Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
J Med Virol ; 96(1): e29379, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38235617

RESUMEN

Although neonates are commonly exposed to vaginal herpes simplex virus (HSV)-2, neonatal herpes is rare. Therefore, we analyzed paired infant and maternal HSV-2 isolates from two cases of mother-to-infant transmission to identify viral factors contributing to vertical transmission. Sixteen infant isolates with neonatal herpes and 27 genital isolates in their third trimester were included. The infant isolates were significantly more temperature-independent than the maternal isolates. Sequence comparison revealed viral UL13 protein kinase (UL13-PK) mutation in the infant isolates in both cases. In the expanded cohort, infant isolates (5/18) had significantly more UL13-PK mutations than genital isolates (1/29). Isolates within 8 days post-birth (3/4) had a significantly higher frequency of UL13-PK mutation than those after 9 days (2/14), suggesting a close association between UL13-PK mutations and vertical transmission. Elongation factor 1-delta was identified as a target of UL13-PK by proteomic analysis of UL13-PK-positive and -negative HepG2 cells. The mixed infant isolates with the intact and mutated UL13-PK conferred altered cell tropism, temperature independence adapting to fetal temperature, and better growth properties in Vero and hepatoblastoma HepG2 cells than in HSV-2 with intact and mutated UL13-PK alone, indicating that viral UL13-PK mutation is essential for vertical HSV-2 transmission.


Asunto(s)
Herpes Simple , Complicaciones Infecciosas del Embarazo , Embarazo , Femenino , Recién Nacido , Humanos , Herpesvirus Humano 2/genética , Madres , Proteómica , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Virales/genética , Mutación , Tropismo , Transmisión Vertical de Enfermedad Infecciosa
2.
Antiviral Res ; 209: 105474, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36511318

RESUMEN

Human cytomegalovirus (CMV) is a ubiquitous ß-herpesvirus that establishes latent asymptomatic infections in healthy individuals but can cause serious infections in immunocompromised people, resulting in increased risk of morbidity and mortality. The current FDA-approved CMV drugs target late stages of the CMV life-cycle. While these drugs are effective in most cases, they have serious drawbacks, including poor oral bioavailability, dose-limiting toxicity, and a low barrier to resistance. Given the clinical relevance of CMV-associated diseases, novel therapies are needed. Thus, a novel class of compounds that inhibits the early stages of the CMV life-cycle was identified and found to block infection of different strains in physiologically relevant cell types. This class of compounds, N-arylpyrimidinamine (NAPA), demonstrated potent anti-CMV activity against ganciclovir-sensitive and -resistant strains in in vitro replication assays, a selectivity index >30, and favorable in vitro ADME properties. Mechanism of action studies demonstrated that NAPA compounds inhibit an early step of virus infection. NAPA compounds are specific inhibitors of cytomegaloviruses and exhibited limited anti-viral activity against other herpesviruses. Collectively, we have identified a novel class of CMV inhibitor that effectively limits viral infection and proliferation.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Humanos , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/etiología , Antivirales/farmacología , Antivirales/uso terapéutico , Ganciclovir/farmacología , Huésped Inmunocomprometido
3.
Mol Pharm ; 20(1): 370-382, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36484496

RESUMEN

DNA viruses are responsible for many diseases in humans. Current treatments are often limited by toxicity, as in the case of cidofovir (CDV, Vistide), a compound used against cytomegalovirus (CMV) and adenovirus (AdV) infections. CDV is a polar molecule with poor bioavailability, and its overall clinical utility is limited by the high occurrence of acute nephrotoxicity. To circumvent these disadvantages, we designed nine CDV prodrug analogues. The prodrugs modulate the polarity of CDV with a long sulfonyl alkyl chain attached to one of the phosphono oxygens. We added capping groups to the end of the alkyl chain to minimize ß-oxidation and focus the metabolism on the phosphoester hydrolysis, thereby tuning the rate of this reaction by altering the alkyl chain length. With these modifications, the prodrugs have excellent aqueous solubility, optimized metabolic stability, increased cellular permeability, and rapid intracellular conversion to the pharmacologically active diphosphate form (CDV-PP). The prodrugs exhibited significantly enhanced antiviral potency against a wide range of DNA viruses in infected human foreskin fibroblasts. Single-dose intravenous and oral pharmacokinetic experiments showed that the compounds maintained plasma and target tissue levels of CDV well above the EC50 for 24 h. These experiments identified a novel lead candidate, NPP-669. NPP-669 demonstrated efficacy against CMV infections in mice and AdV infections in hamsters following oral (p.o.) dosing at a dose of 1 mg/kg BID and 0.1 mg/kg QD, respectively. We further showed that NPP-669 at 30 mg/kg QD did not exhibit histological signs of toxicity in mice or hamsters. These data suggest that NPP-669 is a promising lead candidate for a broad-spectrum antiviral compound.


Asunto(s)
Infecciones por Citomegalovirus , Organofosfonatos , Profármacos , Ratones , Humanos , Animales , Antivirales/farmacocinética , Disponibilidad Biológica , Profármacos/farmacología , Citosina , Cidofovir
4.
Nat Prod Res ; 36(13): 3286-3296, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33287588

RESUMEN

Allobetulone E-ring rearrangement under treating with HClO4 in Ac2O under reflux afforded new triterpenoids: 3,28-diacetoxy-21-acetyl-2(3),20(21)-18α,19ßH-ursandiene 3 and 3,28-diacetoxy-2(3),18(19)-oleandiene 4. 18α,19ßH-Ursanes were transformed at A- and E-rings into indolo- and bis-furfurylidene 7 derivatives. Structure elucidation was performed using COSY, NOESY, HSQC and HMBC experiments, and X-Ray analysis for 3. The potential of newly obtained 18α,19ßH-ursanes was evaluated against HCMV and HPV-11, the NCI-60 cancer cell panel and inhibition of α-glucosidase. All of the compounds have shown viral inhibition towards HCMV compared to standard drug Acyclovir. 3ß-Acetoxy-21ß-acetyl-20ß,28-epoxy-18α,19ßН-ursane 1 showed moderate activity (EC50 4.87 µM) towards the HCMV-resistant isolate (GDGr K17) compared to standard drug Cidofovir and was four times more potent than Ganciclovir. Compound 7 inhibited the cell growth of the three melanoma and one colon cancer cell. 3-Oxo-21ß-acetyl-20ß,28-epoxy-18α,19ßН-ursane 5 and compound 7 inhibited α-glucosidase with IC50 28.0 µM and 4.0 µM being from 6 to 44 times more active than acarbose.


Asunto(s)
Triterpenos , alfa-Glucosidasas , Antivirales/farmacología , Línea Celular Tumoral , Estructura Molecular , Triterpenos/química
5.
Antiviral Res ; 195: 105179, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34530009

RESUMEN

Orthopoxviruses such as variola and monkeypox viruses continue to threaten the human population. Monkeypox virus is endemic in central and western Africa and outbreaks have reached as far as the U.S. Although variola virus, the etiologic agent of smallpox, has been eradicated by a successful vaccination program, official and likely clandestine stocks of the virus exist. Moreover, studies with ectromelia virus (the etiological agent of mousepox) have revealed that IL-4 recombinant viruses are significantly more virulent than wild-type viruses even in mice treated with vaccines and/or antivirals. For these reasons, it is critical that antiviral modalities are developed to treat these viruses should outbreaks, or deliberate dissemination, occur. Currently, 2 antivirals (brincidofovir and tecovirimat) are in the U.S. stockpile allowing for emergency use of the drugs to treat smallpox. Both antivirals have advantages and disadvantages in a clinical and emergency setting. Here we report on the efficacy of a recombinant immunoglobulin (rVIG) that demonstrated efficacy against several orthopoxviruses in vitro and in vivo in both a prophylactic and therapeutic fashion. A single intraperitoneal injection of rVIG significantly protected mice when given up to 14 days before or as late as 6 days post challenge. Moreover, rVIG reduced morbidity, as measured by weight-change, as well as several previously established biomarkers of disease. In rVIG treated mice, we found that vDNA levels in blood were significantly reduced, as was ALT (a marker of liver damage) and infectious virus levels in the liver. No apparent adverse events were observed in rVIG treated mice, suggesting the immunoglobulin is well tolerated. These findings suggest that recombinant immunoglobulins could be candidates for further evaluation and possible licensure under the FDA Animal Rule.


Asunto(s)
Antivirales/uso terapéutico , Inmunoglobulinas/uso terapéutico , Orthopoxvirus/efectos de los fármacos , Viruela/tratamiento farmacológico , Vaccinia/tratamiento farmacológico , Animales , Antivirales/administración & dosificación , Benzamidas , Línea Celular , Chlorocebus aethiops , Citosina/análogos & derivados , Femenino , Humanos , Isoindoles , Ratones , Ratones Endogámicos BALB C , Organofosfonatos , Viruela/prevención & control , Viruela/virología , Vacuna contra Viruela/administración & dosificación , Vacunas de ADN/administración & dosificación , Vaccinia/prevención & control , Vaccinia/virología
6.
RSC Med Chem ; 12(5): 804-808, 2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34124679

RESUMEN

The substantial impact of acyclic nucleoside phosphonates (ANPs) on human medicine encourages the synthesis of new ANP analogues with a potentially differentiated antiviral spectrum. Herein, we demonstrate the functionalization of the 2-position of the (R,S)-3-hydroxy-2-(phosphonomethoxy)propyl side-chain of an inactive ANP with a polar cyano group to generate a thymine analogue with selective inhibition of hepatitis B virus (HBV) replication (SI > 302; EC50 = 0.33 µM), without significant antiretroviral activity. These findings suggest new strategies to synthesize unique ANPs with a targeted antiviral profile.

7.
Sci Rep ; 11(1): 656, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436903

RESUMEN

Lectins, carbohydrate-binding proteins, have been regarded as potential antiviral agents, as some can bind glycans on viral surface glycoproteins and inactivate their functions. However, clinical development of lectins has been stalled by the mitogenicity of many of these proteins, which is the ability to stimulate deleterious proliferation, especially of immune cells. We previously demonstrated that the mitogenic and antiviral activities of a lectin (banana lectin, BanLec) can be separated via a single amino acid mutation, histidine to threonine at position 84 (H84T), within the third Greek key. The resulting lectin, H84T BanLec, is virtually non-mitogenic but retains antiviral activity. Decreased mitogenicity was associated with disruption of pi-pi stacking between two aromatic amino acids. To examine whether we could provide further proof-of-principle of the ability to separate these two distinct lectin functions, we identified another lectin, Malaysian banana lectin (Malay BanLec), with similar structural features as BanLec, including pi-pi stacking, but with only 63% amino acid identity, and showed that it is both mitogenic and potently antiviral. We then engineered an F84T mutation expected to disrupt pi-pi stacking, analogous to H84T. As predicted, F84T Malay BanLec (F84T) was less mitogenic than wild type. However, F84T maintained strong antiviral activity and inhibited replication of HIV, Ebola, and other viruses. The F84T mutation disrupted pi-pi stacking without disrupting the overall lectin structure. These findings show that pi-pi stacking in the third Greek key is a conserved mitogenic motif in these two jacalin-related lectins BanLec and Malay BanLec, and further highlight the potential to rationally engineer antiviral lectins for therapeutic purposes.


Asunto(s)
Antivirales/farmacología , Infecciones por VIH/tratamiento farmacológico , Lectinas/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Mitógenos/farmacología , Musa/química , Replicación Viral , Proliferación Celular , Células Cultivadas , Ebolavirus/efectos de los fármacos , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Humanos , Lectinas/química , Lectinas/genética , Leucocitos Mononucleares/virología
8.
Antimicrob Agents Chemother ; 64(11)2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32816736

RESUMEN

Human adenovirus (HAdV) infection is common in the general population and can cause a range of clinical manifestations, among which pneumonia and keratoconjunctivitis are the most common. Although HAdV infections are mostly self-limiting, infections in immunocompromised individuals can be severe. No antiviral drug has been approved for treating adenoviruses. Filociclovir (FCV) is a nucleoside analogue which has successfully completed phase I human clinical safety studies and is now being developed for treatment of human cytomegalovirus (HCMV)-related disease in immunocompromised patients. In this report, we show that FCV is a potent broad-spectrum inhibitor of HAdV types 4 to 8, with 50% effective concentrations (EC50s) ranging between 1.24 and 3.6 µM and a 50% cytotoxic concentration (CC50) of 100 to 150 µM in human foreskin fibroblasts (HFFs). We also show that the prophylactic oral administration of FCV (10 mg/kg of body weight) 1 day prior to virus challenge and then daily for 14 days to immunosuppressed Syrian hamsters infected intravenously with HAdV6 was sufficient to prevent morbidity and mortality. FCV also mitigated tissue damage and inhibited virus replication in the liver. The 10-mg/kg dose had similar effects even when the treatment was started on day 4 after virus challenge. Furthermore, FCV administered at the same dose after intranasal challenge with HAdV6 partially mitigated body weight loss but significantly reduced pathology and virus replication in the lung. These findings suggest that FCV could potentially be developed as a pan-adenoviral inhibitor.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Infecciones por Citomegalovirus , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Cricetinae , Infecciones por Citomegalovirus/tratamiento farmacológico , Humanos , Replicación Viral
9.
J Infect Dis ; 221(Suppl 1): S32-S44, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134483

RESUMEN

The development of therapeutics for cytomegalovirus (CMV) infections, while progressing, has not matched the pace of new treatments of human immunodeficiency virus (HIV) infections; nevertheless, recent developments in the treatment of CMV infections have resulted in improved human health and perhaps will encourage the development of new therapeutic approaches. First, the deployment of ganciclovir and valganciclovir for both the prevention and treatment of CMV infections and disease in transplant recipients has been further improved with the licensure of the efficacious and less toxic letermovir. Regardless, late-onset CMV disease, specifically pneumonia, remains problematic. Second, the treatment of congenital CMV infections with valganciclovir has beneficially improved both hearing and neurologic outcomes, both fundamental advances for these children. In these pediatric studies, viral load was decreased but not eliminated. Thus, an important lesson learned from studies in both populations is the need for new antiviral agents and the necessity for combination therapies as has been shown to be beneficial in the treatment of HIV infections, among others. The development of monoclonal antibodies, sirtuins, and cyclopropovir may provide new treatment options.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Desarrollo de Medicamentos , Acetatos/administración & dosificación , Acetatos/efectos adversos , Acetatos/uso terapéutico , Antivirales/farmacología , Biomarcadores , Estudios Clínicos como Asunto , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/transmisión , Infecciones por Citomegalovirus/virología , Farmacorresistencia Viral , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos , Quinazolinas/uso terapéutico , Proyectos de Investigación , Sirtuinas/administración & dosificación , Sirtuinas/efectos adversos , Sirtuinas/uso terapéutico , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos , Resultado del Tratamiento , Carga Viral
10.
Chem Biodivers ; 16(10): e1900391, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31479201

RESUMEN

A series of novel 2-oxoimidazolidine derivatives were synthesized and their antiviral activities against BK human polyomavirus type 1 (BKPyV) were evaluated in vitro. Bioassays showed that the synthesized compounds 1-{[(4E)-5-(dichloromethylidene)-2-oxoimidazolidin-4-ylidene]sulfamoyl}piperidine-4-carboxylic acid (5) and N-Cyclobutyl-N'-[(4E)-5-(dichloromethylidene)-2-oxoimidazolidin-4-ylidene]sulfuric diamide (4) exhibited moderate activities against BKPyV (EC50 =5.4 and 5.5 µm, respectively) that are comparable to the standard drug Cidofovir. Compound 5 exhibited the same cytotoxicity in HFF cells and selectivity index (SI50 ) as Cidofovir. The selectivity index of compound 4 is three times less than that of Cidofovir due to the higher toxicity of this compound. Hence, these compounds may be taken as lead compound for further development of novel ant-BKPyV agents.


Asunto(s)
Antivirales/farmacología , Virus BK/efectos de los fármacos , Cidofovir/farmacología , Diseño de Fármacos , Imidazolidinas/farmacología , Antivirales/síntesis química , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cidofovir/química , Relación Dosis-Respuesta a Droga , Humanos , Imidazolidinas/síntesis química , Imidazolidinas/química , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
11.
mSphere ; 4(1)2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814317

RESUMEN

More than 14,000 neonates are infected with herpes simplex virus (HSV) annually. Approximately half display manifestations limited to the skin, eyes, or mouth (SEM disease). The rest develop invasive infections that spread to the central nervous system (CNS disease or encephalitis) or throughout the infected neonate (disseminated disease). Invasive HSV disease is associated with significant morbidity and mortality, but the viral and host factors that predispose neonates to these forms are unknown. To define viral diversity within the infected neonatal population, we evaluated 10 HSV-2 isolates from newborns with a range of clinical presentations. To assess viral fitness independently of host immune factors, we measured viral growth characteristics in cultured cells and found diverse in vitro phenotypes. Isolates from neonates with CNS disease were associated with larger plaque size and enhanced spread, with the isolates from cerebrospinal fluid (CSF) exhibiting the most robust growth. We sequenced complete viral genomes of all 10 neonatal viruses, providing new insights into HSV-2 genomic diversity in this clinical setting. We found extensive interhost and intrahost genomic diversity throughout the viral genome, including amino acid differences in more than 90% of the viral proteome. The genes encoding glycoprotein G (gG; US4), glycoprotein I (gI; US7), and glycoprotein K (gK; UL53) and viral proteins UL8, UL20, UL24, and US2 contained variants that were found in association with CNS isolates. Many of these viral proteins are known to contribute to cell spread and neurovirulence in mouse models of CNS disease. This report represents the first application of comparative pathogen genomics to neonatal HSV disease.IMPORTANCE Herpes simplex virus (HSV) causes invasive disease in half of infected neonates, resulting in significant mortality and permanent cognitive morbidity. The factors that contribute to invasive disease are not understood. This study revealed diversity among HSV isolates from infected neonates and detected the first associations between viral genetic variations and clinical disease manifestations. We found that viruses isolated from newborns with encephalitis showed enhanced spread in culture. These viruses contained protein-coding variations not found in viruses causing noninvasive disease. Many of these variations were found in proteins known to impact neurovirulence and viral spread between cells. This work advances our understanding of HSV diversity in the neonatal population and how it may impact disease outcome.


Asunto(s)
Variación Genética , Herpes Simple/virología , Herpesvirus Humano 2/genética , Complicaciones Infecciosas del Embarazo/virología , Línea Celular , Encefalitis Viral/virología , Femenino , Genoma Viral , Genómica , Genotipo , Edad Gestacional , Herpes Simple/complicaciones , Herpesvirus Humano 1/genética , Herpesvirus Humano 2/aislamiento & purificación , Herpesvirus Humano 2/patogenicidad , Humanos , Recién Nacido , Masculino , Fenotipo , Embarazo , Proteínas Virales/genética
12.
J Perinatol ; 39(5): 683-689, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723275

RESUMEN

OBJECTIVE: To determine frequency of hospital-acquired viral respiratory infections (HA-VRI) and associated outcomes in a NICU. STUDY DESIGN: Prospective cohort study conducted from 4 October 2016 to 21 March 2017. Infants hospitalized from birth in the NICU had a weekly nasal swab collected for testing using a multiplex PCR assay capable of detecting 16 different respiratory viruses. RESULTS: Seventy-four infants enrolled, with 5 (6.8%) testing positive for a virus (incidence rate of 1.3/1000 patient days). VRI positive infants had a younger gestational age (median 27 w vs. 32 w, p = 0.048); were hospitalized longer (97 d vs 43 d, p = 0.013); required more antibiotics (8 d vs. 4 d, p = 0.037) and were more likely to be diagnosed with bronchopulmonary dysplasia (p = 0.008) compared to VRI negative infants. CONCLUSION: Respiratory viruses are a frequent cause of HAI in the NICU and are associated with negative outcomes.


Asunto(s)
Infección Hospitalaria/virología , Unidades de Cuidado Intensivo Neonatal , Infecciones del Sistema Respiratorio/virología , Virosis/diagnóstico , Alabama , Displasia Broncopulmonar/diagnóstico , Infección Hospitalaria/diagnóstico , Femenino , Edad Gestacional , Hospitalización , Humanos , Recién Nacido de Bajo Peso , Recién Nacido , Recien Nacido Prematuro , Masculino , Reacción en Cadena de la Polimerasa , Estudios Prospectivos , Infecciones del Sistema Respiratorio/diagnóstico
13.
Org Biomol Chem ; 17(3): 585-597, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30574983

RESUMEN

This paper describes an access to new nitrogen-containing heterocyclic triterpenoids by the reaction of 2,3-indolotriterpenoids with ozone and dimethyldioxirane. The oxidation of indolo-fused 28-oxo-allobetulin or methyl platanoate with ozone led to a mixture of a quinolone as the major product and a nine-membered 2,3-seco-2-oxolactam and three different types of spiroindoles as byproducts. The formation of quinolone and 2,3-seco-2-oxolactam derivatives could be explained by the standard 1,3-dipolar cycloaddition of ozone to the C2(3)-double bond of the triterpene core similar to the products observed in the ozonolysis of indoles in the Witkop-Winterfeldt oxidation (WWO). The formation of spiroindoles was unexpected and could be explained through the 1,2-cycloaddition of ozone to the C2(3)-double bond with consecutive intramolecular rearrangements of the 2,3-epoxy-intermediate. These spiroindoles seem to be novel structures observed in the WWO reaction. The formation of only two isomeric triterpene spiroindolinones was achieved by the oxidation of 2,3-indolo-28-oxo-allobetulin with dimethyldioxirane that could be explained by the rearrangement of the 2,3-epoxy-intermediate. 19ß,28-Epoxy-18α-olean-28-oxo-2-nor-2,3-4'(1H)-quinolone was the most active against HPV-11 with EC50 0.45 µM and SI50 322 in a primary assay and SI90 < 10 against HPV-16 in a secondary assay. The oxidative transformations of indolotriterpenoids have great potential for further modifications towards the preparation of new biologically active compounds.

14.
Sci Rep ; 8(1): 16662, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413769

RESUMEN

Acyclovir (ACV) is an effective antiviral agent for treating lytic Herpes Simplex virus, type 1 (HSV-1) infections, and it has dramatically reduced the mortality rate of herpes simplex encephalitis. However, HSV-1 resistance to ACV and its derivatives is being increasingly documented, particularly among immunocompromised individuals. The burgeoning drug resistance compels the search for a new generation of more efficacious anti-herpetic drugs. We have previously shown that trans-dihydrolycoricidine (R430), a lycorane-type alkaloid derivative, effectively inhibits HSV-1 infections in cultured cells. We now report that R430 also inhibits ACV-resistant HSV-1 strains, accompanied by global inhibition of viral gene transcription and enrichment of H3K27me3 methylation on viral gene promoters. Furthermore, we demonstrate that R430 prevents HSV-1 reactivation from latency in an ex vivo rodent model. Finally, among a panel of DNA viruses and RNA viruses, R430 inhibited Zika virus with high therapeutic index. Its therapeutic index is comparable to standard antiviral drugs, though it has greater toxicity in non-neuronal cells than in neuronal cells. Synthesis of additional derivatives could enable more efficacious antivirals and the identification of active pharmacophores.


Asunto(s)
Alcaloides de Amaryllidaceae/farmacología , Antivirales/farmacología , Infecciones por Virus ADN/tratamiento farmacológico , Virus ADN/efectos de los fármacos , Infecciones por Virus ARN/tratamiento farmacológico , Virus ARN/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Células Cultivadas , Chlorocebus aethiops , Infecciones por Virus ADN/virología , Humanos , Ratones , Infecciones por Virus ARN/virología , Células Vero
15.
Antiviral Res ; 159: 104-112, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30287226

RESUMEN

The search for new compounds with a broad spectrum of antiviral activity is important and requires the evaluation of many compounds against several distinct viruses. Researchers attempting to develop new antiviral therapies for DNA virus infections currently use a variety of cell lines, assay conditions and measurement methods to determine in vitro drug efficacy, making it difficult to compare results from within the same laboratory as well as between laboratories. In this paper we describe a common assay platform designed to facilitate the parallel evaluation of antiviral activity against herpes simplex virus type 1, herpes simplex virus type 2, varicella-zoster virus, cytomegalovirus, vaccinia virus, cowpox virus, and adenovirus. The automated assays utilize monolayers of primary human foreskin fibroblast cells in 384-well plates as a common cell substrate and cytopathic effects and cytotoxicity are quantified with CellTiter-Glo. Data presented demonstrate that each of the assays is highly robust and yields data that are comparable to those from other traditional assays, such as plaque reduction assays. The assays proved to be both accurate and robust and afford an in depth assessment of antiviral activity against the diverse class of viruses with very small quantities of test compounds. In an accompanying paper, we present a standardized approach to evaluating antivirals against lymphotropic herpesviruses and polyomaviruses and together these studies revealed new activities for reference compounds. This approach has the potential to accelerate the development of broad spectrum therapies for the DNA viruses.


Asunto(s)
Adenoviridae/efectos de los fármacos , Antivirales/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Orthopoxvirus/efectos de los fármacos , Ensayo de Placa Viral/normas , Células Cultivadas , Citomegalovirus/efectos de los fármacos , Efecto Citopatogénico Viral , Infecciones por Virus ADN/tratamiento farmacológico , Fibroblastos , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 3/efectos de los fármacos , Humanos
16.
Antiviral Res ; 159: 122-129, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30287227

RESUMEN

The search for new compounds with a broad spectrum of antiviral activity is important and requires the evaluation of many compounds against several distinct viruses. Researchers attempting to develop new antiviral therapies for DNA virus infections currently use a variety of cell lines, assay conditions and measurement methods to determine in vitro drug efficacy, making it difficult to compare results from within the same laboratory as well as between laboratories. In this paper, we describe the assessment of antiviral activity of a set of nucleoside analogs against BK polyomavirus, JC polyomavirus, Epstein-Barr virus, human herpesvirus 6B, and human herpesvirus 8 in an automated 384-well format and utilize qPCR assays to measure the accumulation of viral DNA. In an accompanying paper, we present a standardized approach to evaluating antivirals against additional herpesviruses, orthopoxviruses, and adenovirus. Together, they reveal new activities for reference compounds and help to define the spectrum of antiviral activity for a set of nucleoside analogs against a set of 12 DNA viruses that infect humans including representative human herpesviruses, orthopoxviruses, adenoviruses, and polyomaviruses. This analysis helps provide perspective on combinations of agents that would help provide broad coverage of significant pathogens in immunocompromised patients as well as against emerging infections.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas/normas , Herpesviridae/efectos de los fármacos , Nucleósidos/farmacología , Poliomavirus/efectos de los fármacos , Automatización de Laboratorios , ADN Viral/análisis , Descubrimiento de Drogas/métodos , Humanos , Replicación Viral/efectos de los fármacos
17.
Antiviral Res ; 159: 153-174, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30227153

RESUMEN

The recent approval of letermovir marks a new era of therapy for human cytomegalovirus (HCMV) infections, particularly for the prevention of HCMV disease in hematopoietic stem cell transplant recipients. For almost 30 years ganciclovir has been the therapy of choice for these infections and by today's standards this drug exhibits only modest antiviral activity that is often insufficient to completely suppress viral replication, and drives the selection of drug-resistant variants that continue to replicate and contribute to disease. While ganciclovir remains the therapy of choice, additional drugs that inhibit novel molecular targets, such as letermovir, will be required as highly effective combination therapies are developed not only for the treatment of immunocompromised hosts, but also for congenitally infected infants. Sustained efforts, largely in the biotech industry and academia, have identified additional highly active lead compounds that have progressed into clinical studies with varying levels of success and at least two have the potential to be approved in the near future. Some of the new drugs in the pipeline inhibit new molecular targets, remain effective against isolates that have developed resistance to existing therapies, and promise to augment existing therapeutic regimens. Here, we will describe some of the unique features of HCMV biology and discuss their effect on therapeutic needs. Existing drugs will also be discussed and some of the more promising candidates will be reviewed with an emphasis on those progressing through clinical studies. The in vitro and in vivo antiviral activity, spectrum of antiviral activity, and mechanism of action of new compounds will be reviewed to provide an update on potential new therapies for HCMV infections that have progressed significantly in recent years.


Asunto(s)
Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Ensayos Clínicos como Asunto , Citomegalovirus/fisiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Ratones
18.
Xenotransplantation ; 25(4): e12427, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30264882

RESUMEN

BACKGROUND: Recent advances in xenotransplantation have produced organs from pigs that are well tolerated in primate models because of genetic changes engineered to delete major antigens from donor animals. To ensure the safety of human transplant recipients, it will be essential to understand both the spectrum of infectious agents in donor pigs and their potential to be transmitted to immunocompromised transplant recipients. Equally important will be the development of new highly sensitive diagnostic methods for use in the detection of these agents in donor animals and for the monitoring of transplant recipients. METHODS: Herein, we report the development of a panel of 30 quantitative polymerase chain reaction (qPCR) assays for infectious agents with the potential to be transmitted to the human host. The reproducibility, sensitivity and specificity of each assay were evaluated and were found to exhibit analytic sensitivity that was similar to that of quantitative assays used to perform viral load testing of human viruses in clinical laboratories. RESULTS: This analytical approach was used to detect nucleic acids of infectious agents present in specimens from 9 sows and 22 piglets derived by caesarean section. The most commonly detected targets in adult animals were Mycoplasma species and two distinct herpesviruses, porcine lymphotrophic herpesvirus 2 and 3. A total of 14 piglets were derived from three sows infected with either or both herpesviruses, yet none tested positive for the viruses indicating that vertical transmission of these viruses is inefficient. CONCLUSIONS: The data presented demonstrate that procedures in place are highly sensitive and can specifically detect nucleic acids from target organisms in the panel, thus ensuring the safety of organs for transplantation as well as the monitoring of patients potentially receiving them.


Asunto(s)
Herpesviridae/patogenicidad , Xenoinjertos/virología , Enfermedades de los Porcinos/virología , Trasplante Heterólogo/efectos adversos , Animales , Citomegalovirus/genética , Humanos , Reproducibilidad de los Resultados , Porcinos , Enfermedades de los Porcinos/diagnóstico
19.
Antiviral Res ; 153: 1-9, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29510156

RESUMEN

Human adenoviruses (AdV) cause generally mild infections of the respiratory and GI tracts as well as some other tissues. However, AdV can cause serious infection in severely immunosuppressed individuals, especially pediatric patients undergoing allogeneic hematopoietic stem cell transplantation, where mortality rates are up to 80% with disseminated disease. Despite the seriousness of AdV disease, there are no drugs approved specifically to treat AdV infections. We report here that USC-087, an N-alkyl tyrosinamide phosphonate ester prodrug of HPMPA, the adenine analog of cidofovir, is highly effective against multiple AdV types in cell culture. USC-087 is also effective against AdV-C6 in our immunosuppressed permissive Syrian hamster model. In this model, hamsters are immunosuppressed by treatment with high dose cyclophosphamide. Injection of AdV-C6 (or AdV-C5) intravenously leads to a disseminated infection that resembles the disease seen in humans, including death. We have tested the efficacy of orally-administered USC-087 against the median lethal dose of intravenously administered AdV-C6. USC-087 completely prevented or significantly decreased mortality when administered up to 4 days post challenge. USC-087 also prevented or significantly decreased liver damage caused by AdV-C6 infection, and suppressed virus replication even when administered 4 days post challenge. These results imply that USC-087 is a promising candidate for drug development against HAdV infections.


Asunto(s)
Adenina/análogos & derivados , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Antivirales/administración & dosificación , Organofosfonatos/administración & dosificación , Profármacos/administración & dosificación , Tirosina/análogos & derivados , Adenina/administración & dosificación , Administración Oral , Animales , Modelos Animales de Enfermedad , Huésped Inmunocomprometido , Hígado/patología , Mesocricetus , Análisis de Supervivencia , Resultado del Tratamiento , Tirosina/administración & dosificación
20.
Antiviral Res ; 149: 1-6, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29113740

RESUMEN

Pritelivir, a helicase-primase inhibitor, has excellent in vitro and in vivo activity against human herpes simplex virus (HSV). Mice lethally infected with HSV type 1 or 2, including acyclovir-resistant strains, were treated 72 h after infection for 7 days with pritelivir or acyclovir. Both drugs were administered orally twice daily either alone or in combination. Dosages of pritelivir from 0.3 to 30 mg/kg reduced mortality (P < 0.001) against HSV-1, E-377. With an acyclovir resistant HSV-1, 11360, pritelivir at 1 and 3 mg/kg increased survival (P < 0.005). With HSV-2, MS infected mice, all dosages higher than the 0.3 mg/kg dose of pritelivir were effective (P < 0.005). For acyclovir resistant HSV-2, strain 12247, pritelivir dosages of 1-3 mg/kg significantly improved survival (P < 0.0001). Combination therapies of pritelivir at 0.1 or 0.3 mg/kg/dose with acyclovir (10 mg/kg/dose) were protective (P < 0.0001) when compared to the vehicle treated group against HSV-2, strain MS (in line with previous data using HSV-1). An increased mean days to death (P < 0.05) was also observed and was indicative of a potential synergy. Pharmacokinetic studies were performed to determine pritelivir concentrations and a dose dependent relationship was found in both plasma and brain samples regardless of infection status or time of initiation of dosing. In summary, pritelivir was shown to be active when treatment was delayed to 72 h post viral inoculation and appeared to synergistically inhibit mortality in this model in combination with acyclovir. We conclude pritelivir has potent and resistance-breaking antiviral efficacy with potential for the treatment of potentially life-threatening HSV type 1 and 2 infections, including herpes simplex encephalitis.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Encefalitis por Herpes Simple/virología , Piridinas/farmacología , Tiazoles/farmacología , Aciclovir/administración & dosificación , Aciclovir/farmacocinética , Animales , Antivirales/administración & dosificación , Antivirales/farmacocinética , Modelos Animales de Enfermedad , Quimioterapia Combinada , Encefalitis por Herpes Simple/tratamiento farmacológico , Encefalitis por Herpes Simple/mortalidad , Encefalitis por Herpes Simple/patología , Femenino , Humanos , Ratones , Piridinas/administración & dosificación , Piridinas/farmacocinética , Sulfonamidas , Tiazoles/administración & dosificación , Tiazoles/farmacocinética , Distribución Tisular , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...