Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
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.
Antiviral Res ; 192: 105120, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34126139

RESUMO

In recent years, a number of novel filoviruses (e.g. Lloviu virus (LLOV) and Bombali virus (BOMV)) have been discovered. While antibody-based therapeutics have recently been approved for treatment of infections with the filovirus Ebola virus (EBOV), no treatment options for novel filoviruses currently exist. Further, the development of antivirals against them is complicated by the fact that only sequence information, but no actual virus isolates, are available. To address this issue, we developed a reverse genetics-based minigenome system for BOMV, which allows us to assess the activity of the BOMV polymerase. Together with similar systems that we have developed for other filoviruses in the past (i.e. LLOV and Reston virus (RESTV)), we then assessed the efficiency of remdesivir, a known inhibitor of the EBOV polymerase that has recently been tested in a clinical trial for efficacy against Ebola disease. We show that remdesivir is indeed also active against the polymerases of BOMV, LLOV, and RESTV, with comparable IC50 values to its activity against EBOV. This suggests that treatment with remdesivir might represent a viable option in case of infections with novel filoviruses.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , Filoviridae/efeitos dos fármacos , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Monofosfato de Adenosina/farmacologia , Alanina/farmacologia , Linhagem Celular , Ebolavirus/efeitos dos fármacos , Filoviridae/classificação , Filoviridae/genética , Humanos , Concentração Inibidora 50 , Filogenia , Replicação Viral/efeitos dos fármacos
3.
Proc Natl Acad Sci U S A ; 117(48): 30687-30698, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33184176

RESUMO

The SARS-CoV-2 pandemic has made it clear that we have a desperate need for antivirals. We present work that the mammalian SKI complex is a broad-spectrum, host-directed, antiviral drug target. Yeast suppressor screening was utilized to find a functional genetic interaction between proteins from influenza A virus (IAV) and Middle East respiratory syndrome coronavirus (MERS-CoV) with eukaryotic proteins that may be potential host factors involved in replication. This screening identified the SKI complex as a potential host factor for both viruses. In mammalian systems siRNA-mediated knockdown of SKI genes inhibited replication of IAV and MERS-CoV. In silico modeling and database screening identified a binding pocket on the SKI complex and compounds predicted to bind. Experimental assays of those compounds identified three chemical structures that were antiviral against IAV and MERS-CoV along with the filoviruses Ebola and Marburg and two further coronaviruses, SARS-CoV and SARS-CoV-2. The mechanism of antiviral activity is through inhibition of viral RNA production. This work defines the mammalian SKI complex as a broad-spectrum antiviral drug target and identifies lead compounds for further development.


Assuntos
Antivirais/farmacologia , Coronavirus/efeitos dos fármacos , Filoviridae/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Complexos Multiproteicos/metabolismo , Orthomyxoviridae/efeitos dos fármacos , Linhagem Celular , Genes Supressores , Modelos Moleculares , Terapia de Alvo Molecular , Ligação Proteica , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
4.
Antiviral Res ; 183: 104932, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32946918

RESUMO

Ebolaviruses and marburgviruses, members of the family Filoviridae, are known to cause fatal diseases often associated with hemorrhagic fever. Recent outbreaks of Ebola virus disease in West African countries and the Democratic Republic of the Congo have made clear the urgent need for the development of therapeutics and vaccines against filoviruses. Using replication-incompetent vesicular stomatitis virus (VSV) pseudotyped with the Ebola virus (EBOV) envelope glycoprotein (GP), we screened a chemical compound library to obtain new drug candidates that inhibit filoviral entry into target cells. We discovered a biaryl sulfonamide derivative that suppressed in vitro infection mediated by GPs derived from all known human-pathogenic filoviruses. To determine the inhibitory mechanism of the compound, we monitored each entry step (attachment, internalization, and membrane fusion) using lipophilic tracer-labeled ebolavirus-like particles and found that the compound efficiently blocked fusion between the viral envelope and the endosomal membrane during cellular entry. However, the compound did not block the interaction of GP with the Niemann-Pick C1 protein, which is believed to be the receptor of filoviruses. Using replication-competent VSVs pseudotyped with EBOV GP, we selected escape mutants and identified two EBOV GP amino acid residues (positions 47 and 66) important for the interaction with this compound. Interestingly, these amino acid residues were located at the base region of the GP trimer, suggesting that the compound might interfere with the GP conformational change required for membrane fusion. These results suggest that this biaryl sulfonamide derivative is a novel fusion inhibitor and a possible drug candidate for the development of a pan-filovirus therapeutic.


Assuntos
Filoviridae/efeitos dos fármacos , Sulfonamidas/química , Sulfonamidas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Chlorocebus aethiops , Descoberta de Drogas , Ebolavirus/efeitos dos fármacos , Filoviridae/classificação , Infecções por Filoviridae/tratamento farmacológico , Infecções por Filoviridae/virologia , Células HEK293 , Doença pelo Vírus Ebola/tratamento farmacológico , Humanos , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus/efeitos dos fármacos , Receptores Virais/metabolismo , Células Vero
5.
J Med Chem ; 63(19): 11085-11099, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32886512

RESUMO

Filoviridae, including Ebola (EBOV) and Marburg (MARV) viruses, are emerging pathogens that pose a serious threat to public health. No agents have been approved to treat filovirus infections, representing a major unmet medical need. The selective estrogen receptor modulator (SERM) toremifene was previously identified from a screen of FDA-approved drugs as a potent EBOV viral entry inhibitor, via binding to EBOV glycoprotein (GP). A focused screen of ER ligands identified ridaifen-B as a potent dual inhibitor of EBOV and MARV. Optimization and reverse-engineering to remove ER activity led to a novel compound 30 (XL-147) showing potent inhibition against infectious EBOV Zaire (0.09 µM) and MARV (0.64 µM). Mutagenesis studies confirmed that inhibition of EBOV viral entry is mediated by the direct interaction with GP. Importantly, compound 30 displayed a broad-spectrum antifilovirus activity against Bundibugyo, Tai Forest, Reston, and Menglà viruses and is the first submicromolar antiviral agent reported for some of these strains, therefore warranting further development as a pan-filovirus inhibitor.


Assuntos
Antivirais/farmacologia , Filoviridae/efeitos dos fármacos , Receptores de Estrogênio/efeitos dos fármacos , Antivirais/química , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Filoviridae/fisiologia , Humanos , Ligantes , Fusão de Membrana/efeitos dos fármacos , Modelos Biológicos , Relação Estrutura-Atividade
6.
Eur J Med Chem ; 204: 112595, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32707357

RESUMO

Filoviruses, including Ebolavirus (EBOV), Marburgvirus (MARV) and Cuevavirus, cause hemorrhagic fevers in humans with up to 90% mortality rates. In the 2014-2016 West Africa Ebola epidemic, there are 15,261 laboratory confirmed cases and 11,325 total deaths. The lack of effective vaccines and medicines for the prevention and treatment of filovirus infection in humans stresses the urgency to develop antiviral therapeutics against filovirus-associated diseases. Our previous study identified a histamine receptor antagonist compound CP19 as an entry inhibitor against both EBOV and MARV. The preliminary structure-activity relationship (SAR) studies of CP19 showed that its piperidine, coumarin and linker were related with its antiviral activities. In this study, we performed detailed SAR studies on these groups with synthesized CP19 derivatives. We discovered that 1) the piperidine group could be optimized with heterocycles, 2) the substitution groups of C3 and C4 of coumarin should be relatively large hydrophobic groups and 3) the linker part should be least substituted. Based on the SAR analysis, we synthesized compound 32 as a potent entry inhibitor of EBOV and MARV (IC50 = 0.5 µM for EBOV and 1.5 µM for MARV). The mutation studies of Ebola glycoprotein and molecular docking studies showed that the coumarin and its substituted groups of compound 32 bind to the pocket of Ebola glycoprotein in a similar way to the published entry inhibitor compound 118a. However, the carboxamide group of compound 32 does not have strong interaction with N61 as compound 118a does. The coumarin skeleton structure and the binding model of compound 32 elucidated by this study could be utilized to guide further design and optimization of entry inhibitors targeting the filovirus glycoproteins.


Assuntos
Cumarínicos/química , Cumarínicos/farmacologia , Desenho de Fármacos , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Antivirais/química , Antivirais/farmacologia , Filoviridae/metabolismo , Terapia de Alvo Molecular , Piperidinas/química , Relação Estrutura-Atividade
7.
Virus Genes ; 56(2): 150-167, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32076918

RESUMO

The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.


Assuntos
Antivirais/uso terapêutico , Arbovírus/efeitos dos fármacos , Bioterrorismo/prevenção & controle , Viroses/tratamento farmacológico , Arbovírus/patogenicidade , Filoviridae/efeitos dos fármacos , Filoviridae/patogenicidade , Humanos , Orthobunyavirus/efeitos dos fármacos , Orthobunyavirus/patogenicidade , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/patogenicidade , Paramyxovirinae/efeitos dos fármacos , Paramyxovirinae/patogenicidade , Poxviridae/efeitos dos fármacos , Poxviridae/patogenicidade , Viroses/virologia
8.
mBio ; 10(4)2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31337716

RESUMO

The filoviruses are etiological agents of life-threatening hemorrhagic fever with high mortality rate and risk of potential outbreak. Among members of this family, the Ebola (EBOV), Sudan (SUDV), and Marburg (MARV) viruses are considered the most pathogenic for humans. The ebolavirus nucleoprotein (NP) is the most abundant protein in infected cells and is essential for viral transcription and replication; thus, it represents an attractive target for therapeutic intervention. Here, we present the structure of SUDV NP in complex with the amino-terminal portion of the phosphoprotein VP35 at 2.3 Å. This structure captures VP35 chaperoning SUDV NP in a monomeric and RNA-free state. This transient state has been proposed to be key to maintaining a pool of monomeric and RNA-free NPs prior to NP-NP polymerization and encapsidation of the viral RNA genome. This structure also reveals a newly visualized interaction between NP and VP35, a well-defined beta sheet that is not present in previous structures. Affinity binding assays demonstrate that this beta sheet is essential for maintaining the high-affinity interaction between VP35 and a hydrophobic pocket on SUDV NP, and electron microscopy indicates the importance of this binding interaction to the oligomeric state and assembly of NP in human cells. Complementary structure-directed mutagenesis identifies critical residues conserved across the filovirus family that could be targeted by broadly effective antivirals.IMPORTANCE Outbreaks of the filoviruses can be unpredictable in timing, location, and identity of the causative virus, with each of Ebola virus, Sudan virus, Bundibugyo virus, and Marburg virus reemerging in the last several years to cause human disease with 30 to 90% lethality. The 2014-2016 outbreak in particular, with nearly 30,000 patients, highlighted the ability of these viruses to emerge unexpectedly and spread rapidly. Two ebolavirus outbreaks have emerged this year, yet we still lack FDA-approved drugs with pan-filovirus activity to treat existing and emergent ebolaviruses. For all filoviruses, the interaction between the nucleoprotein and the phosphoprotein is essential for the virus life cycle and is a potential target for therapeutic intervention. In this report, we describe the crystal structure of the SUDV nucleoprotein with the interacting domain of the viral phosphoprotein, and we identify residues critical for high-affinity interaction and for control of the oligomeric state of the nucleoprotein. Structural comparison of this heterodimer with other members of the filovirus family allowed us to find conserved and essential atomic features that will facilitate understanding of the virus life cycle and the rational design of antivirals.


Assuntos
Ebolavirus/efeitos dos fármacos , Filoviridae/efeitos dos fármacos , Nucleoproteínas/química , Proteínas Virais Reguladoras e Acessórias/química , Cristalografia por Raios X , Filoviridae/patogenicidade , Fosfoproteínas/química , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas
9.
Antiviral Res ; 170: 104569, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31356830

RESUMO

Viral hemorrhagic fevers (VHFs) cause thousands of fatalities every year, but the treatment options for their management remain very limited. In particular, the development of therapeutic interventions is restricted by the lack of commercial viability of drugs targeting individual VHF agents. This makes approaches like drug repurposing and/or the identification of broad range therapies (i.e. those directed at host responses or common proviral factors) highly attractive. However, the identification of candidates for such antiviral repurposing or of host factors/pathways important for the virus life cycle is reliant on high-throughput screening (HTS). Recently, such screening work has been increasingly facilitated by the availability of reverse genetics-based approaches, including tools such as full-length clone (FLC) systems to generate reporter-expressing viruses or various life cycle modelling (LCM) systems, many of which have been developed and/or greatly improved during the last years. In particular, since LCM systems are capable of modelling specific steps in the life cycle, they are a valuable tool for both targeted screening (i.e. for inhibitors of a specific pathway) and mechanism of action studies. This review seeks to summarize the currently available reverse genetics systems for negative-sense VHF causing viruses (i.e. arenaviruses, bunyaviruses and filoviruses), and to highlight the recent advancements made in applying these systems for HTS to identify either antivirals or new virus-host interactions that might hold promise for the development of future treatments for the infections caused by these deadly but neglected virus groups.


Assuntos
Arenaviridae/genética , Bunyaviridae/genética , Filoviridae/genética , Febres Hemorrágicas Virais/virologia , Ensaios de Triagem em Larga Escala , Genética Reversa/métodos , Antivirais/isolamento & purificação , Arenaviridae/efeitos dos fármacos , Bunyaviridae/efeitos dos fármacos , Filoviridae/efeitos dos fármacos , Genoma Viral , Interações entre Hospedeiro e Microrganismos , Humanos
10.
Curr Opin Virol ; 35: 42-56, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31003196

RESUMO

The filovirus family includes some of the deadliest viruses known, including Ebola virus and Marburg virus. These viruses cause periodic outbreaks of severe disease that can be spread from person to person, making the filoviruses important public health threats. There remains a need for approved drugs that target all or most members of this virus family. Small molecule inhibitors that target conserved functions hold promise as pan-filovirus therapeutics. To date, compounds that effectively target virus entry, genome replication, gene expression, and virus egress have been described. The most advanced inhibitors are nucleoside analogs that target viral RNA synthesis reactions.


Assuntos
Antivirais/farmacologia , Desenvolvimento de Medicamentos , Ebolavirus/efeitos dos fármacos , Filoviridae/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antivirais/química , Antivirais/isolamento & purificação , Ensaios Clínicos como Assunto , Infecções por Filoviridae/tratamento farmacológico , Doença pelo Vírus Ebola/tratamento farmacológico , Humanos , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos
11.
Viruses ; 11(3)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30832223

RESUMO

Filoviruses, such as Ebola virus (EBOV) and Marburg virus, are causative agents of unpredictable outbreaks of severe hemorrhagic fevers in humans and non-human primates. For infection, filoviral particles need to be internalized and delivered to intracellular vesicles containing cathepsin proteases and the viral receptor Niemann-Pick C1. Previous studies have shown that EBOV triggers macropinocytosis of the viral particles in a glycoprotein (GP)-dependent manner, but the molecular events required for filovirus internalization remain mostly unknown. Here we report that the diacylglycerol kinase inhibitor, R-59-022, blocks EBOV GP-mediated entry into Vero cells and bone marrow-derived macrophages. Investigation of the mode of action of the inhibitor revealed that it blocked an early step in entry, more specifically, the internalization of the viral particles via macropinocytosis. Finally, R-59-022 blocked viral entry mediated by a panel of pathogenic filovirus GPs and inhibited growth of replicative Ebola virus. Taken together, our studies suggest that R-59-022 could be used as a tool to investigate macropinocytic uptake of filoviruses and could be a starting point for the development of pan-filoviral therapeutics.


Assuntos
Diacilglicerol Quinase/antagonistas & inibidores , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Pirimidinonas/farmacologia , Tiazóis/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Chlorocebus aethiops , Ebolavirus/fisiologia , Células HEK293 , Humanos , Macrófagos/virologia , Marburgvirus/fisiologia , Pinocitose/efeitos dos fármacos , Receptores Virais , Células Vero , Replicação Viral/efeitos dos fármacos
12.
Antiviral Res ; 162: 90-100, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550800

RESUMO

Filoviruses, which include Ebola virus (EBOV) and Marburg virus, are negative-sense RNA viruses associated with sporadic outbreaks of severe viral hemorrhagic fever characterized by uncontrolled virus replication. The extreme virulence and emerging nature of these zoonotic pathogens make them a significant threat to human health. Replication of the filovirus genome and production of viral RNAs require the function of a complex of four viral proteins, the nucleoprotein (NP), viral protein 35 (VP35), viral protein 30 (VP30) and large protein (L). The latter performs the enzymatic activities required for production of viral RNAs and capping of viral mRNAs. Although it has been recognized that interactions between the virus-encoded components of the EBOV RNA polymerase complex are required for viral RNA synthesis reactions, specific molecular details have, until recently, been lacking. New efforts have combined structural biology and molecular virology to reveal in great detail the molecular basis for critical protein-protein interactions (PPIs) necessary for viral RNA synthesis. These efforts include recent studies that have identified a range of interacting host factors and in some instances demonstrated unique mechanisms by which they act. For a select number of these interactions, combined use of mutagenesis, over-expressing of peptides corresponding to PPI interfaces and identification of small molecules that disrupt PPIs have demonstrated the functional significance of virus-virus and virus-host PPIs and suggest several as potential targets for therapeutic intervention.


Assuntos
Filoviridae/fisiologia , Interações entre Hospedeiro e Microrganismos , RNA Viral/biossíntese , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Filoviridae/efeitos dos fármacos , Humanos , Marburgvirus/efeitos dos fármacos , Marburgvirus/fisiologia , Ligação Proteica , Proteínas Virais/antagonistas & inibidores , Viroses/tratamento farmacológico
13.
Antiviral Res ; 157: 47-56, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29981374

RESUMO

Ebola and Marburg are filoviruses and biosafety level 4 pathogens responsible for causing severe hemorrhagic fevers in humans with mortality rates up to 90%. The most recent outbreak in West Africa resulted in approximately 11,310 deaths in 28,616 reported cases. Currently there are no FDA-approved vaccines or therapeutics to treat infections of these deadly viruses. Recently we screened an FDA-approved drug library and identified numerous G protein-coupled receptor (GPCR) antagonists including antihistamines possessing anti-filovirus properties. Antihistamines are attractive targets for drug repurposing because of their low cost and ease of access due to wide use. In this report we identify common over the counter antihistamines, such as diphenhydramine (Benadryl) and chlorcyclizine (Ahist) as potential candidates for repurposing as anti-filovirus agents. Furthermore, we demonstrate that this potential is wide-spread through the 1st generation of H1-specific antihistamines but is not present in newer drugs or drugs targeting H2, H3 and H4 receptors. We showed that the filovirus entry inhibition is not dependent on the classical antagonism of cell surface histamine or muscarinic acetylcholine receptors but occurs in the endosome, like the cathepsin inhibitor CA-074. Finally, using extensive docking studies we showed the potential for these drugs to bind directly to the EBOV-GP at the same site as toremifene. These findings suggest that the 1st generation antihistamines are excellent candidates for repurposing as anti-filovirus therapeutics and can be further optimized for removal of unwanted histamine or muscarinic receptor interactions without loss of anti-filovirus efficacy.


Assuntos
Antivirais/farmacologia , Reposicionamento de Medicamentos , Filoviridae/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos/farmacologia , Células A549 , Difenidramina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Piperazinas/farmacologia , Internalização do Vírus/efeitos dos fármacos
14.
Eur J Med Chem ; 149: 45-55, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29494844

RESUMO

Preventing filoviruses in the entry stage is an attractive antiviral strategy. Taking aloperine, a Chinese natural herb with an endocyclic skeleton, as the lead, 23 new aloperine derivatives were synthesized and evaluated for their anti-filovirus activities including ebola virus (EBOV) and marburg virus (MARV) using pseudotyped virus model. Structure-activity relationship (SAR) analysis indicated that the introduction of a 12N-dichlorobenzyl group was beneficial for the potency. Compound 2e exhibited the most potent anti-EBOV and anti-MARV effects both in vitro and in vivo. It also displayed a good pharmacokinetic and safety profile in vivo, indicating an ideal druglike feature. The primary mechanism study showed that 2e could block a late stage of viral entry, mainly through inhibiting cysteine cathepsin B activity of host components. We consider compound 2e to be a promising broad-spectrum anti-filovirus agent with the advantages of a unique chemical scaffold and a specific biological mechanism.


Assuntos
Antivirais/farmacologia , Filoviridae/efeitos dos fármacos , Piperidinas/síntese química , Piperidinas/uso terapêutico , Internalização do Vírus/efeitos dos fármacos , Antivirais/síntese química , Catepsina B/antagonistas & inibidores , Clorobenzenos , Humanos , Piperidinas/farmacologia , Quinolizidinas , Relação Estrutura-Atividade
15.
ACS Infect Dis ; 4(5): 673-685, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29457711

RESUMO

The development of novel therapeutics and vaccines to treat or prevent disease caused by filoviruses, such as Ebola and Marburg viruses, depends on the availability of animal models that faithfully recapitulate clinical hallmarks of disease as it is observed in humans. In particular, small animal models (such as mice and guinea pigs) are historically and frequently used for the primary evaluation of antiviral countermeasures, prior to testing in nonhuman primates, which represent the gold-standard filovirus animal model. In the past several years, however, the filovirus field has witnessed the continued refinement of the mouse and guinea pig models of disease, as well as the introduction of the hamster and ferret models. We now have small animal models for most human-pathogenic filoviruses, many of which are susceptible to wild type virus and demonstrate key features of disease, including robust virus replication, coagulopathy, and immune system dysfunction. Although none of these small animal model systems perfectly recapitulates Ebola virus disease or Marburg virus disease on its own, collectively they offer a nearly complete set of tools in which to carry out the preclinical development of novel antiviral drugs.


Assuntos
Modelos Animais de Doenças , Infecções por Filoviridae/tratamento farmacológico , Infecções por Filoviridae/prevenção & controle , Filoviridae/patogenicidade , Animais , Cricetinae , Furões , Filoviridae/efeitos dos fármacos , Cobaias , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Doença do Vírus de Marburg/tratamento farmacológico , Doença do Vírus de Marburg/prevenção & controle , Camundongos , Vacinas Virais
16.
Antiviral Res ; 150: 183-192, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29305306

RESUMO

There are no approved medications for the treatment of Marburg or Ebola virus infection. In two previous articles (Martin et al., 2016, Martin et al., 2017), we reviewed surface glycoprotein and replication proteins structure/function relationship to decipher the molecular mechanisms of filovirus life cycle and identify antiviral strategies. In the present article, we recapitulate knowledge about the viral proteins involved in filovirus assembly and budding. First we describe the structural data available for viral proteins associated with virus assembly and virion egress and then, we integrate the structural features of these proteins in the functional context of the viral replication cycle. Finally, we summarize recent advances in the development of innovative antiviral strategies to target filovirus assembly and egress. The development of such prophylactic or post-exposure treatments could help controlling future filovirus outbreaks.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Antivirais/química , Descoberta de Drogas/métodos , Filoviridae/classificação , Genoma Viral , Genômica/métodos , Humanos , Relação Estrutura-Atividade , Proteínas Virais/antagonistas & inibidores
17.
Expert Rev Anti Infect Ther ; 16(1): 67-76, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29210303

RESUMO

INTRODUCTION: During the 2014-2016 Ebolavirus (EBOV) outbreak, several candidate therapeutics were used in EBOV-infected patients in clinical trials and under expanded access for emergency use. This review will focus briefly on medications used during the outbreak. We will discuss current therapeutic candidates and their status and will then turn to a related and essential topic: supportive care and the standard of care for filovirus infected patients. Potential benefits and pitfalls of combination therapies for filoviruses will be discussed. Areas covered: Clinical trials of therapeutics targeting EBOV; clinical usage of therapeutics during recent EBOV outbreak; potential need for combination therapy; role of supportive care in treatment of Ebola virus disease (EVD). Expert commentary: In the absence of another large scale EBOV outbreak, the path to therapeutic product licensure in the United States of America (USA) would need to be via the FDA Animal Rule. However, human data may be needed to supplement animal data. The future of filovirus therapeutics may therefore benefit by establishing the ability to implement clinical trials in an outbreak setting in a timely fashion. Supportive care guidelines for filovirus infection should be defined and established as standard of care for treatment of EVD.


Assuntos
Antivirais/uso terapêutico , Surtos de Doenças , Doença pelo Vírus Ebola/tratamento farmacológico , Animais , Antivirais/administração & dosagem , Aprovação de Drogas , Desenho de Fármacos , Quimioterapia Combinada , Ebolavirus/efeitos dos fármacos , Ebolavirus/isolamento & purificação , Filoviridae/efeitos dos fármacos , Filoviridae/isolamento & purificação , Infecções por Filoviridae/tratamento farmacológico , Infecções por Filoviridae/epidemiologia , Doença pelo Vírus Ebola/epidemiologia , Humanos
18.
Antiviral Res ; 149: 154-163, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175127

RESUMO

Members of the family Filoviridae cause severe, often fatal disease in humans, for which there are no approved vaccines and only a few experimental drugs tested in animal models. Retro-2, a small molecule that inhibits retrograde trafficking of bacterial and plant toxins inside host cells, has been demonstrated to be effective against a range of bacterial and virus pathogens, both in vitro and in animal models. Here, we demonstrated that Retro-2 and its derivatives, Retro-2.1 and compound 25, blocked infection by Ebola virus and Marburg virus in vitro. We show that the derivatives were more potent inhibitors of infection as compared to the parent compound. Pseudotyped virus assays indicated that the compounds affected virus entry into cells while virus particle localization to Niemann-Pick C1-positive compartments showed that they acted at a late step in virus entry. Our work demonstrates a potential for Retro-type drugs to be developed into anti-filoviral therapeutics.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Filoviridae/efeitos dos fármacos , Tiofenos/farmacologia , Animais , Antivirais/química , Benzamidas/química , Linhagem Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Filoviridae/fisiologia , Humanos , Marburgvirus/efeitos dos fármacos , Marburgvirus/fisiologia , Testes de Sensibilidade Microbiana , Estrutura Molecular , Tiofenos/química , Internalização do Vírus/efeitos dos fármacos
19.
ACS Infect Dis ; 3(3): 190-198, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-28152588

RESUMO

The 2014 Ebola outbreak in West Africa, the largest outbreak on record, highlighted the need for novel approaches to therapeutics targeting Ebola virus (EBOV). Within the EBOV replication complex, the interaction between polymerase cofactor, viral protein 35 (VP35), and nucleoprotein (NP) is critical for viral RNA synthesis. We recently identified a peptide at the N-terminus of VP35 (termed NPBP) that is sufficient for interaction with NP and suppresses EBOV replication, suggesting that the NPBP binding pocket can serve as a potential drug target. Here we describe the development and validation of a sensitive high-throughput screen (HTS) using a fluorescence polarization assay. Initial hits from this HTS include the FDA-approved compound tolcapone, whose potency against EBOV infection was validated in a nonfluorescent secondary assay. High conservation of the NP-VP35 interface among filoviruses suggests that this assay has the capacity to identify pan-filoviral inhibitors for development as antivirals.


Assuntos
Antivirais/farmacologia , Filoviridae/fisiologia , Nucleoproteínas/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Sequência de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Sequência Conservada , Avaliação Pré-Clínica de Medicamentos , Filoviridae/efeitos dos fármacos , Filoviridae/genética , Polarização de Fluorescência , Ensaios de Triagem em Larga Escala , Técnicas In Vitro , Modelos Moleculares , Ligação Proteica/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral/efeitos dos fármacos
20.
Antiviral Res ; 141: 48-61, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28192094

RESUMO

Filoviruses are important pathogens that cause severe and often fatal hemorrhagic fever in humans, for which no approved vaccines and antiviral treatments are yet available. In an earlier article (Martin et al., Antiviral Research, 2016), we reviewed the role of the filovirus surface glycoprotein in replication and as a target for drugs and vaccines. In this review, we focus on recent findings on the filovirus replication machinery and how they could be used for the identification of new therapeutic targets and the development of new antiviral compounds. First, we summarize the recent structural and functional advances on the molecules involved in filovirus replication/transcription cycle, particularly the NP, VP30, VP35 proteins, and the "large" protein L, which harbors the RNA-dependent RNA polymerase (RdRp) and mRNA capping activities. These proteins are essential for viral mRNA synthesis and genome replication, and consequently they constitute attractive targets for drug design. We then describe how these insights into filovirus replication mechanisms and the structure/function characterization of the involved proteins have led to the development of new and innovative antiviral strategies that may help reduce the filovirus disease case fatality rate through post-exposure or prophylactic treatments.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Proteínas Virais/metabolismo , Replicação Viral , Antivirais/química , Antivirais/metabolismo , Antivirais/uso terapêutico , Desenho de Fármacos , Ebolavirus/química , Ebolavirus/efeitos dos fármacos , Ebolavirus/metabolismo , Infecções por Filoviridae/tratamento farmacológico , Marburgvirus/química , Marburgvirus/efeitos dos fármacos , Marburgvirus/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/isolamento & purificação , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA