily translate into safe and effective intranasal prophylaxis to reduce transmission of SARS-CoV-2.


Assuntos
COVID-19/transmissão , Lipopeptídeos/administração & dosagem , Fusão de Membrana/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/administração & dosagem , Internalização do Vírus/efeitos dos fármacos , Administração Intranasal , Animais , COVID-19/prevenção & controle , COVID-19/virologia , Chlorocebus aethiops , Modelos Animais de Doenças , Desenho de Fármacos , Furões , Lipopeptídeos/química , Lipopeptídeos/farmacocinética , Lipopeptídeos/farmacologia , Camundongos , Profilaxia Pré-Exposição , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Distribuição Tecidual , Células Vero , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacocinética , Inibidores de Proteínas Virais de Fusão/farmacologia
10.
J Gen Virol ; 102(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33054904

RESUMO

Although enveloped viruses canonically mediate particle entry through virus-cell fusion, certain viruses can spread by cell-cell fusion, brought about by receptor engagement and triggering of membrane-bound, viral-encoded fusion proteins on the surface of cells. The formation of pathogenic syncytia or multinucleated cells is seen in vivo, but their contribution to viral pathogenesis is poorly understood. For the negative-strand paramyxoviruses respiratory syncytial virus (RSV) and Nipah virus (NiV), cell-cell spread is highly efficient because their oligomeric fusion protein complexes are active at neutral pH. The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has also been reported to induce syncytia formation in infected cells, with the spike protein initiating cell-cell fusion. Whilst it is well established that fusion protein-specific antibodies can block particle attachment and/or entry into the cell (canonical virus neutralization), their capacity to inhibit cell-cell fusion and the consequences of this neutralization for the control of infection are not well characterized, in part because of the lack of specific tools to assay and quantify this activity. Using an adapted bimolecular fluorescence complementation assay, based on a split GFP-Renilla luciferase reporter, we have established a micro-fusion inhibition test (mFIT) that allows the identification and quantification of these neutralizing antibodies. This assay has been optimized for high-throughput use and its applicability has been demonstrated by screening monoclonal antibody (mAb)-mediated inhibition of RSV and NiV fusion and, separately, the development of fusion-inhibitory antibodies following NiV vaccine immunization in pigs. In light of the recent emergence of coronavirus disease 2019 (COVID-19), a similar assay was developed for SARS-CoV-2 and used to screen mAbs and convalescent patient plasma for fusion-inhibitory antibodies. Using mFITs to assess antibody responses following natural infection or vaccination is favourable, as this assay can be performed entirely at low biocontainment, without the need for live virus. In addition, the repertoire of antibodies that inhibit cell-cell fusion may be different to those that inhibit particle entry, shedding light on the mechanisms underpinning antibody-mediated neutralization of viral spread.


Assuntos
Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , COVID-19/diagnóstico , Infecções por Henipavirus/diagnóstico , Ensaios de Triagem em Larga Escala , Infecções por Vírus Respiratório Sincicial/diagnóstico , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/metabolismo , COVID-19/imunologia , COVID-19/virologia , Fusão Celular , Convalescença , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Soros Imunes/química , Luciferases/genética , Luciferases/metabolismo , Modelos Moleculares , Vírus Nipah/imunologia , Vírus Nipah/patogenicidade , Conformação Proteica , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Suínos , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/metabolismo , Inibidores de Proteínas Virais de Fusão/farmacologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
11.
J Med Chem ; 63(15): 8043-8045, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32786236

RESUMO

Inhibitors of the respiratory syncytial virus (RSV) fusion protein block entry of the virus into the cell and have shown varying efficacy in a human challenge model of RSV disease. Trials in patient populations are yet to show significant benefits. Jonckers et al. ( J. Med. Chem. 2020, DOI: 10.1021/acs.jmedchem.0c00226) describe the discovery of JNJ-53718678 which can now claim the leading position in clinical evaluation. For RSV inhibitors, the current status of the clinical development of the compound is discussed.


Assuntos
Antivirais/química , Imidazolidinas/química , Indóis/química , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/química , Antivirais/farmacologia , Antivirais/uso terapêutico , Cristalografia por Raios X/métodos , Humanos , Imidazolidinas/farmacologia , Imidazolidinas/uso terapêutico , Indóis/farmacologia , Indóis/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/fisiologia , Resultado do Tratamento , Inibidores de Proteínas Virais de Fusão/farmacologia
12.
Int J Mol Sci ; 21(11)2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32481690

RESUMO

In the past 17 years, three novel coronaviruses have caused severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and the coronavirus disease 2019 (COVID-19). As emerging infectious diseases, they were characterized by their novel pathogens and transmissibility without available clinical drugs or vaccines. This is especially true for the newly identified COVID-19 caused by SARS coronavirus 2 (SARS-CoV-2) for which, to date, no specific antiviral drugs or vaccines have been approved. Similar to SARS and MERS, the lag time in the development of therapeutics is likely to take months to years. These facts call for the development of broad-spectrum anti-coronavirus drugs targeting a conserved target site. This review will systematically describe potential broad-spectrum coronavirus fusion inhibitors, including antibodies, protease inhibitors, and peptide fusion inhibitors, along with a discussion of their advantages and disadvantages.


Assuntos
Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Inibidores de Proteínas Virais de Fusão/uso terapêutico , Animais , Betacoronavirus/fisiologia , Humanos , Inibidores de Proteases/efeitos adversos , Inibidores de Proteases/farmacologia , SARS-CoV-2 , Inibidores de Proteínas Virais de Fusão/efeitos adversos , Inibidores de Proteínas Virais de Fusão/farmacologia , Internalização do Vírus/efeitos dos fármacos
13.
J Med Chem ; 63(15): 8046-8058, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32407115

RESUMO

Respiratory syncytial virus (RSV) is a seasonal virus that infects the lungs and airways of 64 million children and adults every year. It is a major cause of acute lower respiratory tract infection and is associated with significant morbidity and mortality. Despite the large medical and economic burden, treatment options for RSV-associated bronchiolitis and pneumonia are limited and mainly consist of supportive care. This publication covers the medicinal chemistry efforts resulting in the identification of JNJ-53718678, an orally bioavailable RSV inhibitor that was shown to be efficacious in a phase 2a challenge study in healthy adult subjects and that is currently being evaluated in hospitalized infants and adults. Cocrystal structures of several new derivatives helped in rationalizing some of the structure-activity relationship (SAR) trends observed.


Assuntos
Antivirais/química , Descoberta de Drogas/métodos , Imidazolidinas/química , Indóis/química , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/química , Administração Oral , Antivirais/administração & dosagem , Cristalografia por Raios X/métodos , Células HeLa , Humanos , Imidazolidinas/administração & dosagem , Indóis/administração & dosagem , Estrutura Secundária de Proteína , Vírus Sincicial Respiratório Humano/fisiologia , Inibidores de Proteínas Virais de Fusão/administração & dosagem
15.
J Infect Dis ; 222(9): 1468-1477, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31971597

RESUMO

BACKGROUND: Presatovir is an oral respiratory syncytial virus (RSV) fusion inhibitor targeting RSV F protein. In a double-blind, placebo-controlled study in healthy adults experimentally infected with RSV (Memphis-37b), presatovir significantly reduced viral load and clinical disease severity in a dose-dependent manner. METHODS: Viral RNA from nasal wash samples was amplified and the F gene sequenced to monitor presatovir resistance. Effects of identified amino acid substitutions on in vitro susceptibility to presatovir, viral fitness, and clinical outcome were assessed. RESULTS: Twenty-eight treatment-emergent F substitutions were identified. Of these, 26 were tested in vitro; 2 were not due to lack of recombinant virus recovery. Ten substitutions did not affect presatovir susceptibility, and 16 substitutions reduced RSV susceptibility to presatovir (2.9- to 410-fold). No substitutions altered RSV susceptibility to palivizumab or ribavirin. Frequency of phenotypically resistant substitutions was higher with regimens containing lower presatovir dose and shorter treatment duration. Participants with phenotypic presatovir resistance had significantly higher nasal viral load area under the curve relative to those without, but substitutions did not significantly affect peak viral load or clinical manifestations of RSV disease. CONCLUSIONS: Emergence of presatovir-resistant RSV occurred during therapy but did not significantly affect clinical efficacy in participants with experimental RSV infection.


Assuntos
Indazóis/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Inibidores de Proteínas Virais de Fusão/uso terapêutico , Adolescente , Adulto , Substituição de Aminoácidos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Farmacorresistência Viral/genética , Humanos , Pessoa de Meia-Idade , Vírus Sinciciais Respiratórios/genética , Carga Viral/efeitos dos fármacos , Adulto Jovem
16.
J Am Chem Soc ; 141(32): 12648-12656, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31268705

RESUMO

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) cause lower respiratory infection in infants and young children. There are no vaccines for these pathogens, and existing treatments have limited or questionable efficacy. Infection by HPIV3 or RSV requires fusion of the viral and cell membranes, a process mediated by a trimeric fusion glycoprotein (F) displayed on the viral envelope. Once triggered, the pre-fusion form of F undergoes a series of conformational changes that first extend the molecule to allow for insertion of the hydrophobic fusion peptide into the target cell membrane and then refold the trimeric assembly into an energetically stable post-fusion state, a process that drives the merger of the viral and host cell membranes. Peptides derived from defined regions of HPIV3 F inhibit infection by HPIV3 by interfering with the structural transitions of the trimeric F assembly. Here we describe lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F that potently inhibit infection by both HPIV3 and RSV. The lead peptide inhibits RSV infection as effectively as does a peptide corresponding to the RSV HRC domain itself. We show that the inhibitors bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F with high affinity. Co-crystal structures of inhibitors bound to the HRN domains of HPIV3 or RSV F reveal remarkably different modes of binding in the N-terminal segment of the inhibitor.


Assuntos
Lipopeptídeos/farmacologia , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/farmacologia , Proteínas Virais de Fusão/farmacologia , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Lipopeptídeos/metabolismo , Testes de Sensibilidade Microbiana , Vírus da Parainfluenza 3 Humana/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Mucosa Respiratória/virologia , Vírus Sinciciais Respiratórios/química , Inibidores de Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos
17.
Sci Adv ; 5(4): eaav4580, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30989115

RESUMO

Continuously emerging highly pathogenic human coronaviruses (HCoVs) remain a major threat to human health, as illustrated in past SARS-CoV and MERS-CoV outbreaks. The development of a drug with broad-spectrum HCoV inhibitory activity would address this urgent unmet medical need. Although previous studies have suggested that the HR1 of HCoV spike (S) protein is an important target site for inhibition against specific HCoVs, whether this conserved region could serve as a target for the development of broad-spectrum pan-CoV inhibitor remains controversial. Here, we found that peptide OC43-HR2P, derived from the HR2 domain of HCoV-OC43, exhibited broad fusion inhibitory activity against multiple HCoVs. EK1, the optimized form of OC43-HR2P, showed substantially improved pan-CoV fusion inhibitory activity and pharmaceutical properties. Crystal structures indicated that EK1 can form a stable six-helix bundle structure with both short α-HCoV and long ß-HCoV HR1s, further supporting the role of HR1 region as a viable pan-CoV target site.


Assuntos
Infecções por Coronavirus/virologia , Coronavirus/efeitos dos fármacos , Coronavirus/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/antagonistas & inibidores , Glicoproteína da Espícula de Coronavírus/química , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Coronavirus/classificação , Coronavirus/genética , Infecções por Coronavirus/tratamento farmacológico , Modelos Animais de Doenças , Humanos , Fusão de Membrana/efeitos dos fármacos , Camundongos , Modelos Moleculares , Peptídeos/química , Peptídeos/farmacologia , Filogenia , Conformação Proteica , Domínios Proteicos , Solubilidade
18.
Science ; 363(6431)2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30846569

RESUMO

Recent characterization of broadly neutralizing antibodies (bnAbs) against influenza virus identified the conserved hemagglutinin (HA) stem as a target for development of universal vaccines and therapeutics. Although several stem bnAbs are being evaluated in clinical trials, antibodies are generally unsuited for oral delivery. Guided by structural knowledge of the interactions and mechanism of anti-stem bnAb CR6261, we selected and optimized small molecules that mimic the bnAb functionality. Our lead compound neutralizes influenza A group 1 viruses by inhibiting HA-mediated fusion in vitro, protects mice against lethal and sublethal influenza challenge after oral administration, and effectively neutralizes virus infection in reconstituted three-dimensional cell culture of fully differentiated human bronchial epithelial cells. Cocrystal structures with H1 and H5 HAs reveal that the lead compound recapitulates the bnAb hotspot interactions.


Assuntos
Anticorpos Neutralizantes/química , Materiais Biomiméticos/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/prevenção & controle , Piperazinas/farmacologia , Piridinas/farmacologia , Tetrazóis/farmacologia , Inibidores de Proteínas Virais de Fusão/farmacologia , Internalização do Vírus/efeitos dos fármacos , Administração Oral , Animais , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/farmacocinética , Brônquios/virologia , Células Cultivadas , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Células Madin Darby de Rim Canino , Camundongos , Piperazinas/administração & dosagem , Piperazinas/farmacocinética , Piridinas/administração & dosagem , Piridinas/farmacocinética , Mucosa Respiratória/virologia , Tetrazóis/administração & dosagem , Tetrazóis/farmacocinética , Inibidores de Proteínas Virais de Fusão/administração & dosagem , Inibidores de Proteínas Virais de Fusão/farmacocinética
20.
Mol Pharm ; 15(11): 5005-5018, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30226777

RESUMO

New therapeutic alternatives to fight against the spread of HIV-1 are based on peptides designed to inhibit the early steps of HIV-1 fusion in target cells. However, drawbacks, such as bioavailability, short half-life, rapid clearance, and poor ability to cross the physiological barriers, make such peptides unattractive for the pharmaceutical industry. Here we developed, optimized, and characterized polymeric nanoparticles (NPs) coated with glycol chitosan to incorporate and release an HIV-1 fusion inhibitor peptide (E1) inside the vaginal mucosa. The NPs were prepared by a modified double emulsion method, and optimization was carried out by a factorial design. In vitro, ex vivo, and in vivo studies were carried out to evaluate the optimized formulation. The results indicate that the physicochemical features of these NPs enable them to incorporate and release HIV fusion inhibitor peptides to the vaginal mucosa before the fusion step takes place.


Assuntos
Portadores de Fármacos/química , HIV-1/efeitos dos fármacos , Peptídeos/administração & dosagem , Inibidores de Proteínas Virais de Fusão/administração & dosagem , Administração Intravaginal , Animais , Quitosana/química , Desenho de Fármacos , Feminino , Proteína gp41 do Envelope de HIV/antagonistas & inibidores , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , HIV-1/fisiologia , Modelos Animais , Mucosa/efeitos dos fármacos , Mucosa/metabolismo , Mucosa/virologia , Nanopartículas/química , Tamanho da Partícula , Peptídeos/química , Peptídeos/farmacocinética , Suínos , Vagina/efeitos dos fármacos , Vagina/metabolismo , Vagina/virologia , Proteínas do Envelope Viral/química , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/farmacocinética , Internalização do Vírus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...