Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 149: 1-6, 2018 01.
Article in English | MEDLINE | ID: mdl-29113740

ABSTRACT

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.


Subject(s)
Acyclovir/pharmacology , Antiviral Agents/pharmacology , Encephalitis, Herpes Simplex/virology , Pyridines/pharmacology , Thiazoles/pharmacology , Acyclovir/administration & dosage , Acyclovir/pharmacokinetics , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Disease Models, Animal , Drug Therapy, Combination , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/mortality , Encephalitis, Herpes Simplex/pathology , Female , Humans , Mice , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Sulfonamides , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Tissue Distribution , Treatment Outcome
2.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25473037

ABSTRACT

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Subject(s)
Epigenesis, Genetic , Herpesviridae Infections/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/physiology , Animals , Disease Models, Animal , Female , Genome, Viral , Guinea Pigs , Histone Demethylases , Mice , Mice, Inbred BALB C , Monoamine Oxidase Inhibitors/chemistry , Phenotype , Protein Structure, Tertiary , Rabbits , Recurrence , Tranylcypromine/chemistry , Vagina/virology , Virus Activation , Virus Latency , Virus Replication/drug effects , Virus Shedding
3.
Int Trends Immun ; 2(2): 87-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-25215309

ABSTRACT

Natural serine protease inhibitors (serpins) elicit sensing of a microbial cell intruder and activate an intrinsic cellular immune response in HIV and HCV infected cells. Here, we demonstrate in vitro inhibition of HSV with serpin antithrombin III (ATIII) early during infection pointing towards inhibition of an entry event. We also found reduction of mortality from 90% to 40% in an abrasion mice model demonstrating a strong reduction of infection in vivo. Our data also indicated that this treatment might be suitable for drug-resistant viruses since high inhibition of an acyclovir-resistant HSV-1 strain was found. Thus, an ATIII tropical treatment might be used for immunocompromised patients where prolonged treatment leads to drug resistant HSV-1 strains. Understanding how ATIII regulates HSV-1 infections may reveal new avenues for therapeutic interventions.

4.
mBio ; 4(1): e00558-12, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23386436

ABSTRACT

Cellular processes requiring access to the DNA genome are regulated by an overlay of epigenetic modifications, including histone modification and chromatin remodeling. Similar to the cellular host, many nuclear DNA viruses that depend upon the host cell's transcriptional machinery are also subject to the regulatory impact of chromatin assembly and modification. Infection of cells with alphaherpesviruses (herpes simplex virus [HSV] and varicella-zoster virus [VZV]) results in the deposition of nucleosomes bearing repressive histone H3K9 methylation on the viral genome. This repressive state is modulated by the recruitment of a cellular coactivator complex containing the histone H3K9 demethylase LSD1 to the viral immediate-early (IE) gene promoters. Inhibition of the activity of this enzyme results in increased repressive chromatin assembly and suppression of viral gene expression during lytic infection as well as reactivation from latency in a mouse ganglion explant model. However, available small-molecule LSD1 inhibitors are not originally designed to inhibit LSD1, but rather monoamine oxidases (MAO) in general. Thus, their specificity for and potency to LSD1 is low. In this study, a novel specific LSD1 inhibitor was identified that potently repressed HSV IE gene expression, genome replication, and reactivation from latency. Importantly, the inhibitor also suppressed primary infection of HSV in vivo in a mouse model. Based on common control of a number of DNA viruses by epigenetic modulation, it was also demonstrated that this LSD1 inhibitor blocks initial gene expression of the human cytomegalovirus and adenovirus type 5. IMPORTANCE Epigenetic mechanisms, including histone modification and chromatin remodeling, play important regulatory roles in all cellular processes requiring access to the genome. These mechanisms are often altered in disease conditions, including various cancers, and thus represent novel targets for drugs. Similarly, many viral pathogens are regulated by an epigenetic overlay that determines the outcome of infection. Therefore, these epigenetic targets also represent novel antiviral targets. Here, a novel inhibitor was identified with high specificity and potency for the histone demethylase LSD1, a critical component of the herpes simplex virus (HSV) gene expression paradigm. This inhibitor was demonstrated to have potent antiviral potential in both cultured cells and animal models. Thus, in addition to clearly demonstrating the critical role of LSD1 in regulation of HSV infection, as well as other DNA viruses, the data extends the therapeutic potential of chromatin modulation inhibitors from the focused field of oncology to the arena of antiviral agents.


Subject(s)
Antiviral Agents/metabolism , Histone Demethylases/antagonists & inhibitors , Host-Pathogen Interactions/drug effects , Simplexvirus/drug effects , Simplexvirus/physiology , Virus Activation/drug effects , Virus Replication/drug effects , Adenoviridae/drug effects , Adenoviridae/physiology , Animals , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Disease Models, Animal , Female , Herpes Simplex/drug therapy , Humans , Mice , Mice, Inbred BALB C , Simplexvirus/growth & development , Viral Load
5.
J Virol ; 86(9): 5304-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22379082

ABSTRACT

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Subject(s)
Genetic Vectors/genetics , Interleukin-12/genetics , Simplexvirus/genetics , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Aotidae , Brain/pathology , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Female , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Glioma/genetics , Glioma/mortality , Glioma/therapy , Humans , Interleukin-12/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, SCID , Simplexvirus/drug effects , Survival Analysis , Virus Replication/drug effects , Virus Replication/genetics , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem ; 20(8): 2669-74, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22417649

ABSTRACT

Synthesis of 6-deoxycyclopropavir (10), a prodrug of cyclopropavir (1) and its in vitro and in vivo antiviral activity is described. 2-Amino-6-chloropurine methylenecyclopropane 13 was transformed to its 6-iodo derivative 14 which was reduced to prodrug 10. It is converted to cyclopropavir (1) by the action of xanthine oxidase and this reaction can also occur in vivo. Compound 10 lacked significant in vitro activity against human cytomegalovirus (HCMV), human herpes virus 1 and 2 (HSV-1 and HSV-2), human immunodeficiency virus type 1 (HIV-1), human hepatitis B virus (HBV), Epstein-Barr virus (EBV), vaccinia virus and cowpox virus. In contrast, prodrug 10 given orally was as active as cyclopropavir (1) reported previously [Kern, E. R.; Bidanset, D. J.; Hartline, C. B.; Yan, Z.; Zemlicka, J.; Quenelle, D. C. et al. Antimicrob. Agents Chemother. 2004, 48, 4745] against murine cytomegalovirus (MCMV) infection in mice and against HCMV in severe combined immunodeficient (SCID) mice.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cyclopropanes/chemical synthesis , Cyclopropanes/pharmacology , Guanine/analogs & derivatives , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Cowpox virus/drug effects , Cyclopropanes/chemistry , Cytomegalovirus/drug effects , Guanine/chemical synthesis , Guanine/chemistry , Guanine/pharmacology , HIV-1/drug effects , Hepatitis B virus/drug effects , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Herpesvirus 4, Human/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Microbial Sensitivity Tests , Molecular Structure , Prodrugs/administration & dosage , Vaccinia virus/drug effects
7.
Antivir Chem Chemother ; 22(3): 131-7, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22095521

ABSTRACT

BACKGROUND: N-methanocarbathymidine (N-MCT) has previously been shown to be effective against lethal orthopoxvirus and herpes simplex virus type-1 infections in mice. In this investigation, the antiviral activity of N-MCT was assessed against herpes simplex virus type-2 (HSV-2) in BALB/c mice. METHODS: BALB/c mice were infected intranasally with a lethal challenge dose of HSV-2. N-MCT was administered orally twice daily to mice using doses of 0.01 to 100 mg/kg to determine effects on survival and on viral replication in organ and central nervous system (CNS) samples. RESULTS: N-MCT provided significant protection from mortality even when treatments were delayed until 3 days after viral infection. Viral replication in organ and CNS samples from N-MCT-treated mice was reduced below the limit of detection after 4 days of treatment. CONCLUSIONS: These results indicated that low dose N-MCT treatment was more effective than acyclovir therapy. N-MCT may be effective against HSV disease in humans and is currently undergoing preclinical evaluation. In particular, its potential use as a combination therapy for HSV, with its differing metabolism from acyclovir, make it a promising compound to develop for human use.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Herpes Simplex/drug therapy , Herpesvirus 2, Human/drug effects , Thymidine/analogs & derivatives , Administration, Oral , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Herpes Simplex/virology , Humans , Mice , Mice, Inbred BALB C , Survival Rate , Thymidine/administration & dosage , Thymidine/pharmacology
8.
Antimicrob Agents Chemother ; 55(10): 4728-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21788472

ABSTRACT

Although acyclovir (ACV) has proven to be of value in the therapy of certain herpes simplex virus (HSV) infections, there is a need for more effective therapies, particularly for serious infections in neonates and immunocompromised individuals, where resistance to this drug can be problematic. CMX001 is an orally bioavailable lipid conjugate of cidofovir that is substantially less nephrotoxic than the parent drug and has excellent antiviral activity against all the human herpesviruses. This compound retains full antiviral activity against ACV-resistant laboratory and clinical isolates. The combined efficacy of CMX001 and ACV was evaluated in a new real-time PCR combination assay, which demonstrated that the combination synergistically inhibited the replication of HSV in cell culture. This was also confirmed in murine models of HSV infection, where the combined therapy with these two drugs synergistically reduced mortality. These results suggest that CMX001 may be effective in the treatment of ACV-resistant HSV infections and as an adjunct therapy in individuals with suboptimal responses to ACV.


Subject(s)
Acyclovir/pharmacology , Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Organophosphonates/pharmacology , Acyclovir/therapeutic use , Acyclovir/toxicity , Animals , Antiviral Agents/therapeutic use , Antiviral Agents/toxicity , Cells, Cultured , Cytosine/pharmacology , Cytosine/therapeutic use , Cytosine/toxicity , Drug Resistance, Viral , Drug Synergism , Drug Therapy, Combination , Female , Herpes Simplex/virology , Humans , Mice , Mice, Inbred BALB C , Organophosphonates/therapeutic use , Organophosphonates/toxicity
9.
J Infect Dis ; 202(10): 1492-9, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20923374

ABSTRACT

CMX001, an orally active lipid conjugate of cidofovir, is 50 times more active in vitro against herpes simplex virus (HSV) replication than acyclovir or cidofovir. These studies compared the efficacy of CMX001 to acyclovir in BALB/c mice inoculated intranasally with HSV types 1 or 2. CMX001 was effective in reducing mortality using doses of 5 to 1.25 mg/kg administered orally once daily, even when treatments were delayed 48-72 h post viral inoculation. Organ samples obtained from mice treated with CMX001 had titers 3-5 log(10) plaque-forming units per gram of tissue lower than samples obtained from mice treated with acyclovir, including 5 different regions of the brain. Detectable concentrations of drug-related radioactivity were documented in the central nervous system of mice after oral administration of (14)C-CMX001. These studies indicate that CMX001 penetrates the blood-brain barrier, is a potent inhibitor of HSV replication in disseminated infections and central nervous system infections, and is superior to acyclovir.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Cytosine/analogs & derivatives , Herpes Simplex/drug therapy , Herpesvirus 1, Human , Herpesvirus 2, Human , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Administration, Oral , Animals , Cytosine/administration & dosage , Cytosine/pharmacokinetics , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Mice , Mice, Inbred BALB C , Tissue Distribution
10.
Viruses ; 2(12): 2681-95, 2010 12.
Article in English | MEDLINE | ID: mdl-21994637

ABSTRACT

Although a large number of compounds have been identified with antiviral activity against orthopoxviruses in tissue culture systems, it is highly preferred that these compounds have activity in vivo before they can be seriously considered for further development. One of the most commonly used animal models for the confirmation of this activity has been the use of mice infected with either vaccinia or cowpox viruses. These model systems have the advantage that they are relatively inexpensive, readily available and do not require any special containment facilities; therefore, relatively large numbers of compounds can be evaluated in vivo for their activity. The two antiviral agents that have progressed from preclinical studies to human safety trials for the treatment of orthopoxvirus infections are the cidofovir analog, CMX001, and an inhibitor of extracellular virus formation, ST-246. These compounds are the ones most likely to be used in the event of a bioterror attack. The purpose of this communication is to review the advantages and disadvantages of using mice infected with vaccinia and cowpox virus as surrogate models for human orthopoxvirus infections and to summarize the activity of CMX001 and ST-246 in these model infections.

11.
Antimicrob Agents Chemother ; 53(12): 5251-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770274

ABSTRACT

A series of 4'-thionucleosides were synthesized and evaluated for activities against orthopoxviruses and herpesviruses. We reported previously that one analog, 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU), exhibits good activity both in vitro and in vivo against two orthopoxviruses. This compound also has good activity in cell culture against many of the herpesviruses. It inhibited the replication of herpes simplex virus type 1 (HSV-1), HSV-2, and varicella-zoster virus with 50% effective concentrations (EC(50)s) of 0.1, 0.5, and 2 microM, respectively. It also inhibited the replication of human cytomegalovirus (HCMV) with an EC(50) of 5.9 microM but did not selectively inhibit Epstein-Barr virus, human herpesvirus 6, or human herpesvirus 8. While acyclovir-resistant strains of HSV-1 and HSV-2 were comparatively resistant to 4'-thioIDU, it retained modest activity (EC(50)s of 4 to 12 microM) against these strains. Some ganciclovir-resistant strains of HCMV also exhibited reduced susceptibilities to the compound, which appeared to be related to the specific mutations in the DNA polymerase, consistent with the observed incorporation of the compound into viral DNA. The activity of 4'-thioIDU was also evaluated using mice infected intranasally with the MS strain of HSV-2. Although there was no decrease in final mortality rates, the mean length of survival after inoculation increased significantly (P < 0.05) for all animals receiving 4'-thioIDU. The findings from the studies presented here suggest that 4'-thioIDU is a good inhibitor of some herpesviruses, as well as orthopoxviruses, and this class of compounds warrants further study as a therapy for infections with these viruses.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Herpesviridae Infections/drug therapy , Herpesviridae/drug effects , Pyrimidine Nucleosides/pharmacology , Pyrimidine Nucleosides/therapeutic use , Virus Replication/drug effects , Animals , Antiviral Agents/adverse effects , Antiviral Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/physiology , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Fluorescent Antibody Technique, Indirect , Herpesviridae/genetics , Herpesviridae Infections/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/genetics , Herpesvirus 6, Human/drug effects , Herpesvirus 6, Human/genetics , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Molecular Structure , Pyrimidine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/chemistry , Viral Proteins/genetics , Viral Proteins/physiology
12.
Antimicrob Agents Chemother ; 53(2): 572-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19029322

ABSTRACT

As part of a program to identify new compounds that have activity against orthopoxviruses, a number of 4'-thionucleosides were synthesized and evaluated for their efficacies against vaccinia and cowpox viruses. Seven compounds that were active at about 1 microM against both viruses in human cells but that did not have significant toxicity were identified. The 5-iodo analog, 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU), was selected as a representative molecule; and this compound also inhibited viral DNA synthesis at less than 1 microM but only partially inhibited the replication of a recombinant vaccinia virus that lacked a thymidine kinase. This compound retained complete activity against cidofovir- and ST-246-resistant mutants. To determine if this analog had activity in an animal model, mice were infected intranasally with vaccinia or cowpox virus and treatment with 4'-thioIDU was given intraperitoneally or orally twice daily at 50, 15, 5, or 1.5 mg/kg of body weight beginning at 24 to 120 h postinfection and was continued for 5 days. Almost complete protection (87%) was observed when treatment with 1.5 mg/kg was begun at 72 h postinfection, and significant protection (73%) was still obtained when treatment with 5 mg/kg was initiated at 96 h. Virus titers in the liver, spleen, and kidney were reduced by about 4 log(10) units and about 2 log(10) units in mice infected with vaccinia virus and cowpox virus, respectively. These results indicate that 4'-thioIDU is a potent, nontoxic inhibitor of orthopoxvirus replication in cell culture and experimental animal infections and suggest that it may have potential for use in the treatment of orthopoxvirus infections in animals and humans.


Subject(s)
Antiviral Agents/therapeutic use , Nucleosides/therapeutic use , Orthopoxvirus , Poxviridae Infections/drug therapy , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cowpox/drug therapy , Cytopathogenic Effect, Viral/drug effects , DNA, Viral/biosynthesis , DNA, Viral/genetics , Drug Resistance, Viral , Humans , Mice , Mice, Inbred BALB C , Poxviridae Infections/virology , Structure-Activity Relationship , Vaccinia/drug therapy , beta-Galactosidase/metabolism
13.
Antiviral Res ; 80(2): 223-4, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18573279

ABSTRACT

These studies were performed to determine the effect of AD-472, an attenuated human herpes simplex virus (HSV) type 2 or HSV-2 glycoprotein D (gD) when combined with an adjuvant, GPI-0100, a semi-synthetic Quillaja Saponin analog in a genital HSV-2 infection in guinea pigs. While animals immunized with either vaccine had reduced clinical disease, GPI-0100 only improved the efficacy of gD and did not affect the efficacy of the live vaccine. Neither vaccine had any therapeutic effect if administered 24 h after viral infection.


Subject(s)
Adjuvants, Immunologic , Herpes Genitalis/prevention & control , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/immunology , Saponins/immunology , Vaccination , Vaccines, Attenuated/immunology , Viral Envelope Proteins/immunology , Animals , Female , Guinea Pigs , Herpes Genitalis/immunology , Humans
14.
Virol J ; 5: 58, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18479513

ABSTRACT

BACKGROUND: The emergence of drug resistant viruses, together with the possibility of increased virulence, is an important concern in the development of new antiviral compounds. Cidofovir (CDV) is a phosphonate nucleotide that is approved for use against cytomegalovirus retinitis and for the emergency treatment of smallpox or complications following vaccination. One mode of action for CDV has been demonstrated to be the inhibition of the viral DNA polymerase. RESULTS: We have isolated several CDV resistant (CDVR) vaccinia viruses through a one step process, two of which have unique single mutations within the DNA polymerase. An additional resistant virus isolate provides evidence of a second site mutation within the genome involved in CDV resistance. The CDVR viruses were 3-7 fold more resistant to the drug than the parental viruses. The virulence of the CDVR viruses was tested in mice inoculated intranasally and all were found to be attenuated. CONCLUSION: Resistance to CDV in vaccinia virus can be conferred individually by at least two different mutations within the DNA polymerase gene. Additional genes may be involved. This one step approach for isolating resistant viruses without serial passage and in the presence of low doses of drug minimizes unintended secondary mutations and is applicable to other potential antiviral agents.


Subject(s)
Cytosine/analogs & derivatives , Drug Resistance, Viral , Organophosphonates/pharmacology , Vaccinia virus/drug effects , Vaccinia virus/isolation & purification , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Cidofovir , Cytosine/pharmacology , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Female , Humans , Mice , Models, Molecular , Mutation , Vaccinia/mortality , Vaccinia/virology , Vaccinia virus/pathogenicity , Vaccinia virus/physiology , Vero Cells , Viral Plaque Assay , Virulence
15.
Antiviral Res ; 79(2): 133-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18336926

ABSTRACT

We utilized BALB/c mice infected with murine CMV (MCMV) or severe combined immunodeficient (SCID) mice implanted with human fetal tissue and infected with HCMV to determine the efficacy of (S)-9-[3-hydroxy-2-(phophonomethoxy)propyl]adenine ((S)-HPMPA), hexadecyloxypropyl-(S)-HPMPA (HDP-(S)-HPMPA) or octadecyloxyethyl-(S)-HPMPA (ODE-(S)-HPMPA). In MCMV-infected BALB/c mice, oral HDP-(S)-HPMPA at 30 mg/kg significantly reduced mortality when started 24-48 h post inoculation. In the experimental HCMV infection, oral administration of vehicle or 10mg/kg of (S)-HPMPA, HDP-(S)-HPMPA or ODE-(S)-HPMPA was initiated 24h after infection and continued for 28 consecutive days. Cidofovir (CDV), at 20mg/kg given i.p., was used as a positive control. HDP-(S)-HPMPA or ODE-(S)-HPMPA significantly reduced viral replication compared to vehicle-treated mice, while oral (S)-HPMPA was ineffective.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Herpesviridae Infections/drug therapy , Organophosphonates/therapeutic use , Adenine/administration & dosage , Adenine/pharmacology , Adenine/therapeutic use , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Cidofovir , Cytomegalovirus/drug effects , Cytosine/administration & dosage , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Liver/virology , Mice , Mice, Inbred BALB C , Mice, SCID , Muromegalovirus/drug effects , Organophosphonates/administration & dosage , Organophosphonates/pharmacology , Survival Analysis
16.
Virol J ; 5: 39, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18321387

ABSTRACT

BACKGROUND: The vaccinia virus (VV) F2L gene encodes a functional deoxyuridine triphosphatase (dUTPase) that catalyzes the conversion of dUTP to dUMP and is thought to minimize the incorporation of deoxyuridine residues into the viral genome. Previous studies with with a complex, multigene deletion in this virus suggested that the gene was not required for viral replication, but the impact of deleting this gene alone has not been determined in vitro or in vivo. Although the crystal structure for this enzyme has been determined, its potential as a target for antiviral therapy is unclear. RESULTS: The F2L gene was replaced with GFP in the WR strain of VV to assess its effect on viral replication. The resulting virus replicated well in cell culture and its replication kinetics were almost indistinguishable from those of the wt virus and attained similar titers. The virus also appeared to be as pathogenic as the WR strain suggesting that it also replicated well in mice. Cells infected with the dUTPase mutant would be predicted to affect pyrimidine deoxynucleotide pools and might be expected to exhibit altered susceptibility to pyrimidine analogs. The antiviral activity of cidofovir and four thymidine analogs were evaluated both in the mutant and the parent strain of this virus. The dUTPase knockout remained fully susceptible to cidofovir and idoxuridine, but was hypersensitive to the drug (N)-methanocarbathymidine, suggesting that pyrimidine metabolism was altered in cells infected with the mutant virus. The absence of dUTPase should reduce cellular dUMP pools and may result in a reduced conversion to dTMP by thymidylate synthetase or an increased reliance on the salvage of thymidine by the viral thymidine kinase. CONCLUSION: We confirmed that F2L was not required for replication in cell culture and determined that it does not play a significant role on virulence of the virus in intranasally infected mice. The recombinant virus is hypersensitive to (N)-methanocarbathymidine and may reflect metabolic differences in the mutant virus.


Subject(s)
Antiviral Agents/pharmacology , Pyrophosphatases/metabolism , Thymidine/analogs & derivatives , Vaccinia virus/physiology , Vaccinia/virology , Virus Replication/drug effects , Animals , Female , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Pyrimidines/metabolism , Pyrophosphatases/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Thymidine/pharmacology , Vaccinia virus/drug effects , Vaccinia virus/enzymology , Vaccinia virus/pathogenicity , Viral Plaque Assay , Virulence
17.
Antimicrob Agents Chemother ; 51(11): 3940-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17846137

ABSTRACT

We have previously reported that (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, or (S)-HPMPA, is active in vitro against cowpox virus (CV) and vaccinia virus (VV) but is not active orally in animals. However, the ether lipid esters of (S)-HPMPA, hexadecyloxypropyl-[(S)-HPMPA] [HDP-(S)-HPMPA] and octadecyloxyethyl-[(S)-HPMPA] [ODE-(S)-HPMPA], had significantly enhanced activity in vitro and are orally bioavailable in mice. In the current study, HDP-(S)-HPMPA and ODE-(S)-HPMPA were prepared in water and administered once daily by oral gavage to mice at doses of 30, 10, and 3 mg/kg of body weight for 5 days beginning 24, 48, or 72 h after inoculation with CV or VV. Oral HDP-(S)-HPMPA and ODE-(S)-HPMPA were both highly effective (P < 0.001) at preventing mortality due to CV at 30 mg/kg, even when treatments were delayed until up to 72 h postinfection. ODE-(S)-HPMPA or HDP-(S)-HPMPA were also highly effective (P < 0.001) at preventing mortality in mice infected with VV at 30 mg/kg when treatments were delayed until to 48 or 72 h postinfection, respectively. Protection against both viruses was associated with a significant reduction of virus replication in the liver, spleen, and kidney but not in the lung. These data indicate that HDP-(S)-HPMPA and ODE-(S)-HPMPA are active when given orally against lethal CV and VV infections in mice, and further evaluation is warranted to provide additional information on the potential of these orally active compounds for treatment of human orthopoxvirus infection.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Cowpox/drug therapy , Organophosphonates/therapeutic use , Vaccinia virus/drug effects , Adenine/chemistry , Adenine/pharmacokinetics , Adenine/therapeutic use , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Cowpox/virology , Kidney/drug effects , Kidney/metabolism , Kidney/virology , Liver/drug effects , Liver/metabolism , Liver/virology , Lung/drug effects , Lung/metabolism , Lung/virology , Mice , Molecular Structure , Organophosphonates/chemistry , Organophosphonates/pharmacokinetics , Spleen/drug effects , Spleen/metabolism , Spleen/virology
18.
Antimicrob Agents Chemother ; 51(11): 4118-24, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17724153

ABSTRACT

The combination of ST-246 and hexadecyloxypropyl-cidofovir or CMX001 was evaluated for synergistic activity in vitro against vaccinia virus and cowpox virus (CV) and in vivo against CV. In cell culture the combination was highly synergistic against both viruses, and the results suggested that combined treatment with these agents might offer superior efficacy in vivo. For animal models, ST-246 was administered orally with or without CMX001 to mice lethally infected with CV. Treatments began 1, 3, or 6 days postinfection using lower dosages than previously used for single-drug treatment. ST-246 was given at 10, 3, or 1 mg/kg of body weight with or without CMX001 at 3, 1, or 0.3 mg/kg to evaluate potential synergistic interactions. Treatment beginning 6 days post-viral inoculation with ST-246 alone only increased the mean day to death at 10 or 3 mg/kg but had no effect on survival. CMX001 alone also had no effect on survival. When the combination of the two drugs was begun 6 days after viral infection using various dosages of the two, a synergistic reduction in mortality was observed. No evidence of increased toxicity was noted with the combination either in vitro or in vivo. These results indicate that combinations of ST-246 and CMX001 are synergistic both in vitro and in vivo and suggest that combination therapy using ST-246 and CMX001 for treatment of orthopoxvirus disease in humans or animals may provide an additional benefit over the use of the two drugs by themselves.


Subject(s)
Benzamides/pharmacology , Cytosine/analogs & derivatives , Isoindoles/pharmacology , Organophosphonates/pharmacology , Orthopoxvirus/drug effects , Administration, Oral , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Benzamides/administration & dosage , Benzamides/therapeutic use , Cells, Cultured , Chlorocebus aethiops , Cowpox virus/drug effects , Cytosine/pharmacology , Cytosine/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Female , Humans , Isoindoles/administration & dosage , Isoindoles/therapeutic use , Male , Mice , Mice, Inbred BALB C , Organophosphonates/therapeutic use , Poxviridae Infections/drug therapy , Poxviridae Infections/virology , Treatment Outcome , Vaccinia virus/drug effects , Vero Cells , Virus Replication/drug effects
19.
Drug Deliv ; 14(3): 119-27, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17454031

ABSTRACT

Two poly(DL-lactide-co-glycolide) microsphere formulations (A, 10% wt/wt, and B, 23% wt/wt, 1-10 microns) were evaluated for intracellular delivery of rifabutin using the J774 murine and Mono Mac 6 (MM6) human monocytic cell lines. Within 7 days, formulation A released 100% in both cell lines and B released 53 and 67% in the J774 and MM6, respectively. Intracellular release of rifabutin with both formulations caused significant reduction of intracellularly replicating Mycobacterium avium (MAC). In MAC-infected beige mice, formulation B (50 mg, intraperitoneal days 0 and 7) completely eliminated infection by 21 days (p < 0.001), similar to a rifabutin daily oral regimen.


Subject(s)
Antibiotics, Antitubercular/administration & dosage , Antibiotics, Antitubercular/pharmacology , Macrophages/microbiology , Mycobacterium avium-intracellulare Infection/drug therapy , Rifabutin/administration & dosage , Rifabutin/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Chemistry, Pharmaceutical , Colony Count, Microbial , Female , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microspheres , Monocytes/microbiology , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium phlei/drug effects , Phagocytosis/drug effects , Serum Bactericidal Test
20.
Antimicrob Agents Chemother ; 51(2): 689-95, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17116683

ABSTRACT

ST-246 was evaluated for activity against cowpox virus (CV), vaccinia virus (VV), and ectromelia virus (ECTV) and had an in vitro 50% effective concentration (EC50) of 0.48 microM against CV, 0.05 microM against VV, and 0.07 microM against ECTV. The selectivity indices were >208 and >2,000 for CV and VV, respectively. The in vitro antiviral activity of ST-246 was significantly greater than that of cidofovir, which had an EC50 of 41.1 microM against CV and 29.2 microM against VV, with selectivity indices of >7 and >10, respectively. ST-246 administered once daily by oral gavage to mice infected intranasally with CV beginning 4 h or delayed until 72 h postinoculation was highly effective when given for a 14-day duration using 100, 30, or 10 mg/kg of body weight. When 100 mg/kg of ST-246 was administered to VV-infected mice, a duration of 5 days was sufficient to significantly reduce mortality even when treatment was delayed 24 h postinoculation. Viral replication in liver, spleen, and kidney, but not lung, of CV- or VV-infected mice was reduced by ST-246 compared to levels for vehicle-treated mice. When 100 mg/kg of ST-246 was given once daily to mice infected by the intranasal route with ECTV, treatment for 10 days prevented mortality even when treatment was delayed up to 72 h after viral inoculation. Viral replication in target organs of ECTV-infected mice was also reduced.


Subject(s)
Benzamides/administration & dosage , Indoles/administration & dosage , Orthopoxvirus/drug effects , Poxviridae Infections/drug therapy , Administration, Oral , Animals , Disease Models, Animal , Female , Isoindoles , Mice , Organ Specificity , Orthopoxvirus/physiology , Time Factors , Treatment Outcome , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...