Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Mol Pharm ; 20(1): 370-382, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36484496

ABSTRACT

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.


Subject(s)
Cytomegalovirus Infections , Organophosphonates , Prodrugs , Mice , Humans , Animals , Antiviral Agents/pharmacokinetics , Biological Availability , Prodrugs/pharmacology , Cytosine , Cidofovir
2.
Antimicrob Agents Chemother ; 64(11)2020 10 20.
Article in English | MEDLINE | ID: mdl-32816736

ABSTRACT

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.


Subject(s)
Adenovirus Infections, Human , Adenoviruses, Human , Cytomegalovirus Infections , Adenovirus Infections, Human/drug therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cricetinae , Cytomegalovirus Infections/drug therapy , Humans , Virus Replication
3.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28250128

ABSTRACT

Syrian hamsters are permissive for the replication of species C human adenoviruses (HAdV-C). The virus replicates to high titers in the liver of these animals after intravenous infection, while respiratory infection results in virus replication in the lung. Here we show that two types belonging to species C, HAdV-C5 and HAdV-C6, replicate to significantly different extents and cause pathology with significantly different severities, with HAdV-C6 replicating better and inducing more severe and more widespread lesions. The virus burdens in the livers of HAdV-C6-infected hamsters are higher than the virus burdens in HAdV-C5-infected ones because more of the permissive hepatocytes get infected. Furthermore, when hamsters are infected intravenously with HAdV-C6, live, infectious virus can be isolated from the lung and the kidney, which is not seen with HAdV-C5. Similarly to mouse models, in hamsters, HAdV-C6 is sequestered by macrophages to a lesser degree than HAdV-C5. Depletion of Kupffer cells from the liver greatly increases the replication of HAdV-C5 in the liver, while it has only a modest effect on the replication of HAdV-C6. Elimination of Kupffer cells also dramatically increases the pathology induced by HAdV-C5. These findings indicate that in hamsters, pathology resulting from intravenous infection with adenoviruses is caused mostly by replication in hepatocytes and not by the abortive infection of Kupffer cells and the following cytokine storm.IMPORTANCE Immunocompromised human patients can develop severe, often lethal adenovirus infections. Respiratory adenovirus infection among military recruits is a serious problem, in some cases requiring hospitalization of the patient. Furthermore, adenovirus-based vectors are frequently used as experimental viral therapeutic agents. Thus, it is imperative that we investigate the pathogenesis of adenoviruses in a permissive animal model. Syrian hamsters are susceptible to infection with certain human adenoviruses, and the pathology accompanying these infections is similar to what is observed with adenovirus-infected human patients. We demonstrate that replication in permissive cells in a susceptible host animal is a major part of the mechanism by which systemic adenovirus infection induces pathology, as opposed to the chiefly immune-mediated pathology observed in nonsusceptible hosts. These findings support the use of compounds inhibiting adenovirus replication as a means to block adenovirus-induced pathology.


Subject(s)
Adenovirus Infections, Human/pathology , Adenovirus Infections, Human/virology , Adenoviruses, Human/pathogenicity , Liver/virology , Viral Load , Virus Replication , Adenoviruses, Human/classification , Adenoviruses, Human/physiology , Animals , Cell Line , Cricetinae , Disease Models, Animal , Humans , Kidney/virology , Kupffer Cells/virology , Liver/pathology , Lung/virology , Macrophages/virology , Mesocricetus
5.
PLoS Pathog ; 11(8): e1005084, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26291525

ABSTRACT

Human adenoviruses have been studied extensively in cell culture and have been a model for studies in molecular, cellular, and medical biology. However, much less is known about adenovirus replication and pathogenesis in vivo in a permissive host because of the lack of an adequate animal model. Presently, the most frequently used permissive immunocompetent animal model for human adenovirus infection is the Syrian hamster. Species C human adenoviruses replicate in these animals and cause pathology that is similar to that seen with humans. Here, we report findings with a new Syrian hamster strain in which the STAT2 gene was functionally knocked out by site-specific gene targeting. Adenovirus-infected STAT2 knockout hamsters demonstrated an accentuated pathology compared to the wild-type control animals, and the virus load in the organs of STAT2 knockout animals was 100- to 1000-fold higher than that in wild-type hamsters. Notably, the adaptive immune response to adenovirus is not adversely affected in STAT2 knockout hamsters, and surviving hamsters cleared the infection by 7 to 10 days post challenge. We show that the Type I interferon pathway is disrupted in these hamsters, revealing the critical role of interferon-stimulated genes in controlling adenovirus infection. This is the first study to report findings with a genetically modified Syrian hamster infected with a virus. Further, this is the first study to show that the Type I interferon pathway plays a role in inhibiting human adenovirus replication in a permissive animal model. Besides providing an insight into adenovirus infection in humans, our results are also interesting from the perspective of the animal model: STAT2 knockout Syrian hamster may also be an important animal model for studying other viral infections, including Ebola-, hanta-, and dengue viruses, where Type I interferon-mediated innate immunity prevents wild type hamsters from being effectively infected to be used as animal models.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/pathogenicity , Disease Models, Animal , Interferon Type I/immunology , STAT2 Transcription Factor/deficiency , Adenoviridae Infections/pathology , Adenoviruses, Human/immunology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cricetinae , Flow Cytometry , Gene Knockout Techniques , Humans , Mesocricetus , Reverse Transcriptase Polymerase Chain Reaction , STAT2 Transcription Factor/immunology
6.
J Virol ; 88(4): 2291-300, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24335311

ABSTRACT

Oncolytic viruses based on adenovirus type 5 (Ad5) have been developed as a new class of therapeutic agents for cancers that are resistant to conventional therapies. Clinical experience shows that these agents are safe, but virotherapy alone has not achieved long-term cure in cancer patients. The vast majority of oncolytic adenoviruses used in clinical trials to date have deletion of the E3B genes. It has been demonstrated that the antitumor potency of the E3B-deleted mutant (dl309) is inferior to adenovirus with E3B genes intact. Tumors treated with dl309 show markedly greater macrophage infiltration than E3B-intact adenovirus. However, the functional mechanisms for this were not previously known. Here, we demonstrate that deletion of E3B genes increases production of chemokines by monocytes after adenovirus infection and increases monocyte migration. The E3B 14,700-Da protein (E3B-14.7K) inhibits STAT1 function by preventing its phosphorylation and nuclear translocation. The STAT1 inhibitor, fludarabine, rescues the effect of E3B-14.7K deletion by downregulating target chemokine expression in human and murine monocytes and results in an enhanced antitumor efficacy with dl309 in vivo. These findings have important implications for clinical use of E3B-deleted oncolytic adenovirus and other E3B-deleted adenovirus vector-based therapy.


Subject(s)
Adenoviridae/physiology , Adenovirus E3 Proteins/metabolism , Monocytes/metabolism , Oncolytic Viruses/physiology , STAT1 Transcription Factor/metabolism , Active Transport, Cell Nucleus/drug effects , Adenoviridae/metabolism , Adenovirus E3 Proteins/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Line , DNA, Complementary/biosynthesis , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , Oncolytic Viruses/metabolism , Phosphorylation/drug effects , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/antagonists & inhibitors , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
7.
Antimicrob Agents Chemother ; 58(12): 7171-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224011

ABSTRACT

Adenovirus infections of immunocompromised patients can develop into deadly multiorgan or systemic disease. The virus is especially threatening for pediatric allogeneic hematopoietic stem cell transplant recipients; according to some studies, 10% or more of these patients succumb to disease resulting from adenovirus infection. At present, there is no drug approved for the treatment or prevention of adenovirus infections. Compounds that are approved to treat other virus infections are used off-label to combat adenovirus, but only anecdotal evidence of the efficacy of these drugs exists. Ganciclovir, a drug approved for the treatment of herpesvirus infection, was previously reported to be effective against human adenoviruses in vitro. To model adenovirus infections in immunocompromised humans, we examined ganciclovir's efficacy in immunosuppressed Syrian hamsters intravenously infected with type 5 human adenovirus (Ad5). This animal model is permissive for Ad5 replication, and the animals develop symptoms similar to those seen in humans. We demonstrate that ganciclovir suppresses Ad5 replication in the liver of infected hamsters and that it mitigates the consequences of Ad5 infections in these animals when administered prophylactically or therapeutically. We show that ganciclovir inhibits Ad5 DNA synthesis and late gene expression. The mechanism of action for the drug is not clear; preliminary data suggest that it exerts its antiadenoviral effect by directly inhibiting the adenoviral DNA polymerase. While more extensive studies are required, we believe that ganciclovir is a promising drug candidate to treat adenovirus infections. Brincidofovir, a drug with proven activity against Ad5, was used as a positive control in the prophylactic experiment.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Ganciclovir/pharmacology , Immunocompromised Host , Viral Proteins/antagonists & inhibitors , Adenoviridae Infections/immunology , Adenoviridae Infections/mortality , Adenoviridae Infections/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/growth & development , Adenoviruses, Human/pathogenicity , Animals , Body Weight/drug effects , Cell Line, Tumor , Cytosine/analogs & derivatives , Cytosine/pharmacology , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Female , Gene Expression , Humans , Male , Mesocricetus , Organophosphonates/pharmacology , Survival Analysis , Transaminases/blood , Viral Load/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
8.
Antimicrob Agents Chemother ; 58(12): 7451-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267681

ABSTRACT

Herpesviruses are large double-stranded DNA viruses that cause serious human diseases. Herpesvirus DNA replication depends on multiple processes typically catalyzed by nucleotidyltransferase superfamily (NTS) enzymes. Therefore, we investigated whether inhibitors of NTS enzymes would suppress replication of herpes simplex virus 1 (HSV-1) and HSV-2. Eight of 42 NTS inhibitors suppressed HSV-1 and/or HSV-2 replication by >10-fold at 5 µM, with suppression at 50 µM reaching ∼1 million-fold. Five compounds in two chemical families inhibited HSV replication in Vero and human foreskin fibroblast cells as well as the approved drug acyclovir did. The compounds had 50% effective concentration values as low as 0.22 µM with negligible cytotoxicity in the assays employed. The inhibitors suppressed accumulation of viral genomes and infectious particles and blocked events in the viral replication cycle before and during viral DNA replication. Acyclovir-resistant mutants of HSV-1 and HSV-2 remained highly sensitive to the NTS inhibitors. Five of six NTS inhibitors of the HSVs also blocked replication of another herpesvirus pathogen, human cytomegalovirus. Therefore, NTS enzyme inhibitors are promising candidates for new herpesvirus treatments that may have broad efficacy against members of the herpesvirus family.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Nucleotidyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Acyclovir/pharmacology , Animals , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/enzymology , Cytomegalovirus/growth & development , DNA Replication/drug effects , DNA, Viral/antagonists & inhibitors , DNA, Viral/genetics , DNA, Viral/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Viral/drug effects , Fibroblasts/drug effects , Fibroblasts/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/growth & development , Herpesvirus 2, Human/enzymology , Herpesvirus 2, Human/growth & development , Humans , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Primary Cell Culture , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
9.
Antiviral Res ; 222: 105799, 2024 02.
Article in English | MEDLINE | ID: mdl-38190973

ABSTRACT

Adenovirus infections of immunocompromised humans are a significant source of morbidity and mortality. Presently, there is no drug specifically approved for the treatment of adenovirus infections by the FDA. The state-of-the-art treatment of such infections is the off-label use of cidofovir, an acyclic nucleotide phosphonate. While cidofovir inhibits adenovirus replication, it has dose-limiting kidney toxicity. There is an apparent need for a better compound to treat adenovirus infections. To this end, we have been developing acyclic nucleotide phosphonate prodrugs that utilize an amino acid scaffold equipped with a lipophilic modifier. Here, we compare the antiviral potential of two prodrugs of HPMPA that differ only in the amino acid-based promoiety: USC-087, based on an N-hexadecyl tyrosinamide, and USC-093, based on an N-hexadecyl serinamide. Oral administration of both compounds was very efficacious against disseminated HAdV-C6 infection in immunosuppressed Syrian hamsters, suppressing virus replication and mitigating pathology even when treatment was withheld until 4 days after challenge. We saw only marginal efficacy after respiratory infection of hamsters, which may reflect suboptimal distribution to the lung. Importantly, neither compound induced intestinal toxicity, which was observed as the major adverse effect in clinical trials of brincidofovir, a prodrug of cidofovir which also contains a C-16 modifier. Notably, we found that there was a significant difference in the nephrotoxicity of the two compounds: USC-087 caused significant kidney toxicity while USC-093 did not, at effective doses. These findings will be valuable guidepoints in the future evolution of this new class of potential prodrugs to treat adenovirus infections.


Subject(s)
Adenine/analogs & derivatives , Adenoviridae Infections , Adenovirus Infections, Human , Organophosphonates , Prodrugs , Tyrosine/analogs & derivatives , Cricetinae , Animals , Humans , Adenovirus Infections, Human/drug therapy , Cidofovir/pharmacology , Cidofovir/therapeutic use , Mesocricetus , Antiviral Agents/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , Adenoviridae , Virus Replication , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Adenoviridae Infections/drug therapy , Cytosine/pharmacology , Cytosine/therapeutic use , Amino Acids/pharmacology , Nucleotides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL