Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38003463

RESUMEN

Stimulator of interferon genes (STING) agonists have shown potent anti-tumor efficacy in various mouse tumor models and have the potential to overcome resistance to immune checkpoint inhibitors (ICI) by linking the innate and acquired immune systems. First-generation STING agonists are administered intratumorally; however, a systemic delivery route would greatly expand the clinical use of STING agonists. Biochemical and cell-based experiments, as well as syngeneic mouse efficacy models, were used to demonstrate the anti-tumoral activity of ALG-031048, a novel STING agonist. In vitro, ALG-031048 is highly stable in plasma and liver microsomes and is resistant to degradation via phosphodiesterases. The high stability in biological matrices translated to good cellular potency in a HEK 293 STING R232 reporter assay, efficient activation and maturation of primary human dendritic cells and monocytes, as well as long-lasting, antigen-specific anti-tumor activity in up to 90% of animals in the CT26 mouse colon carcinoma model. Significant reductions in tumor growth were observed in two syngeneic mouse tumor models following subcutaneous administration. Combinations of ALG-031048 and ICIs further enhanced the in vivo anti-tumor activity. This initial demonstration of anti-tumor activity after systemic administration of ALG-031048 warrants further investigation, while the combination of systemically administered ALG-031048 with ICIs offers an attractive approach to overcome key limitations of ICIs in the clinic.


Asunto(s)
Neoplasias del Colon , Neoplasias , Ratones , Animales , Humanos , Células HEK293 , Neoplasias/patología , Neoplasias del Colon/tratamiento farmacológico , Modelos Animales de Enfermedad , Inmunoterapia , Microambiente Tumoral
2.
J Virol ; 97(10): e0072223, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37754761

RESUMEN

IMPORTANCE: Chronic hepatitis B is the most important cause of liver cancer worldwide and affects more than 290 million people. Current treatments are mostly suppressive and rarely lead to a cure. Therefore, there is a need for novel and curative drugs that target the host or the causative agent, hepatitis B virus itself. Capsid assembly modulators are an interesting class of antiviral molecules that may one day become part of curative treatment regimens for chronic hepatitis B. Here we explore the characteristics of a particularly interesting subclass of capsid assembly modulators. These so-called non-HAP CAM-As have intriguing properties in cell culture but also clear virus-infected cells from the mouse liver in a gradual and sustained way. We believe they represent a considerable improvement over previously reported molecules and may one day be part of curative treatment combinations for chronic hepatitis B.


Asunto(s)
Antivirales , Cápside , Virus de la Hepatitis B , Hepatitis B Crónica , Ensamble de Virus , Animales , Humanos , Ratones , Antivirales/clasificación , Antivirales/farmacología , Antivirales/uso terapéutico , Cápside/química , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/efectos de los fármacos , Proteínas de la Cápside/metabolismo , Células Cultivadas , Virus de la Hepatitis B/química , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/metabolismo , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Técnicas In Vitro , Ensamble de Virus/efectos de los fármacos , Modelos Animales de Enfermedad
3.
Hepatology ; 78(4): 1252-1265, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37102495

RESUMEN

BACKGROUND AND AIMS: Effective therapies leading to a functional cure for chronic hepatitis B are still lacking. Class A capsid assembly modulators (CAM-As) are an attractive modality to address this unmet medical need. CAM-As induce aggregation of the HBV core protein (HBc) and lead to sustained HBsAg reductions in a chronic hepatitis B mouse model. Here, we investigate the underlying mechanism of action for CAM-A compound RG7907. APPROACH AND RESULTS: RG7907 induced extensive HBc aggregation in vitro , in hepatoma cells, and in primary hepatocytes. In the adeno-associated virus (AAV)-HBV mouse model, the RG7907 treatment led to a pronounced reduction in serum HBsAg and HBeAg, concomitant with clearance of HBsAg, HBc, and AAV-HBV episome from the liver. Transient increases in alanine transaminase, hepatocyte apoptosis, and proliferation markers were observed. These processes were confirmed by RNA sequencing, which also uncovered a role for interferon alpha and gamma signaling, including the interferon-stimulated gene 15 (ISG15) pathway. Finally, the in vitro observation of CAM-A-induced HBc-dependent cell death through apoptosis established the link of HBc aggregation to in vivo loss of infected hepatocytes. CONCLUSIONS: Our study unravels a previously unknown mechanism of action for CAM-As such as RG7907 in which HBc aggregation induces cell death, resulting in hepatocyte proliferation and loss of covalently closed circular DNA or its equivalent, possibly assisted by an induced innate immune response. This represents a promising approach to attain a functional cure for chronic hepatitis B.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Ratones , Animales , Virus de la Hepatitis B , Antígenos de Superficie de la Hepatitis B/metabolismo , Cápside/metabolismo , Hepatocitos/metabolismo , Interferón-alfa/farmacología , Hepatitis B/metabolismo , ADN Viral/genética
4.
Biochem Biophys Res Commun ; 555: 134-139, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813272

RESUMEN

There is an urgent need for antivirals targeting the SARS-CoV-2 virus to fight the current COVID-19 pandemic. The SARS-CoV-2 main protease (3CLpro) represents a promising target for antiviral therapy. The lack of selectivity for some of the reported 3CLpro inhibitors, specifically versus cathepsin L, raises potential safety and efficacy concerns. ALG-097111 potently inhibited SARS-CoV-2 3CLpro (IC50 = 7 nM) without affecting the activity of human cathepsin L (IC50 > 10 µM). When ALG-097111 was dosed in hamsters challenged with SARS-CoV-2, a robust and significant 3.5 log10 (RNA copies/mg) reduction of the viral RNA copies and 3.7 log10 (TCID50/mg) reduction in the infectious virus titers in the lungs was observed. These results provide the first in vivo validation for the SARS-CoV-2 3CLpro as a promising therapeutic target for selective small molecule inhibitors.


Asunto(s)
Amidas/farmacología , Tratamiento Farmacológico de COVID-19 , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Modelos Animales de Enfermedad , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , Amidas/farmacocinética , Animales , COVID-19/virología , Catepsina L/antagonistas & inhibidores , Línea Celular , Cricetinae , Inhibidores de Cisteína Proteinasa/farmacocinética , Femenino , Humanos , Concentración 50 Inhibidora , Masculino , Mesocricetus/virología , Reproducibilidad de los Resultados , SARS-CoV-2/crecimiento & desarrollo , Serina Endopeptidasas , Especificidad por Sustrato , Replicación Viral/efectos de los fármacos
5.
PLoS One ; 15(12): e0240338, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33306682

RESUMEN

Thyroid hormones are important modulators of metabolic activity in mammals and alter cholesterol and fatty acid levels through activation of the nuclear thyroid hormone receptor (THR). Currently, there are several THRß agonists in clinical trials for the treatment of non-alcoholic steatohepatitis (NASH) that have demonstrated the potential to reduce liver fat and restore liver function. In this study, we tested three THRß-agonism-based NASH treatment candidates, GC-1 (sobetirome), MGL-3196 (resmetirom), and VK2809, and compared their selectivity for THRß and their ability to modulate the expression of genes specific to cholesterol and fatty acid biosynthesis and metabolism in vitro using human hepatic cells and in vivo using a rat model. Treatment with GC-1 upregulated the transcription of CPT1A in the human hepatocyte-derived Huh-7 cell line with a dose-response comparable to that of the native THR ligand, triiodothyronine (T3). VK2809A (active parent of VK2809), MGL-3196, and VK2809 were approximately 30-fold, 1,000-fold, and 2,000-fold less potent than T3, respectively. Additionally, these relative potencies were confirmed by quantification of other direct gene targets of THR, namely, ANGPTL4 and DIO1. In primary human hepatocytes, potencies were conserved for every compound except for VK2809, which showed significantly increased potency that was comparable to that of its active counterpart, VK2809A. In high-fat diet fed rats, a single dose of T3 significantly reduced total cholesterol levels and concurrently increased liver Dio1 and Me1 RNA expression. MGL-3196 treatment resulted in concentration-dependent decreases in total and low-density lipoprotein cholesterol with corresponding increases in liver gene expression, but the compound was significantly less potent than T3. In conclusion, we have implemented a strategy to rank the efficacy of THRß agonists by quantifying changes in the transcription of genes that lead to metabolic alterations, an effect that is directly downstream of THR binding and activation.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores beta de Hormona Tiroidea/agonistas , Transcripción Genética/efectos de los fármacos , Acetatos/farmacología , Acetatos/uso terapéutico , Proteína 4 Similar a la Angiopoyetina/metabolismo , Animales , Línea Celular Tumoral , LDL-Colesterol/sangre , LDL-Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Hepatocitos , Humanos , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Organofosfonatos/farmacología , Organofosfonatos/uso terapéutico , Fenoles/farmacología , Fenoles/uso terapéutico , Cultivo Primario de Células , Piridazinas/farmacología , Piridazinas/uso terapéutico , Ratas , Uracilo/análogos & derivados , Uracilo/farmacología , Uracilo/uso terapéutico
6.
Antiviral Res ; 182: 104924, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32896566

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the COVID-19 pandemic that began in 2019. The coronavirus 3-chymotrypsin-like cysteine protease (3CLpro) controls replication and is therefore considered a major target for antiviral discovery. This study describes the evaluation of SARS-CoV-2 3CLpro inhibitors in a novel self-assembled monolayer desorption ionization mass spectrometry (SAMDI-MS) enzymatic assay. Compared with a traditional FRET readout, the label-free SAMDI-MS assay offers greater sensitivity and eliminates false positive inhibition from compound interference with the optical signal. The SAMDI-MS assay was optimized and validated with known inhibitors of coronavirus 3CLpro such as GC376 (IC50 = 0.060 µM), calpain inhibitors II and XII (IC50 ~20-25 µM). The FDA-approved drugs shikonin, disulfiram, and ebselen did not inhibit SARS-CoV-2 3CLpro activity in the SAMDI-MS assay under physiologically relevant reducing conditions. The three drugs did not directly inhibit human ß-coronavirus OC-43 or SARS-CoV-2 in vitro, but instead induced cell death. In conclusion, the SAMDI-MS 3CLpro assay, combined with antiviral and cytotoxic assessment, provides a robust platform to evaluate antiviral agents directed against SARS-CoV-2.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Betacoronavirus/enzimología , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Proteínas no Estructurales Virales/antagonistas & inhibidores , COVID-19 , Proteasas 3C de Coronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Glicoproteínas/farmacología , Células HeLa , Humanos , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , SARS-CoV-2 , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Tratamiento Farmacológico de COVID-19
7.
J Med Chem ; 62(9): 4555-4570, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-30951311

RESUMEN

We report the synthesis and biological evaluation of a series of 4'-fluoro-2'- C-substituted uridines. Triphosphates of the uridine analogues exhibited a potent inhibition of hepatitis C virus (HCV) NS5B polymerase with IC50 values as low as 27 nM. In an HCV subgenomic replicon assay, the phosphoramidate prodrugs of these uridine analogues demonstrated a very potent activity with EC50 values as low as 20 nM. A lead compound AL-335 (53) demonstrated high levels of the nucleoside triphosphate in vitro in primary human hepatocytes and Huh-7 cells as well as in dog liver following a single oral dose. Compound 53 was selected for the clinical development where it showed promising results in phase 1 and 2 trials.


Asunto(s)
Alanina/análogos & derivados , Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Profármacos/farmacología , Nucleótidos de Uracilo/farmacología , Uridina/análogos & derivados , Alanina/síntesis química , Alanina/farmacología , Animales , Antivirales/síntesis química , Línea Celular Tumoral , Perros , Hepacivirus/enzimología , Hepatitis C/tratamiento farmacológico , Humanos , Inhibidores de la Síntesis del Ácido Nucleico/síntesis química , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforamidas , Profármacos/síntesis química , Replicón/efectos de los fármacos , Nucleótidos de Uracilo/síntesis química , Nucleótidos de Uracilo/metabolismo , Uridina/síntesis química , Uridina/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores
8.
Antiviral Res ; 143: 151-161, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28412183

RESUMEN

Recent cases of severe toxicity during clinical trials have been associated with antiviral ribonucleoside analogs (e.g. INX-08189 and balapiravir). Some have hypothesized that the active metabolites of toxic ribonucleoside analogs, the triphosphate forms, inadvertently target human mitochondrial RNA polymerase (POLRMT), thus inhibiting mitochondrial RNA transcription and protein synthesis. Others have proposed that the prodrug moiety released from the ribonucleoside analogs might instead cause toxicity. Here, we report the mitochondrial effects of several clinically relevant and structurally diverse ribonucleoside analogs including NITD-008, T-705 (favipiravir), R1479 (parent nucleoside of balapiravir), PSI-7851 (sofosbuvir), and INX-08189 (BMS-986094). We found that efficient substrates and chain terminators of POLRMT, such as the nucleoside triphosphate forms of R1479, NITD-008, and INX-08189, are likely to cause mitochondrial toxicity in cells, while weaker chain terminators and inhibitors of POLRMT such as T-705 ribonucleoside triphosphate do not elicit strong in vitro mitochondrial effects. Within a fixed 3'-deoxy or 2'-C-methyl ribose scaffold, changing the base moiety of nucleotides did not strongly affect their inhibition constant (Ki) against POLRMT. By swapping the nucleoside and prodrug moieties of PSI-7851 and INX-08189, we demonstrated that the cell-based toxicity of INX-08189 is mainly caused by the nucleoside component of the molecule. Taken together, these results show that diverse 2' or 4' mono-substituted ribonucleoside scaffolds cause mitochondrial toxicity. Given the unpredictable structure-activity relationship of this ribonucleoside liability, we propose a rapid and systematic in vitro screen combining cell-based and biochemical assays to identify the early potential for mitochondrial toxicity.


Asunto(s)
Antivirales/toxicidad , Mitocondrias/efectos de los fármacos , Ribonucleósidos/química , Ribonucleósidos/toxicidad , Adenosina/análogos & derivados , Amidas/toxicidad , Línea Celular/efectos de los fármacos , Citidina/análogos & derivados , Citidina/toxicidad , ARN Polimerasas Dirigidas por ADN/efectos de los fármacos , Guanosina Monofosfato/análogos & derivados , Guanosina Monofosfato/toxicidad , Humanos , Concentración 50 Inhibidora , Proteínas Mitocondriales/metabolismo , Nucleósidos/toxicidad , Profármacos/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Pirazinas/toxicidad , ARN/metabolismo , ARN Mitocondrial , Sofosbuvir/toxicidad , Relación Estructura-Actividad , Sitio de Iniciación de la Transcripción/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
9.
Bioorg Med Chem Lett ; 23(20): 5650-3, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23999044

RESUMEN

The discovery and development of antimicrobial agents that do not give rise to resistance remains an ongoing challenge. Our efforts in this regard continue to reveal new potential therapeutic agents with differing physicochemical properties while retaining the effective N,N-dichloroamine pharmacophore as the key antimicrobial warhead. In this Letter, we disclose agents containing polyol units as a water solubilizing group. These sulfonyl-polyol agents show broad spectrum bactericidal and virucidal activity. These compounds show 1 h MBC's of 16-512 µg/mL against Escherichia coli and 4-256 µg/mL against Staphylococcus aureus at neutral pH, and 1-h IC50's of 4.5-32 µM against Adenovirus 5 and 0.7-3.0 µM against Herpes simplex virus 1. The lead compounds were tested in a tissue culture irritancy assay and showed only minimal irritation at the highest concentrations tested.


Asunto(s)
Aminas/química , Antiinfecciosos/química , Polímeros/química , Adenoviridae/metabolismo , Aminas/síntesis química , Aminas/farmacología , Animales , Antiinfecciosos/síntesis química , Antiinfecciosos/farmacología , Línea Celular Tumoral , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Herpesvirus Humano 1/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus/efectos de los fármacos , Células Vero
10.
Invest Ophthalmol Vis Sci ; 54(2): 1244-51, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23341010

RESUMEN

PURPOSE: Viral conjunctivitis is a highly contagious infection often causing major epidemics. A safe broad-spectrum antiviral agent is needed to treat this unmet medical need. The purpose of this study is to demonstrate that in vitro NVC-422 is a safe, broad-spectrum topical virucidal agent with activity against ophthalmic viral pathogens. METHODS: The virucidal activity of NVC-422 against several serotypes of human adenovirus (HAdV), coxsackievirus A24, enterovirus 70, and herpes simplex-virus-1 (HSV-1) was tested in standard in vitro titer reduction assays with or without tears. An in vitro irritancy score for NVC-422 was determined using the MatTek EpiOcular tissue system. RESULTS: NVC-422 reduced the viral titer of HAdV-5, HAdV-8, HAdV-19, HAdV-37, and HSV-1 by at least 4 logs after 1 hour incubation at 250 µM. Incubation of coxsackievirus A24 and enterovirus 70 with 2.5 mM NVC-422 for 1 hour reduced the viral titer by 4 logs and 4.5 logs, respectively. The virucidal activity of NVC-422 is maintained in the presence of 10% synthetic tears. In the EpiOcular corneal tissue model, NVC-422 was nonirritating at concentrations up to 41 mM. CONCLUSIONS: NVC-422 has potent, rapid in vitro virucidal activity against major causes of conjunctivitis. Its broad-spectrum virucidal activity combined with favorable safety profile validates NVC-422 as a potential new therapeutic agent against viral conjunctivitis.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Conjuntiva/virología , Conjuntivitis Viral/tratamiento farmacológico , Taurina/análogos & derivados , Células Cultivadas , Conjuntiva/patología , Conjuntivitis Viral/patología , Conjuntivitis Viral/virología , Humanos , Taurina/farmacología
11.
Antiviral Res ; 92(3): 470-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22024427

RESUMEN

Human adenoviral conjunctivitis is a highly contagious eye infection affecting millions of people world-wide. If untreated, it can further develop into keratitis, corneal ulceration, scarring and possible blindness. Despite the significant patient morbidity and socio-economic costs, it is an unmet medical need with no FDA approved treatment. Here, we demonstrate the virucidal activity of NVC-422 (N,N-dichloro-2,2-dimethyltaurine) against adenovirus type 5 (Ad5) and investigated its mechanism of action of Ad5 inactivation. NVC-422 inhibits Ad5-induced loss of cell viability in vitro with 50% inhibitory concentration (IC(50)) ranging from 9 to 23 µM. NVC-422 does not cause any cytotoxicity at concentrations as high as 250 µM. Invitro, NVC-422 inactivates Ad5 but does not interfere with viral replication, indicating that NVC-422 acts on the extracellular adenovirus as a virucidal agent. NVC-422 inactivates Ad5 by oxidative inactivation of key viral proteins such as fiber and hexon as evidenced by SDS-PAGE, Western blotting and reversed-phase HPLC. These data, combined with measurements of the kinetics of the NVC-422 reactivity with selected amino acids, indicate that the changes in the viral proteins are caused by the selective oxidation of sulfur-containing amino acids. The conformational changes of the viral proteins result in the destruction of the viral morphology as shown by transmission electron microscopy. In summary, NVC-422 exhibits virucidal activity against Ad5 by the oxidative inactivation of key viral proteins, leading to the loss of viral integrity and infectivity.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Taurina/análogos & derivados , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Adenovirus Humanos/ultraestructura , Antivirales/química , Antivirales/uso terapéutico , Línea Celular , Conjuntivitis Viral/tratamiento farmacológico , Cisteína/química , Humanos , Metionina/química , Oxidación-Reducción , Compuestos de Sulfhidrilo/química , Taurina/química , Taurina/farmacología , Taurina/uso terapéutico , Proteínas Virales/química , Inactivación de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
12.
Antiviral Res ; 87(2): 213-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20470829

RESUMEN

Dengue virus (DENV), an emerging pathogen from the Flaviviridae family with neither vaccine nor antiviral treatment available, causes a serious worldwide public health threat. In theory, there are several ways by which small molecules could inhibit the replication cycle of DENV. Here, we show that the nucleoside analogue beta-d-2'-ethynyl-7-deaza-adenosine inhibits representative strains of all four serotypes of DENV with an EC(50) around or below 1microM. Using membrane-associated native replicase complex as well as recombinant RNA polymerase from each DENV serotype in enzymatic assays, we provide evidence that beta-d-2'-ethynyl-7-deaza-adenosine triphosphate (2'E-7D-ATP) targets viral replication at the polymerase active site by competing with the natural nucleotide substrate with an apparent K(i) of 0.060+/-0.016microM. In single-nucleotide incorporation experiments, the catalytic efficiency of 2'E-7D-ATP is 10-fold lower than for natural ATP, and the incorporated nucleotide analogue causes immediate chain termination. A combination of bioinformatics and site-directed mutagenesis demonstrates that 2'E-7D-ATP is equipotent across all serotypes because the nucleotide binding site residues are conserved in dengue virus. Overall, beta-d-2'-ethynyl-7-deaza-adenosine provides a promising scaffold for the development of inhibitors of dengue virus polymerase.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Antivirales/farmacología , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Virus del Dengue/enzimología , Inhibidores Enzimáticos/farmacología , Animales , Antivirales/química , Sitios de Unión , Línea Celular , Biología Computacional , Secuencia Conservada , Cricetinae , Inhibidores Enzimáticos/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Mutagénesis Sitio-Dirigida
14.
Antimicrob Agents Chemother ; 54(2): 734-41, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19995923

RESUMEN

In passaging experiments, we isolated HIV strains resistant to MAb3952, a chemokine (C-C motif) receptor 5 (CCR5) monoclonal antibody (MAb) that binds to the second extracellular domain (extracellular loop 2 [ECL-2]) of CCR5. MAb3952-resistant viruses remain CCR5-tropic and are cross-resistant to a second ECL-2-specific antibody. Surprisingly, MAb3952-resistant viruses were more susceptible to RoAb13, a CCR5 antibody binding to the N terminus of CCR5. Using CCR5 receptor mutants, we show that MAb3952-resistant virus strains preferentially use the N terminus of CCR5, while the wild-type viruses preferentially use ECL-2. We propose this switch in the CCR5 binding site as a novel mechanism of HIV resistance.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Epítopos/inmunología , Infecciones por VIH/tratamiento farmacológico , VIH/efectos de los fármacos , Receptores CCR5/inmunología , Fármacos Anti-VIH/farmacología , Sitios de Unión/genética , Línea Celular Tumoral , Células Cultivadas , Farmacorresistencia Viral/genética , Epítopos/química , Epítopos/genética , VIH/genética , Infecciones por VIH/virología , Humanos , Receptores CCR5/química , Receptores CCR5/genética
16.
J Biol Chem ; 284(8): 5175-85, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19097993

RESUMEN

In this study, we describe a novel CD4-targeting bifunctional human immunodeficiency virus (HIV-1) fusion inhibitor (CD4-BFFI) that blocks HIV-1 entry by inhibiting both HIV-1 attachment and fusion and is highly potent against both R5 and X4 HIV-1 viruses in various antiviral assays, including peripheral blood mononuclear cell (PBMC) infection assays. Previously, we have reported a CCR5 antibody-based bifunctional HIV-1 fusion inhibitor (BFFI) that was highly active in blocking R5 HIV-1 infection but was ineffective against X4 viruses infecting human PBMCs (Kopetzki, E., Jekle, A., Ji, C., Rao, E., Zhang, J., Fischer, S., Cammack, N., Sankuratri, S., and Heilek, G. (2008) Virology J. 5, 56-65). CD4-BFFI, which consists of two HIV-1 fusion inhibitor (FI) T-651 variant peptides recombinantly fused to the Fc end of a humanized anti-CD4 monoclonal antibody, has demonstrated more than 100-fold greater antiviral activity than T-651 variant or the parental CD4 monoclonal antibody. Mechanistic studies revealed that CD4-BFFI primarily blocks the HIV-1-cell fusion step through its FI peptide moieties. The enhanced antiviral activity of CD4-BFFI is most likely due to avid binding of the bivalent FI peptides as well as the increased local concentration of CD4-BFFI via attachment to the target cell surface receptor CD4. In vivo pharmacokinetic studies demonstrated that CD4-BFFI was stable in monkey blood, and a dose of 10 mg/kg maintained serum concentrations greater than 2,000-fold over the IC(90) value for 7 days postdosing. This novel bifunctional inhibitor with improved potency and favorable pharmacokinetic properties may offer a novel approach for HIV-1 therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD4 , Inhibidores de Fusión de VIH/farmacología , VIH-1/inmunología , Fragmentos Fc de Inmunoglobulinas/farmacología , Leucocitos Mononucleares/metabolismo , Péptidos/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacocinética , Antagonistas de los Receptores CCR5 , Células CHO , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Inhibidores de Fusión de VIH/farmacocinética , Humanos , Leucocitos Mononucleares/virología , Macaca fascicularis , Péptidos/farmacocinética , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología
17.
Antiviral Res ; 74(2): 125-37, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17166600

RESUMEN

To identify monoclonal antibodies (mAbs) with high potency and novel recognition sites, more than 25,000 of mouse hybridomas were screened and 4 novel anti-human CCR5 mAbs ROAb12, ROAb13, ROAb14, and ROAb18 showing potent activity in cell-cell fusion (CCF) assay were identified. These mAbs demonstrated potent antiviral activities in both single-cycle HIV infection (IC(50) range: 0.16-4.3 microg/ml) and PBMC viral replication (IC(50) range: 0.02-0.04 microg/ml) assays. These potent antiviral effects were donor-independent. All 4 mAbs were also highly potent in the PhenoSense assay against 29 HIV isolates covering clade A through G. In all antiviral assays, these mAbs showed potency superior to the previously reported mAb 2D7 in side-by-side comparison studies. All 4 mAbs were also fully active against viruses resistant to HIV fusion inhibitor enfuvirtide and CCR5 antagonist maraviroc. Although ROAb12, ROAb14, and ROAb18 inhibited RANTES, MIP1alpha and MIP1beta binding and cell activation, the other novel mAb ROAb13 was inactive in inhibiting cell activation by these three ligands. Furthermore, highly synergistic antiviral effects were found between mAb ROAb13 and 2D7 or ROAb12. In addition, none of these mAbs showed agonist activity or caused internalization of the CCR5 receptor.


Asunto(s)
Fármacos Anti-VIH/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , VIH/efectos de los fármacos , VIH/fisiología , Receptores CCR5/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antagonistas de los Receptores CCR5 , Fusión Celular , Línea Celular , Quimiocina CCL5/metabolismo , Cricetinae , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Receptores CCR5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA