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










Base de datos
Intervalo de año de publicación
1.
Arthritis Rheumatol ; 73(5): 769-778, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33169522

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) is a major autoimmune disease that causes synovitis and joint damage. Although clinical trials have been performed using interleukin-10 (IL-10), an antiinflammatory cytokine, as a potential treatment of RA, the therapeutic effects of IL-10 have been limited, potentially due to insufficient residence in lymphoid organs, where antigen recognition primarily occurs. This study was undertaken to engineer an IL-10-serum albumin (SA) fusion protein and evaluate its effects in 2 murine models of RA. METHODS: SA-fused IL-10 (SA-IL-10) was recombinantly expressed. Mice with collagen antibody-induced arthritis (n = 4-7 per group) or collagen-induced arthritis (n = 9-15 per group) were injected intravenously with wild-type IL-10 or SA-IL-10, and the retention of SA-IL-10 in the lymph nodes (LNs), immune cell composition in the paws, and therapeutic effect of SA-IL-10 on mice with arthritis were assessed. RESULTS: SA fusion to IL-10 led to enhanced accumulation in the mouse LNs compared with unmodified IL-10. Intravenous SA-IL-10 treatment restored immune cell composition in the paws to a normal status, elevated the frequency of suppressive alternatively activated macrophages, reduced IL-17A levels in the paw-draining LN, and protected joint morphology. Intravenous SA-IL-10 treatment showed similar efficacy as treatment with an anti-tumor necrosis factor antibody. SA-IL-10 was equally effective when administered intravenously, locally, or subcutaneously, which is a benefit for clinical translation of this molecule. CONCLUSION: SA fusion to IL-10 is a simple but effective engineering strategy for RA therapy and has potential for clinical translation.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Articulaciones del Pie/efectos de los fármacos , Interleucina-10/farmacología , Ganglios Linfáticos/inmunología , Macrófagos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/farmacología , Animales , Células Presentadoras de Antígenos/metabolismo , Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Pie , Articulaciones del Pie/inmunología , Articulaciones del Pie/metabolismo , Articulaciones del Pie/patología , Miembro Posterior , Antígenos de Histocompatibilidad Clase I/metabolismo , Inyecciones Intravenosas , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-6/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Ingeniería de Proteínas , Transporte de Proteínas , Receptores Fc/metabolismo , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/inmunología , Inhibidores del Factor de Necrosis Tumoral/farmacología
2.
Nat Biomed Eng ; 5(5): 387-398, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33046864

RESUMEN

Interleukin-4 (IL-4) suppresses the development of multiple sclerosis in a murine model of experimental autoimmune encephalomyelitis (EAE). Here, we show that, in mice with EAE, the accumulation and persistence in the lymph nodes and spleen of a systemically administered serum albumin (SA)-IL-4 fusion protein leads to higher efficacy in preventing disease development than the administration of wild-type IL-4 or of the clinically approved drug fingolimod. We also show that the SA-IL-4 fusion protein prevents immune-cell infiltration in the spinal cord, decreases integrin expression in antigen-specific CD4+ T cells, increases the number of granulocyte-like myeloid-derived suppressor cells (and their expression of programmed-death-ligand-1) in spinal cord-draining lymph nodes and decreases the number of T helper 17 cells, a pathogenic cell population in EAE. In mice with chronic EAE, SA-IL-4 inhibits immune-cell infiltration into the spinal cord and completely abrogates immune responses to myelin antigen in the spleen. The SA-IL-4 fusion protein may be prophylactically and therapeutically advantageous in the treatment of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/administración & dosificación , Interleucina-4/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Albúmina Sérica/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Semivida , Inmunosupresores/farmacocinética , Inmunosupresores/farmacología , Inyecciones Intravenosas , Ganglios Linfáticos/química , Ganglios Linfáticos/inmunología , Ratones , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Bazo/química , Bazo/inmunología , Células Th17/efectos de los fármacos
4.
Arthritis Res Ther ; 21(1): 298, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31870429

RESUMEN

BACKGROUND: Although disease in a majority of rheumatoid arthritis (RA) patients is often initially limited to one or a few joints, currently approved medications including anti-tumor necrosis factor-α antibody (α-TNF) are injected systemically. Given that α-TNF systemic injection typically does not cure RA and involves risk of treatment-related adverse events, one possible approach to enhance therapeutic efficacy and reduce α-TNF systemic exposure is to retain the antibodies in arthritic joints after local administration. The aim of this study was to evaluate the approach of conferring extracellular matrix (ECM) binding affinity to α-TNF antibodies in a RA model. METHODS: α-TNF was chemically conjugated with a promiscuous ECM-binding peptide derived from placenta growth factor 2 (PlGF-2123-144). The binding activity of PlGF-2123-144-conjugated α-TNF (PlGF-2123-144-α-TNF) against ECM proteins was assessed by ELISA and by immunostaining on human cartilage specimens. The effect of conjugation on antibody function was assessed as a neutralizing activity against osteoclast differentiation. Retention at the injection site and therapeutic efficacy of PlGF-2123-144-α-TNF were tested in a collagen antibody-induced arthritis (CAIA) model in the mouse. RESULTS: PlGF-2123-144 peptide conjugation conferred α-TNF with affinity to ECM proteins without impairment of antigen recognition. PlGF-2123-144-α-TNF locally injected at a paw in the CAIA model was retained for at least 96 h at the injection site, whereas unmodified α-TNF was dispersed rapidly after injection. Local treatment with unmodified α-TNF did not suppress the arthritis score relative to isotype controls. By contrast, local administration of PlGF-2123-144-α-TNF suppressed arthritis development almost completely in the treated paw even at a 1000× lower dose. CONCLUSION: These data demonstrate that retention of α-TNF in arthritic joints can suppress arthritis development and enhance therapeutic efficacy. This simple bioengineering approach of ECM-binding peptide conjugation offers a powerful and clinically translational approach to treat RA.


Asunto(s)
Anticuerpos/inmunología , Artritis Reumatoide/inmunología , Matriz Extracelular/inmunología , Inmunoconjugados/inmunología , Factor de Crecimiento Placentario/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/prevención & control , Artritis Reumatoide/metabolismo , Artritis Reumatoide/prevención & control , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Humanos , Inmunoconjugados/metabolismo , Inmunoconjugados/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Factor de Crecimiento Placentario/química , Factor de Crecimiento Placentario/metabolismo , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo
5.
Sci Adv ; 5(11): eaay1971, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31723606

RESUMEN

Enhancing the therapeutic efficacy of drugs for inflammatory diseases is of high demand. One possible approach is targeting drugs to the extracellular matrix of the inflamed area. Here, we target collagens in the matrix, which are inaccessible in most tissues yet are exposed to the bloodstream in the inflamed area because of vascular hyperpermeability. We conferred collagen affinity to anti-tumor necrosis factor-α (α-TNF) antibody by conjugating a collagen-binding peptide (CBP) derived from the sequence of decorin. CBP-α-TNF accumulated in the inflamed paw of the arthritis model, and arthritis development was significantly suppressed by treatment with CBP-α-TNF compared with the unmodified antibody. Similarly, CBP-anti-transforming growth factor-ß (α-TGF-ß) accumulated in the inflamed lung of pulmonary fibrosis model and significantly suppressed pulmonary fibrosis compared with the unmodified antibody. Together, collagen affinity enables the anticytokine antibodies to target arthritis and pulmonary fibrosis accompanied by inflammation, demonstrating a clinically translational approach to treat inflammatory diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticuerpos/uso terapéutico , Colágeno/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Antiinflamatorios/inmunología , Antiinflamatorios/metabolismo , Anticuerpos/inmunología , Anticuerpos/metabolismo , Colágeno/inmunología , Colágeno/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Sialoglicoproteínas/inmunología , Sialoglicoproteínas/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-31405853

RESUMEN

Current therapies against invasive pulmonary aspergillosis (IPA) have a limited cure rate. Given that a delay in treatment initiation may be fatal, a new drug with rapid-onset and potent fungicidal activity is needed. The novel cyclic hexapeptide ASP2397 (currently known as VL-2397) exhibited antifungal activity against Aspergillus fumigatus (including azole-sensitive and azole-resistant isolates), A. terreus, and A. flavus at an MIC range of 1 to 4 µg/ml in human serum. Time-kill curve experiments showed that ASP2397 reduced germinated conidia of A. fumigatus by more than 1 log10 CFU within 6 h. In addition, ASP2397 inhibited hyphal elongation from germinated conidia of A. fumigatus, A. terreus, and A. flavus more rapidly than voriconazole. Under conditions of delayed treatment initiation in an IPA mouse model, ASP2397 had efficacy superior to that of posaconazole, with 100% survival and over 1 log10 CFU/g reduction in lung fungal burden. Histopathological investigation of lungs also showed that ASP2397 markedly suppressed disease progression. To clarify its mechanism of action, we generated a UV-induced mutant of A. fumigatus with low susceptibility to ASP2397. The mutant had a point mutation in the siderophore transporter gene sit1, which is absent in mammalian cells. These findings suggest that ASP2397 may improve clinical treatment options for IPA.


Asunto(s)
Antifúngicos/farmacología , Aspergillus fumigatus/efectos de los fármacos , Aspergillus fumigatus/metabolismo , Aspergillus/efectos de los fármacos , Aspergillus/metabolismo , Complejos de Coordinación/farmacología , Proteínas Fúngicas/metabolismo , Péptidos Cíclicos/farmacología , Farmacorresistencia Fúngica/genética , Proteínas Fúngicas/genética , Humanos , Pruebas de Sensibilidad Microbiana , Sideróforos/química
7.
Sci Transl Med ; 11(487)2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971453

RESUMEN

Cancer immunotherapy with immune checkpoint inhibitors (CPIs) and interleukin-2 (IL-2) has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T lymphocyte antigen 4 antibody (αCTLA4) + anti-programmed death ligand 1 antibody (αPD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously administered CBD protein accumulated mainly in tumors. CBD conjugation or fusion decreases the systemic toxicity of both αCTLA4 + αPD-L1 combination therapy and IL-2, for example, eliminating hepatotoxicity with the CPI molecules and ameliorating pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells. In an orthotopic breast cancer model, combination treatment with CPI and IL-2 eradicated tumors in 9 of 13 animals with the CBD-modified drugs, whereas it did so in only 1 of 13 animals with the unmodified drugs. Thus, the A3 domain of VWF can be used to improve safety and efficacy of systemically administered tumor drugs with high translational promise.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Colágeno/metabolismo , Citocinas/inmunología , Inmunoterapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Inmunidad , Inyecciones Intravenosas , Interleucina-2/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Dominios Proteicos , Resultado del Tratamiento
8.
Biochem Pharmacol ; 158: 201-206, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30365949

RESUMEN

Existing treatments have limited efficacy against severe infection associated with herpes simplex virus (HSV) and herpes zoster virus (VZV), particularly in immunocompromized patients and those with multidermatomal infection. This issue, along with issues regarding drug resistance, support the need for improved therapeutic options. To investigate the antiviral effect of amenamevir, a VZV and HSV helicase-primase inhibitor, in severe infection conditions, mouse models of severe HSV-1 infection were developed by immunosuppression or multidermatomal infection. Mice with cyclosporin-induced immunosuppression and HSV-1 infection via inoculation of a dorsolateral area of skin were orally treated with amenamevir (10-100 mg/kg/day) for different durations (2-5 days). Immunosuppressed mice maintained high skin HSV-1 titers in the absence of treatment. Amenamevir successfully reduced HSV-1 titers at all tested doses in immunosuppressed mice, but required a longer treatment period to avoid a rebound in viral titers due to immunosuppression. To compare the efficacy of amenamevir and valacyclovir, a murine model of multidermatomal HSV-1 infection was generated by scarifying the dorsolateral area of skin in a line and inoculating broadly with HSV-1. The mice were treated with amenamevir or valacyclovir starting on Day 3, 4, or 5 post-infection for 5 days. Although both drugs similarly reduced disease scores when treatment was started on Day 3, amenamevir also reduced disease severity when treatment was initiated on Day 4, whereas valacyclovir did not. Amenamevir was not affected by the host's immune status in terms of effective oral doses and was more efficacious in treating severe cutaneous infection even when treatment initiation was delayed.


Asunto(s)
Antivirales/uso terapéutico , ADN Helicasas/antagonistas & inhibidores , ADN Primasa/antagonistas & inhibidores , Modelos Animales de Enfermedad , Oxadiazoles/uso terapéutico , Índice de Severidad de la Enfermedad , Infección por el Virus de la Varicela-Zóster/tratamiento farmacológico , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/farmacología , Chlorocebus aethiops , ADN Helicasas/metabolismo , ADN Primasa/metabolismo , Femenino , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/metabolismo , Ratones , Oxadiazoles/farmacología , Resultado del Tratamiento , Infección por el Virus de la Varicela-Zóster/metabolismo , Células Vero , Proteínas Virales/metabolismo
9.
J Infect Chemother ; 24(6): 492-495, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29373265

RESUMEN

The narrow-spectrum macrocyclic antibiotic fidaxomicin is approved for treatment of Clostridium difficile infection in many countries and is currently under evaluation in Japan for this indication. This study was conducted to evaluate the effects of fidaxomicin and its major metabolite, OP-1118, on Clostridium spp. isolated in Nagasaki University Hospital, Japan. Isolates were cultured and antimicrobial susceptibility analyses performed according to the Clinical Laboratory Standards Institute methods. Ninety-eight isolates were obtained between 2012 and 2015, 50 of C. difficile and 48 of eight other Clostridium spp. Fidaxomicin had the lowest minimum inhibitory concentration (MIC) of the antimicrobials tested against C. difficile, with MIC90 (MIC range) 0.12 µg/mL (0.015-0.25), versus vancomycin MIC90 0.5 µg/mL (0.5), metronidazole MIC90 0.5 µg/mL (0.12-0.5), and OP-1118 MIC90 4.0 µg/mL (0.5-4.0). Fidaxomicin and OP-1118 each had a similar spectrum of activity against the other Clostridium spp. C. butyricum and the 29 fidaxomicin- and OP-1118-susceptible C. perfringens isolates had the lowest MIC values, and C. bolteae and C. hathewayi higher. All the C. ramosum isolates (n = 6) and one of 30 C. perfringens isolates had low susceptibility to fidaxomicin and OP-1118 (i.e., MIC >64 µg/mL). In summary, this study showed that fidaxomicin was active against a number of Clostridium spp., including C. difficile. Fidaxomicin was generally more effective than its major metabolite OP-1118, but both showed a similar spectrum of activity, suggesting that OP-1118 contributes to the antimicrobial activity of fidaxomicin. These findings were broadly in accordance with those of similar studies conducted in other settings.


Asunto(s)
Aminoglicósidos/farmacología , Antiinfecciosos/farmacología , Clostridium/efectos de los fármacos , Aminoglicósidos/uso terapéutico , Antiinfecciosos/uso terapéutico , Clostridium/clasificación , Clostridium/aislamiento & purificación , Infecciones por Clostridium/tratamiento farmacológico , Fidaxomicina , Humanos , Japón , Pruebas de Sensibilidad Microbiana
10.
J Infect Chemother ; 23(10): 724-726, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28527649

RESUMEN

We evaluated the susceptibility of 100 Japanese Clostridium difficile isolates to fidaxomicin, a new macrocyclic antibiotic. The minimum inhibitory concentration (MIC) range of fidaxomicin was 0.03-0.5 µg/mL, with a MIC for inhibition of 50% (MIC50) of 0.12 µg/mL, and for inhibition of 90% (MIC90) of 0.25 µg/mL. We also evaluated the susceptibilities of the same 100 C. difficile isolates to vancomycin, metronidazole, moxifloxacin, clindamycin, meropenem, and ampicillin. Of all the antibiotics tested, fidaxomicin showed the most potent antimicrobial activity against this group of C. difficile isolates. MIC levels against C. difficile isolates, including those producing binary toxin, did not substantially differ from those previously reported in Europe, North America and Taiwan.


Asunto(s)
Aminoglicósidos/farmacología , Antiinfecciosos/farmacología , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , Europa (Continente) , Fidaxomicina , Humanos , Japón , Pruebas de Sensibilidad Microbiana/métodos , América del Norte , Taiwán
11.
Drug Metab Pharmacokinet ; 31(4): 323-32, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27461507

RESUMEN

Amenamevir is a novel drug that targets the viral helicase-primase complex. While dose-dependent efficacy had been observed in non-clinical studies, no clear dose dependence has been observed in humans. We therefore developed a pharmacokinetic/pharmacodynamic (PK/PD) model to explain this inconsistency between species and to clarify the immune-related healing of amenamevir in humans. The model consisted of a non-linear kinetic model for a virtual number of virus plaques as a built-in biomarker. Lesion score was defined as an endpoint of antiviral efficacy, and logit model analysis was applied to the ordered-categorical lesion score. The modeling results suggested the time course profiles of lesion score could be explained with the efficacy terms in the logit model, using change in number of virus plaques as an indicator of the effects of amenamevir and time elapsed as an indicator of the healing of the immune response. In humans, the PD effect was almost dose-independent, and immune-related healing may have been the driving force behind the reduction in lesion scores. Drug efficacy is occasionally masked in diseases healed by the immune response, such as genital herpes. The PK/PD model proposed in the present study must be useful for explanation the PK/PD relationship of such drugs.


Asunto(s)
Antivirales/farmacocinética , Herpes Genital/tratamiento farmacológico , Oxadiazoles/farmacocinética , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Células Cultivadas , Chlorocebus aethiops , Femenino , Cobayas , Herpes Genital/metabolismo , Humanos , Modelos Biológicos , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Recurrencia , Simplexvirus/efectos de los fármacos , Células Vero
12.
Antimicrob Agents Chemother ; 57(3): 1339-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23274658

RESUMEN

ASP2151 (amenamevir) is a helicase-primase inhibitor against herpes simplex virus 1 (HSV-1), HSV-2, and varicella zoster virus. Here, to determine and analyze the correlation between the pharmacodynamic (PD) and pharmacokinetic (PK) parameters of ASP2151, we examined the PD profile of ASP2151 using in vitro plaque reduction assay and a murine model of HSV-1 infection. ASP2151 inhibited the in vitro replication of HSV-1 with a mean 50% effective concentration (EC(50)) of 14 ng/ml. In the cutaneously HSV-1-infected mouse model, ASP2151 dose dependently suppressed intradermal HSV-1 growth, with the effect reaching a plateau at a dose of 30 mg/kg of body weight/day. The dose fractionation study showed that intradermal HSV-1 titers were below the detection limit in mice treated with ASP2151 at 100 mg/kg/day divided into two daily doses and at 30 or 100 mg/kg/day divided into three daily doses. The intradermal HSV-1 titer correlated with the maximum concentration of drug in serum (C(max)), the area under the concentration-time curve over 24 h (AUC(24h)), and the time during which the concentration of ASP2151 in plasma was above 100 ng/ml (T(>100)). The continuous infusion of ASP2151 effectively decreased intradermal HSV-1 titers below the limit of detection in mice in which the ASP2151 concentration in plasma reached 79 to 145 ng/ml. Our findings suggest that the antiviral efficacy of ASP2151 is most closely associated with the PK parameter T(>100) in HSV-1-infected mice. Based on these results, we propose that a plasma ASP2151 concentration exceeding 100 ng/ml for 21 to 24 h per day provides the maximum efficacy in HSV-1-infected mice.


Asunto(s)
Antivirales/farmacocinética , ADN Helicasas/antagonistas & inhibidores , ADN Primasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacocinética , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1/efectos de los fármacos , Oxadiazoles/farmacocinética , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/sangre , Antivirales/farmacología , Área Bajo la Curva , ADN Helicasas/metabolismo , ADN Primasa/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacología , Femenino , Herpes Simple/virología , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/crecimiento & desarrollo , Ratones , Ratones Pelados , Oxadiazoles/sangre , Oxadiazoles/farmacología , Piel/efectos de los fármacos , Piel/virología , Ensayo de Placa Viral , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
13.
Antiviral Res ; 97(2): 154-60, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23261844

RESUMEN

ASP2151 (amenamevir) is a helicase-primase complex inhibitor with antiviral activity against herpes simplex virus HSV-1, HSV-2, and varicella-zoster virus (VZV). To assess combination therapy of ASP2151 with existing antiherpes agents against HSV-1, HSV-2, and VZV, we conducted in vitro and in vivo studies of two-drug combinations. The combination activity effect of ASP2151 with nucleoside analogs acyclovir (ACV), penciclovir (PCV), or vidarabine (VDB) was tested via plaque-reduction assay and MTS assay, and the data were analyzed using isobolograms and response surface modeling. In vivo combination therapy of ASP2151 with valaciclovir (VACV) was studied in an HSV-1-infected zosteriform spread mouse model. The antiviral activity of ASP2151 combined with ACV and PCV against ACV-susceptible HSV-1, HSV-2, and VZV showed a statistically significant synergistic effect (P<0.05). ASP2151 with VDB was observed to have additive effects against ACV-susceptible HSV-2 and synergistic effects against VZV. In the mouse model of zosteriform spread, the inhibition of disease progression via combination therapy was more potent than that of either drugs as monotherapy (P<0.05). These results indicate that the combination therapies of ASP2151 with ACV and PCV have synergistic antiherpes effects against HSV and VZV infections and may be feasible in case of severe disease, such as herpes encephalitis or in patients with immunosuppression.


Asunto(s)
Sinergismo Farmacológico , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 3/efectos de los fármacos , Nucleósidos/farmacología , Oxadiazoles/farmacología , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Herpes Zóster/tratamiento farmacológico , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Resultado del Tratamiento , Células Vero , Ensayo de Placa Viral
14.
Biochem Pharmacol ; 84(4): 459-67, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22687623

RESUMEN

ASP2151 is an antiherpes agent targeting the helicase-primase complex of herpes simplex virus (HSV)-1, HSV-2, and varicella-zoster virus (VZV). We characterized the ASP2151-resistant HSV-1 and HSV-2 variants or mutants based on findings from sequencing analysis, growth, pathogenicity, and susceptibility testing, identifying several single base-pair substitutions resulting in amino acid changes in the helicase and primase subunit of ASP2151-resistant mutants. Amino acid alterations in the helicase subunit were clustered near helicase motif IV in the UL5 helicase gene of both HSV-1 and HSV-2, while the primase subunit substitution associated with reduced susceptibility, R367H, was found in ASP2151-resistant HSV-1 mutants. However, while susceptibility in the ASP2151-resistant HSV mutants to existing antiherpes agents was equivalent to that in wild-type HSV strains, ASP2151-resistant HSV mutants showed attenuated in vitro growth capability and in vivo pathogenicity compared with the parent strains. Taken together, our present findings demonstrated that important amino acid substitutions associated with reduced susceptibilities of HSV-1 and HSV-2 to ASP2151 exist in both the helicase and primase subunits of the helicase-primase complex, and that mutations in this complex against ASP2151 might confer defects in viral replication and pathogenicity.


Asunto(s)
Antivirales/farmacología , ADN Helicasas/antagonistas & inhibidores , ADN Primasa/antagonistas & inhibidores , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Oxadiazoles/farmacología , Proteínas Virales/antagonistas & inhibidores , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células Cultivadas , Chlorocebus aethiops , ADN Helicasas/genética , ADN Primasa/genética , Farmacorresistencia Viral , Femenino , Herpes Simple/virología , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 2/enzimología , Herpesvirus Humano 2/genética , Humanos , Ratones , Ratones Pelados , Datos de Secuencia Molecular , Mutación , Ensayo de Placa Viral , Proteínas Virales/genética
15.
Antimicrob Agents Chemother ; 56(7): 3587-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22526302

RESUMEN

ASP2151 (amenamevir) is a helicase-primase inhibitor against herpes simplex virus type 1 (HSV-1), HSV-2, and varicella-zoster virus. To evaluate the anti-HSV activity of ASP2151, susceptibility testing was performed on viruses isolated from patients participating in a placebo- and valacyclovir-controlled proof-of-concept phase II study for recurrent genital herpes. A total of 156 HSV strains were isolated prior to the dosing of patients, and no preexisting variants with less susceptibility to ASP2151 or acyclovir (ACV) were detected. ASP2151 inhibited HSV-1 and HSV-2 replication with mean 50% effective concentrations (EC(50)s) of 0.043 and 0.069 µM, whereas ACV exhibited mean EC(50)s of 2.1 and 3.2 µM, respectively. Notably, the susceptibilities of HSV isolates to ASP2151 and ACV were not altered after dosing with the antiviral agents. Taken together, these results demonstrate that ASP2151 inhibits the replication of HSV clinical isolates more potently than ACV, and HSV resistant to this novel helicase-primase inhibitor as well as ACV may not easily emerge in short-term treatment for recurrent genital herpes patients.


Asunto(s)
Antivirales/farmacología , ADN Helicasas/antagonistas & inhibidores , ADN Primasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Herpes Genital/virología , Oxadiazoles/farmacología , Simplexvirus/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Aciclovir/farmacología , Humanos , Simplexvirus/enzimología
16.
Molecules ; 16(9): 7210-23, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21869749

RESUMEN

ASP2151 is a herpesvirus helicase-primase inhibitor with antiviral activity against varicella zoster virus and herpes simplex virus types 1 (HSV-1) and 2 (HSV-2). Here, we examined the potency and efficacy of ASP2151 against HSV in vitro and in vivo. We found that ASP2151 was more potent in inhibiting the replication of HSV-1 and HSV-2 in Vero cells in the plaque reduction assay and had greater anti-HSV activity in a guinea pig model of genital herpes than did acyclovir and valacyclovir (VACV), respectively. Oral ASP2151 given from the day of infection reduced peak and overall disease scores in a dose-dependent manner, resulting in complete prevention of symptoms at the dose of 30 mg/kg. The 50% effective dose (ED(50)) values for ASP2151 and VACV were 0.37 and 68 mg/kg, respectively, indicating that ASP2151 was 184-fold more potent than VACV. When ASP2151 was administered after the onset of symptoms, the disease course of genital herpes was suppressed more effectively than by VACV, with a significant reduction in disease score observed one day after starting ASP2151 at 30 mg/kg, whereas the therapeutic effect of VACV was only evident three days after treatment at the highest dose tested (300 mg/kg). This indicated that ASP2151 possesses a faster onset of action and wider therapeutic time window than VACV. Further, virus shedding from the genital mucosa was significantly reduced with ASP2151 at 10 and 30 mg/kg but not with VACV, even at 300 mg/kg. Taken together, our present findings demonstrated the superior potency and efficacy of ASP2151 against HSV.


Asunto(s)
Antivirales/farmacología , ADN Helicasas/antagonistas & inhibidores , ADN Primasa/antagonistas & inhibidores , Herpes Genital/tratamiento farmacológico , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 2/enzimología , Oxadiazoles/farmacología , Proteínas Virales/antagonistas & inhibidores , Aciclovir/análogos & derivados , Aciclovir/farmacología , Animales , Antivirales/uso terapéutico , Área Bajo la Curva , Evaluación Preclínica de Medicamentos , Femenino , Cobayas , Herpes Genital/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 2/crecimiento & desarrollo , Oxadiazoles/uso terapéutico , Valaciclovir , Valina/análogos & derivados , Valina/farmacología , Carga Viral/efectos de los fármacos , Ensayo de Placa Viral , Esparcimiento de Virus/efectos de los fármacos
17.
J Antimicrob Chemother ; 65(8): 1733-41, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534624

RESUMEN

OBJECTIVES: To evaluate and describe the anti-herpesvirus effect of ASP2151, amenamevir, a novel non-nucleoside oxadiazolylphenyl-containing herpesvirus helicase-primase complex inhibitor. METHODS: The inhibitory effect of ASP2151 on enzymatic activities associated with a recombinant HSV-1 helicase-primase complex was assessed. To investigate the effect on viral DNA replication, we analysed viral DNA in cells infected with herpesviruses [herpes simplex virus (HSV), varicella-zoster virus (VZV) and human cytomegalovirus]. Sequencing analyses were conducted on an ASP2151-resistant VZV mutant. In vitro and in vivo antiviral activities were evaluated using a plaque reduction assay and an HSV-1-infected zosteriform-spread model in mice. RESULTS: ASP2151 inhibited the single-stranded DNA-dependent ATPase, helicase and primase activities associated with the HSV-1 helicase-primase complex. Antiviral assays revealed that ASP2151, unlike other known HSV helicase-primase inhibitors, exerts equipotent activity against VZV, HSV-1 and HSV-2 through prevention of viral DNA replication. Further, the anti-VZV activity of ASP2151 (EC(50), 0.038-0.10 microM) was more potent against all strains tested than that of aciclovir (EC(50), 1.3-27 microM). ASP2151 was also active against aciclovir-resistant VZV. Amino acid substitutions were found in helicase and primase subunits of ASP2151-resistant VZV. In a mouse zosteriform-spread model, ASP2151 was orally active and inhibited disease progression more potently than valaciclovir. CONCLUSIONS: ASP2151 is a novel herpes helicase-primase inhibitor that warrants further investigation for the potential treatment of both VZV and HSV infections.


Asunto(s)
Antivirales/farmacología , ADN Helicasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 3/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Citomegalovirus/efectos de los fármacos , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Farmacorresistencia Viral , Inhibidores Enzimáticos/uso terapéutico , Femenino , Herpes Zóster/tratamiento farmacológico , Ratones , Pruebas de Sensibilidad Microbiana , Análisis de Secuencia de ADN , Ensayo de Placa Viral , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...