Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Antiviral Res ; 84(2): 131-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19665486

ABSTRACT

Alternative therapies are needed for HSV-1 infections in patients refractory to treatment with Acyclovir (ACV) and its derivatives. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are single-stranded DNA analogues that enter cells readily and reduce target gene expression through steric blockage of complementary RNA. When applied before or soon after infection PPMO targeting the translation-start-site regions of HSV-1 ICP0 or ICP27 mRNA reduced HSV-1 plaque formation by 70-98% in vitro. The ICP0 PPMO also reduced ACV-resistant HSV-1 (strain 615.9) plaque formation by 70-90%, while an equivalent dose of ACV produced only 40-50% inhibition when the treatment was applied between 1 and 3hpi. Seven daily topical treatments of 100microg ICP0 PPMO caused no gross or microscopic damage to the corneas of uninfected mice. Topical application of 10microg ICP0 PPMO to the eyes of HSV-1 infected mice reduced the incidence of eye disease by 37.5-50% compared to controls. This study demonstrates that topically applied PPMO holds promise as an antiviral drug candidate against HSV-1 ocular infection.


Subject(s)
Antiviral Agents/therapeutic use , Herpesvirus 1, Human/drug effects , Immediate-Early Proteins/drug effects , Keratitis, Herpetic/drug therapy , Morpholines/therapeutic use , Ubiquitin-Protein Ligases/drug effects , Acyclovir/pharmacology , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Base Sequence , Chlorocebus aethiops , Drug Resistance, Viral , Herpesvirus 1, Human/physiology , Humans , Keratitis, Herpetic/virology , Mice , Molecular Sequence Data , Morpholines/adverse effects , Morpholines/chemical synthesis , Morpholines/chemistry , Morpholinos , Vero Cells , Viral Proteins/drug effects , Virus Replication/drug effects
2.
Antimicrob Agents Chemother ; 53(5): 2089-99, 2009 May.
Article in English | MEDLINE | ID: mdl-19223614

ABSTRACT

Phosphorodiamidate morpholino oligomers (PMOs) are uncharged nucleic acid-like molecules designed to inactivate the expression of specific genes via the antisense-based steric hindrance of mRNA translation. PMOs have been successful at knocking out viral gene expression and replication in the case of acute viral infections in animal models and have been well tolerated in human clinical trials. We propose that antisense PMOs represent a promising class of therapeutic agents that may be useful for combating filoviral infections. We have previously shown that mice treated with a PMO whose sequence is complementary to a region spanning the start codon of VP24 mRNA were protected against lethal Ebola virus challenge. In the present study, we report on the abilities of two additional VP24-specific PMOs to reduce the cell-free translation of a VP24 reporter, to inhibit the in vitro replication of Ebola virus, and to protect mice against lethal challenge when the PMOs are delivered prior to infection. Additionally, structure-activity relationship evaluations were conducted to assess the enhancement of antiviral efficacy associated with PMO chemical modifications that included conjugation with peptides of various lengths and compositions, positioning of conjugated peptides to either the 5' or the 3' terminus, and the conferring of charge modifications by the addition of piperazine moieties. Conjugation with arginine-rich peptides greatly enhanced the antiviral efficacy of VP24-specific PMOs in infected cells and mice during lethal Ebola virus challenge.


Subject(s)
Antiviral Agents , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Morpholines , Oligonucleotides, Antisense , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Disease Models, Animal , Dose-Response Relationship, Drug , Ebolavirus/genetics , Ebolavirus/physiology , Female , Hemorrhagic Fever, Ebola/virology , Humans , Male , Mice , Mice, Inbred C57BL , Morpholines/chemistry , Morpholines/pharmacology , Morpholines/therapeutic use , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Rabbits , Structure-Activity Relationship , Treatment Outcome , Vero Cells , Virus Replication/drug effects
3.
J Antimicrob Chemother ; 62(3): 555-65, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18567576

ABSTRACT

OBJECTIVES: To determine the antiviral activity of phosphorodiamidate morpholino oligomers (PMO) and peptide-conjugated PMO (PPMO) in AG129 mice infected with dengue 2 virus (DENV-2). METHODS: Antisense PMO and PPMO were designed against the 5' terminal region (5'SL) or the 3'-cyclization sequence region (3'CS) of DENV genomic RNA and administered to AG129 mice before and/or after infection with DENV-2. In addition, cell culture evaluations designed to determine optimum PPMO length, and pharmacokinetic and toxicity analysis of PPMO were also carried out. RESULTS: Mock-treated AG129 mice lived for 9-17 days following intraperitoneal (ip) infection with 10(4)-10(6) pfu of DENV-2 (strain New Guinea C). Intraperitoneal administration of 5'SL or 3'CS PPMO before and after DENV infection produced an increase in the average survival time of up to 8 days. Animals receiving only post-infection PPMO treatment did not benefit significantly. Cell culture studies showed that PPMO of 22-24 bases long produced substantially higher DENV titre reductions than did PPMO that were either shorter or longer. Pharmacokinetic and toxicology analysis with non-infected animals showed that nine consecutive once-daily ip treatments of 10 mg/kg PPMO resulted in high concentrations of PPMO in the liver and caused little impact on overall health. CONCLUSIONS: The data indicate that PPMO had considerable antiviral efficacy against DENV-2 in the AG129 mouse model and that PPMO treatment early in the course of an infection was critical to extending the survival times of DENV-2-infected mice in the AG129 model system.


Subject(s)
Antiviral Agents/therapeutic use , Dengue/drug therapy , Morpholines/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Animals , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Body Weight , Dengue Virus/drug effects , Dengue Virus/genetics , Injections, Intraperitoneal , Liver/chemistry , Mice , Morpholines/adverse effects , Morpholines/pharmacokinetics , Morpholines/pharmacology , Morpholinos , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Plasma/chemistry , Survival Analysis , Viral Plaque Assay
4.
Virology ; 376(2): 357-70, 2008 Jul 05.
Article in English | MEDLINE | ID: mdl-18468653

ABSTRACT

The genus Alphavirus contains members that threaten human health, both as natural pathogens and as potential biological weapons. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) enter cells readily and can inhibit viral replication through sequence-specific steric blockade of viral RNA. Sindbis virus (SINV) has low pathogenicity in humans and is regularly utilized as a model alphavirus. PPMO targeting the 5'-terminal and AUG translation start site regions of the SINV genome blocked the production of infectious SINV in tissue culture. PPMO designed against corresponding regions in Venezuelan equine encephalitis virus (VEEV) were likewise found to be effective in vitro against several strains of VEEV. Mice treated with PPMO before and after VEEV infection were completely protected from lethal outcome while mice receiving only post-infection PPMO treatment were partially protected. Levels of virus in tissue samples correlated with animal survival. Uninfected mice suffered no apparent ill-effects from PPMO treatment. Thus, PPMO appear promising as candidates for therapeutic development against alphaviruses.


Subject(s)
Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Alphavirus/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Morpholines/pharmacology , Morpholines/therapeutic use , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Administration, Intranasal , Alphavirus/genetics , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Design , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalomyelitis, Venezuelan Equine/prevention & control , Injections, Subcutaneous , Mice , Morpholinos , Sindbis Virus/drug effects , Sindbis Virus/physiology , Virus Replication
5.
J Virol ; 80(23): 11510-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987987

ABSTRACT

Coxsackievirus B3 (CVB3) is a primary cause of viral myocarditis, yet no effective therapeutic against CVB3 is available. Nucleic acid-based interventional strategies against various viruses, including CVB3, have shown promise experimentally, but limited stability and inefficient delivery in vivo remain as obstacles to their potential as therapeutics. We employed phosphorodiamidate morpholino oligomers (PMO) conjugated to a cell-penetrating arginine-rich peptide, P007 (to form PPMO), to address these issues. Eight CVB3-specific PPMO were evaluated with HeLa cells and HL-1 cardiomyocytes in culture and in a murine infection model. One of the PPMO (PPMO-6), designed to target a sequence in the 3' portion of the CVB3 internal ribosomal entry site, was found to be especially potent against CVB3. Treatment of cells with PPMO-6 prior to CVB3 infection produced an approximately 3-log(10) decrease in viral titer and largely protected cells from a virus-induced cytopathic effect. A similar antiviral effect was observed when PPMO-6 treatment began shortly after the virus infection period. A/J mice receiving intravenous administration of PPMO-6 once prior to and once after CVB3 infection showed an approximately 2-log(10)-decreased viral titer in the myocardium at 7 days postinfection and a significantly decreased level of cardiac tissue damage, compared to the controls. Thus, PPMO-6 provided potent inhibition of CVB3 amplification both in cell cultures and in vivo and appears worthy of further evaluation as a candidate for clinical development.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus B, Human/drug effects , Morpholines/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacokinetics , Cell Culture Techniques , Enterovirus B, Human/genetics , Enterovirus B, Human/physiology , Mice , Morpholines/chemistry , Myocarditis/drug therapy , Myocarditis/pathology , Myocarditis/virology , Oligodeoxyribonucleotides, Antisense/genetics , Peptides/metabolism
6.
PLoS Pathog ; 2(1): e1, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16415982

ABSTRACT

The filoviruses Marburg virus and Ebola virus (EBOV) quickly outpace host immune responses and cause hemorrhagic fever, resulting in case fatality rates as high as 90% in humans and nearly 100% in nonhuman primates. The development of an effective therapeutic for EBOV is a daunting public health challenge and is hampered by a paucity of knowledge regarding filovirus pathogenesis. This report describes a successful strategy for interfering with EBOV infection using antisense phosphorodiamidate morpholino oligomers (PMOs). A combination of EBOV-specific PMOs targeting sequences of viral mRNAs for the viral proteins (VPs) VP24, VP35, and RNA polymerase L protected rodents in both pre- and post-exposure therapeutic regimens. In a prophylactic proof-of-principal trial, the PMOs also protected 75% of rhesus macaques from lethal EBOV infection. The work described here may contribute to development of designer, "druggable" countermeasures for filoviruses and other microbial pathogens.


Subject(s)
Antisense Elements (Genetics)/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/prevention & control , Morpholines/pharmacology , Animals , Ebolavirus/genetics , Ebolavirus/physiology , Female , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Morpholinos , Protein Biosynthesis/drug effects , RNA, Viral/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins , Virus Replication/drug effects
7.
Virol J ; 2: 91, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16336686

ABSTRACT

L1R, a myristylated late gene product of vaccinia virus, is essential for formation of infectious intracellular mature virions (IMV). In its absence, only viral particles arrested at an immature stage are detected and no infectious progeny virus is produced. Previous studies have shown that the L1R protein is exclusively associated with the IMV membrane and that myristylation is required for correct targeting. The L1R protein contains six cysteine amino acid residues that have all been shown to participate in intramolecular disulphide bonds. However, it was not clear what role, if any, the disulfide bonds play in the membrane topology of the L1R protein. To address this question, a comprehensive library of L1R mutants in which the cysteine residues have been mutated to serine (either individually or in combination) were tested for their ability to rescue a L1R conditional lethal mutant virus under non-permissive conditions. Much to our surprise, we determined that C57 was not essential for production of infectious IMV. These results suggest that protein disulphide isomerases may be involved in reorganization of disulfide bonds within the L1R protein.


Subject(s)
Disulfides/metabolism , Vaccinia virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Cell Line , Gene Expression Regulation, Viral , Humans , Mutation , Protein Conformation , Vaccinia virus/genetics , Viral Envelope Proteins/genetics , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...