Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Antiviral Res ; 199: 105275, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35248614

RÉSUMÉ

Varicella zoster virus (VZV) causes chicken pox and shingles and is prevalent worldwide. Acyclovir and penciclovir (and its prodrugs) are first-line treatments for VZV infections, but they are not highly potent against VZV and resistance may arise in immunocompromised people on long-term therapy. HPMPC (cidofovir) is active against VZV, but cidofovir is not approved for treating VZV diseases, is nephrotoxic, and is not orally bioavailable. Here, we present the synthesis and evaluation of USC-373, a phosphonate prodrug of HPMPC with activity against VZV and other DNA viruses. In cultured fibroblasts, it was potent against VZV Ellen laboratory strain and was not overtly toxic, with EC50 of 4 nM and CC50 of 0.20 µM, producing a selectivity index of 50. In ARPE-19 cells, USC-373 was effective against VZV-ORF57-Luc wild type strain and the acyclovir-resistant isogenic strain. In human skin organ culture, USC-373 formulated in cocoa butter and applied topically prevented VZV-ORF57-Luc spread without toxicity. In NuSkin mice with human skin xenografts, one daily dose of 3 mg/kg was effective by the subcutaneous route, and one daily dose of 10 mg/kg was effective by the oral route. Remarkably, a 10 mg/kg oral dose given every other day was also effective. USC-373 was well tolerated and mice did not lose weight or show signs of distress. The prodrug modifications of USC-373 increase the potency and oral bioavailability compared to its parent nucleoside analog, HPMPC.


Sujet(s)
Phosphonates , Promédicaments , Aciclovir/pharmacologie , Animaux , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Cidofovir/pharmacologie , Herpèsvirus humain de type 3 , Humains , Souris , Techniques de culture d'organes , Phosphonates/pharmacologie , Promédicaments/pharmacologie , Protéines virales régulatrices ou accessoires
2.
Transpl Infect Dis ; 18(5): 785-790, 2016 Oct.
Article de Anglais | MEDLINE | ID: mdl-27481400

RÉSUMÉ

Brincidofovir (BCV) is a broad-spectrum antiviral agent active in vitro against double-stranded DNA viruses including herpesviruses, adenoviruses, polyomaviruses, and poxviruses. We report successful BCV use in management of disseminated acyclovir- and cidofovir-resistant varicella zoster virus in an immunocompromised hematopoietic stem cell transplant patient with chronic graft-versus-host disease who was intolerant to foscarnet.


Sujet(s)
Aciclovir/analogues et dérivés , Antiviraux/usage thérapeutique , Cytosine/analogues et dérivés , Médicaments en essais cliniques/usage thérapeutique , Transplantation de cellules souches hématopoïétiques/effets indésirables , Zona/traitement médicamenteux , Herpèsvirus humain de type 3/physiologie , Leucémie myéloïde chronique BCR-ABL positive/thérapie , Phosphonates/usage thérapeutique , Valine/analogues et dérivés , Aciclovir/administration et posologie , Aciclovir/usage thérapeutique , Adulte , Antibioprophylaxie , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Antiviraux/administration et posologie , Antiviraux/effets indésirables , Cytosine/administration et posologie , Cytosine/effets indésirables , Cytosine/usage thérapeutique , Résistance virale aux médicaments , Médicaments en essais cliniques/administration et posologie , Médicaments en essais cliniques/effets indésirables , Femelle , Foscarnet/administration et posologie , Foscarnet/effets indésirables , Foscarnet/usage thérapeutique , Maladie du greffon contre l'hôte/sang , Maladie du greffon contre l'hôte/traitement médicamenteux , Zona/sang , Zona/virologie , Herpèsvirus humain de type 3/isolement et purification , Humains , Sujet immunodéprimé , Présentation de nouveau médicament de recherche , Phosphonates/administration et posologie , Phosphonates/effets indésirables , Transplantation homologue/effets indésirables , Valaciclovir , Valine/administration et posologie , Valine/usage thérapeutique
3.
Clin Pharmacol Ther ; 97(1): 66-78, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25670384

RÉSUMÉ

The seminal discovery of acyclovir 40 years ago heralded the modern era of truly selective antiviral therapies and this drug remains the therapy of choice for herpes simplex virus infections. Yet by modern standards, its antiviral activity is modest and new drugs against novel molecular targets such as the helicase-primase have the potential to improve clinical outcome, particularly in high-risk patients. A brief synopsis of current therapies for these infections and clinical need is provided to help provide an initial perspective. The function of the helicase-primase complex is then summarized and the development of new inhibitors of the helicase-primase complex, such as pritelivir and amenamevir, is discussed. We review their mechanism of action, propensity for drug resistance, and pharmacokinetic characteristics and discuss their potential to advance current therapeutic options. Strategies that include combinations of these inhibitors with acyclovir are also considered, as they will likely maximize clinical efficacy.


Sujet(s)
Antiviraux/usage thérapeutique , Conception de médicament , Herpès/traitement médicamenteux , Aciclovir/pharmacologie , Aciclovir/usage thérapeutique , Animaux , Antiviraux/pharmacologie , Helicase/antagonistes et inhibiteurs , DNA primase/antagonistes et inhibiteurs , Résistance virale aux médicaments , Herpès/virologie , Humains , Thérapie moléculaire ciblée , Oxadiazoles/pharmacologie , Oxadiazoles/usage thérapeutique , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Sulfonamides , Thiazoles/pharmacologie , Thiazoles/usage thérapeutique , Protéines virales/antagonistes et inhibiteurs
4.
Infect Disord Drug Targets ; 11(5): 504-13, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21827431

RÉSUMÉ

Infections due to resistant human cytomegalovirus (CMV) are an emerging problem, particularly in immunocompromised hosts. When managing such patients, clinicians should be aware of the possibility of developing CMV antiviral resistance, especially while on prolonged therapy or if severe immunosuppression is present. CMV resistance to current antiviral agents is mediated by alterations in either the UL97 kinase or DNA polymerase, encoded by the UL97 and UL54 genes, respectively. UL97 mutations are capable of conferring resistance to ganciclovir, while UL54 mutations can impart resistance to ganciclovir, cidofovir, and foscarnet. If treatment failure is suspected to be due to antiviral resistance, CMV resistance analysis should be obtained. Phenotypic resistance assays performed on clinical isolates measure antiviral susceptibilities directly, but are laborious and time-consuming. Therefore, genotypic resistance analysis has become the more common means of diagnosing CMV resistance. Mutations in UL97 or UL54 may be clinically associated with resistance, but their effect on antiviral susceptibility must be confirmed by marker transfer techniques such as recombinant phenotyping.


Sujet(s)
Antiviraux/pharmacologie , Infections à cytomégalovirus/traitement médicamenteux , Cytomegalovirus/effets des médicaments et des substances chimiques , Animaux , Cytomegalovirus/génétique , Infections à cytomégalovirus/virologie , DNA-directed DNA polymerase/génétique , Résistance virale aux médicaments , Humains , Sujet immunodéprimé , Mutation , Phosphotransferases (Alcohol Group Acceptor)/génétique , Protéines virales/génétique
5.
J Virol ; 75(16): 7592-601, 2001 Aug.
Article de Anglais | MEDLINE | ID: mdl-11462031

RÉSUMÉ

Cytomegalovirus gene UL114, a homolog of mammalian uracil-DNA glycosylase (UNG), is required for efficient viral DNA replication. In quiescent fibroblasts, UNG mutant virus replication is delayed for 48 h and follows the virus-induced expression of cellular UNG. In contrast, mutant virus replication proceeds without delay in actively growing fibroblasts that express host cell UNG. In the absence of viral or host cell UNG expression, mutant virus fails to proceed to late-phase DNA replication, characterized by rapid DNA amplification. The data suggest that uracil incorporated early during wild-type viral DNA replication must be removed by virus or host UNG prior to late-phase amplification and encapsidation into progeny virions. The process of uracil incorporation and excision may introduce strand breaks to facilitate the transition from early-phase replication to late-phase amplification.


Sujet(s)
Cytomegalovirus/physiologie , DNA Glycosylases , N-Glycosyl hydrolases/physiologie , Réplication virale/physiologie , Séquence d'acides aminés , Lignée cellulaire , Réplication de l'ADN , ADN viral , Humains , Données de séquences moléculaires , Alignement de séquences , Uracil-DNA glycosidase
6.
Rev Med Virol ; 11(3): 191-200, 2001.
Article de Anglais | MEDLINE | ID: mdl-11376481

RÉSUMÉ

The complete genetic content of human cytomegalovirus (HCMV) has been difficult to determine, since most strains studied in the laboratory have been extensively passaged in human fibroblast cultures which can change the genetic content as well as the biological properties of the virus. Approximately 13 kb of novel DNA sequences located near the right edge of the unique long (UL) component of the genome has been discovered in Toledo, clinical isolates and certain stocks of Towne. This region of novel sequence, designated the UL/b' region, encodes several interesting proteins including vCXC-1, a potent IL-8 homologue, and UL144, a member of the TNF receptor family. This region is missing from the prototypic laboratory variants of Towne and AD169. In contrast to Toledo and other low passage isolates which have relatively small repeats bracketing the UL component, the Towne and AD169 laboratory variants contain large (>10 kb) b/b' repeats. The large size of these repeats in AD169 and Towne appear to have arisen as compensation for the loss of sequences from the UL/b' region that existed in less passaged variants of these strains. Consequently, many of the haploid genes at the left edge of the prototypic wild-type (wt) UL component are diploid in AD169 and Towne. We hypothesise that this plasticity of the genome at the right edge of the UL component results from extensive passage and adaptation to replication in fibroblasts in vitro. Further work will be required to understand the complete genetic content of wt HCMV.


Sujet(s)
Cytomegalovirus/génétique , Génome viral , Séquence d'acides aminés , Cellules cultivées , Chimiokines CXC/génétique , Cytomegalovirus/croissance et développement , Cytomegalovirus/pathogénicité , Infections à cytomégalovirus/virologie , Fibroblastes , Variation génétique , Humains , Glycoprotéines membranaires/génétique , Données de séquences moléculaires , Alignement de séquences , Protéines virales/génétique , Culture virale , Réplication virale
7.
Proc Natl Acad Sci U S A ; 98(4): 1895-900, 2001 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-11172047

RÉSUMÉ

The human cytomegalovirus UL97 kinase, an important target of antiviral therapy, has an impact on at least two distinct phases of viral replication. Compared with wild-type virus, the UL97 deletion mutant exhibits an early replication defect that reduces DNA accumulation by 4- to 6-fold, as well as a late capsid maturation defect responsible for most of the observed 100- to 1000-fold reduction in replication. Block-release experiments with the antiviral 2-bromo-5,6-dichloro-1-(beta-D-ribofuranosyl)-benzimidazole revealed an important role for UL97 kinase in capsid assembly. Although cleavage of concatemeric DNA intermediates to unit-length genomes remained unaffected, progeny mutant virus maturation was delayed, with accumulation of progeny at significantly reduced levels compared with wild type after release of this block. Transmission electron microscopy confirmed the aberrant accumulation of empty A-like capsids containing neither viral DNA nor an internal scaffold structure, consistent with a failure to stably package DNA in mutant virus-infected cells. The function of UL97 in DNA synthesis as well as capsid assembly suggests that protein phosphorylation mediated by this herpesvirus-conserved kinase increases the efficiency of these two distinct phases of virus replication.


Sujet(s)
Cytomegalovirus/physiologie , Réplication de l'ADN/physiologie , ADN viral/biosynthèse , Phosphotransferases (Alcohol Group Acceptor)/physiologie , Réplication virale/physiologie , Capside/biosynthèse , Cellules cultivées , Cytomegalovirus/génétique , Humains , Protéines virales/métabolisme
8.
J Virol ; 73(7): 5663-70, 1999 Jul.
Article de Anglais | MEDLINE | ID: mdl-10364316

RÉSUMÉ

Human cytomegalovirus encodes a protein kinase (UL97) that confers sensitivity to ganciclovir by phosphorylating it to the monophosphate. The function of this unusual kinase in viral replication is unknown. We constructed two independent isolates of a recombinant virus, RCDelta97, that contain large deletions in this gene and carry a 4.8-kb insertion containing a selectable genetic marker. These mutant viruses were isolated by using a population of primary cells (HEL97) that express this gene from integrated copies of a defective retroviral vector. The recombinant viruses were severely impaired in their ability to replicate in primary fibroblasts, attaining virus titers that were 2 to 3 orders of magnitude lower than those produced by the parent virus. Despite the severe replication deficit, both of these viruses retained the ability to form small, slowly growing plaques in primary fibroblasts, demonstrating that UL97 is not absolutely essential for replication in cell culture. The replication deficit was relieved when UL97 was provided in trans in the complementing cell line, showing that the phenotype was due to a deficiency in UL97. Thus, the UL97 gene product plays a very important role in viral replication in tissue culture and may be a good target for antiviral chemotherapy.


Sujet(s)
Cytomegalovirus/physiologie , Phosphotransferases (Alcohol Group Acceptor)/physiologie , Réplication virale , Animaux , Lignée cellulaire , Cellules cultivées , Cytomegalovirus/génétique , Protéines de liaison à l'ADN/génétique , Expression des gènes , Test de complémentation , Humains , Souris , Cadres ouverts de lecture , Phosphotransferases (Alcohol Group Acceptor)/génétique , Recombinaison génétique , Protéines virales/génétique
9.
J Virol ; 72(9): 6997-7004, 1998 Sep.
Article de Anglais | MEDLINE | ID: mdl-9696791

RÉSUMÉ

Human cytomegalovirus (HCMV) lytic-phase DNA replication initiates at the cis-acting origin of replication, oriLyt. oriLyt is a structurally complex region containing repeat elements and transcription factor binding sites. We identified two site-specific alkali-labile regions within oriLyt which flank an alkali-resistant DNA segment. These alkali-sensitive regions were the result of the degradation of two RNA species embedded within oriLyt and covalently linked to viral DNA. The virus-associated RNA, vRNA, was identified by DNase I treatment of HCMV DNA obtained from sucrose gradient purified virus. This heterogeneous population of vRNA was end labeled and used as a hybridization probe to map the exact location of vRNAs within oriLyt. vRNA-1 is localized between restriction endonuclease sites XhoI at nucleotide (nt) 93799 and SacI at nt 94631 and is approximately 500 bases long. The second vRNA, vRNA-2, lies within a region which exhibits a heterogeneous restriction pattern located between the SphI (nt 92636) and BamHI (nt 93513) and is approximately 300 bases long. This region was previously shown to be required for oriLyt replication (D. G. Anders, M. A. Kacica, G. S. Pari, and S. M. Punturieri, J. Virol. 66:3373-3384, 1992). RNase H analysis determined that vRNA-2 forms a persistent RNA-DNA hybrid structure in the context of the viral genome and in an oriLyt-containing plasmid used in the transient-replication assay.


Sujet(s)
Cytomegalovirus/génétique , ADN viral , ARN viral , Recombinaison génétique , Origine de réplication , Sites de fixation , Lignée cellulaire , Cartographie chromosomique , Humains , Conformation d'acide nucléique , Cartographie de restriction , Hydroxyde de sodium
10.
Virology ; 239(1): 169-75, 1997 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-9426456

RÉSUMÉ

A polymorphism in the UL42-UL43 region of the human cytomegalovirus genome has been characterized by nucleotide sequence analysis, revealing a 929-bp insertion following nt 54,612 relative to the published strain AD169-UK genome sequence (M.S. Chee et al., 1990, Curr. Top. Microbiol Immunol. 154, 125-170). Although AD169-UK exhibited polymorphism in this genomic region, other CMV strains (Towne, Toledo, and AD169-ATCC) carried only the newly characterized longer form. The additional sequence altered the assignment of UL42 and UL43 open reading frames. UL42 decreased in size from 157 to 125 codons, retaining 76 of the previously reported carboxyl terminal codons, and UL43 increased in size from 187 to 423 codons, retaining 185 of the previously reported amino terminal codons. This additional sequence makes UL43 a more conserved betaherpesvirus US22 family member. Only AD169-UK exhibited restriction fragment length polymorphism in this region, suggesting that a deletion occurred during the propagation of this strain in cell culture. The additional sequence should be considered a bona fide part of the cytomegalovirus genome and the AD169 genome size should be corrected to 230,283 bp.


Sujet(s)
Cytomegalovirus/génétique , Génome humain , Polymorphisme génétique , Séquence d'acides aminés , Humains , Données de séquences moléculaires , Analyse de séquence
11.
J Virol ; 70(5): 3018-25, 1996 May.
Article de Anglais | MEDLINE | ID: mdl-8627778

RÉSUMÉ

Human cytomegalovirus (CMV) encodes a gene, UL114, whose product is homologous to the uracil DNA glycosylase and is highly conserved in all herpesviruses. This DNA repair enzyme excises uracil residues in DNA that result from the misincorporation of dUTP or spontaneous deamination of cytosine. We constructed a recombinant virus, RC2620, that contains a large deletion in the UL114 open reading frame and carries a 1.2-kb insert containing the Escherichia coli gpt gene. RC2620 retains the capacity to replicate in primary human fibroblasts and reaches titers that are similar to those produced by the parent virus but exhibits a significantly longer replication cycle. Although the rate of expression of alpha and beta gene products appears to be unaffected by the mutation, DNA synthesis fails to proceed normally. Once initiated, DNA synthesis in mutant virus-infected cells proceeds at the same rate as with wild-type virus, but initiation is delayed by 48 h. The mutant virus also exhibits two predicted phenotypes: (i) hypersensitivity to the nucleoside analog 5-bromodeoxyuridine and (ii) retention of more uracil residues in genomic DNA than the parental virus. Together, these data suggest UL114 is required for the proper excision of uracil residues from viral DNA but in addition plays some role in establishing the correct temporal progression of DNA synthesis and viral replication. Although such involvement has not been previously observed in herpesviruses, a requirement for uracil DNA glycosylase in DNA replication has been observed in poxviruses.


Sujet(s)
Cytomegalovirus/physiologie , DNA Glycosylases , Réparation de l'ADN , Réplication de l'ADN , ADN viral/biosynthèse , N-Glycosyl hydrolases/métabolisme , Réplication virale , Séquence nucléotidique , Broxuridine/toxicité , Division cellulaire , Cellules cultivées , Séquence conservée , Cytomegalovirus/enzymologie , Cytomegalovirus/génétique , Réplication de l'ADN/effets des médicaments et des substances chimiques , Escherichia coli , Fibroblastes , Régulation de l'expression des gènes viraux , Gènes viraux , Herpesviridae/génétique , Humains , Cinétique , Mâle , N-Glycosyl hydrolases/génétique , Cadres ouverts de lecture , Phénotype , Délétion de séquence , Peau , Facteurs temps , Uracil-DNA glycosidase , Réplication virale/effets des médicaments et des substances chimiques
13.
Chemotherapy ; 41(5): 384-95, 1995.
Article de Anglais | MEDLINE | ID: mdl-8521741

RÉSUMÉ

Herpes simplex virus encodes a ribonucleotide reductase that catalyzes the formation of deoxyribonucleotides from ribonucleotides. The enzyme is not essential for either viral DNA synthesis or replication, yet inhibitors of this enzyme suppress viral replication. To clarify the role of the ribonucleotide reductase in virus infection and to evaluate it as an antiviral target, the metabolism of deoxyribonucleotides in infected cells was examined. Our results show that the cellular ribonucleotide reductase is incapable of generating adequate deoxyribonucleoside triphosphate pools to support efficient virus replication. Additionally, we have shown that the virus is unable to efficiently utilize salvaged deoxyribonucleosides from degraded cellular DNA. A selective inhibitor of the viral ribonucleotide reductase, 2-acetylpyridine thiosemicarbazone, decreased deoxyribonucleotide pools in infected cells, thus inhibiting viral DNA synthesis. This compound also inhibited the cellular ribonucleotide reductase to some extent, thereby enhancing its antiviral activity. The antiviral effects of acyclovir were potentiated by 2-acetylpyridine thiosemicarbazone in the wild-type virus but not in the ribonucleotide reductase mutant, ICP6 delta. Collectively, these data strongly suggest that the viral ribonucleotide reductase is an important enzyme in viral replication and a valid target for antiviral chemotherapy.


Sujet(s)
Ribonucleotide reductases/pharmacologie , Simplexvirus/effets des médicaments et des substances chimiques , Aciclovir/pharmacologie , Animaux , Antiviraux , Numération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Dosage immunologique , Macaca mulatta , Réplication virale/effets des médicaments et des substances chimiques
14.
Antiviral Res ; 27(3): 317-23, 1995 Jun.
Article de Anglais | MEDLINE | ID: mdl-8540752

RÉSUMÉ

Ribavirin has been reported to enhance the activity of ddI against HIV. We explored this enhancement of antiviral activity in Rauscher murine leukemia virus (RMuLV) models in vitro and in vivo. The significant finding in these studies was that combinations of the drugs exhibited virus titer reductions that were greater than would be expected if the drug interactions were simply additive. These effects were designated synergistic by the method of Prichard and Shipman (Prichard, M.N. and Shipman, C., Jr. (1990). A three-dimensional model to analyze drug-drug interaction, Antiviral Res. 14, 181-206). In addition to the antiviral synergy, we also observed some synergistic toxicity in the animal model.


Sujet(s)
Didéoxyinosine/pharmacologie , Leucémie expérimentale/traitement médicamenteux , Virus de Rauscher/effets des médicaments et des substances chimiques , Infections à Retroviridae/traitement médicamenteux , Ribavirine/pharmacologie , Infections à virus oncogènes/traitement médicamenteux , Animaux , Lignée cellulaire , Synergie des médicaments , Leucémie expérimentale/virologie , Mâle , Souris , Virus de Rauscher/isolement et purification , Prise de poids
15.
Antimicrob Agents Chemother ; 37(3): 540-5, 1993 Mar.
Article de Anglais | MEDLINE | ID: mdl-8384816

RÉSUMÉ

The development of new drugs effective against human viral diseases has proven to be both difficult and time-consuming. Indeed, there are but 10 drugs licensed for such applications in the United States today. An attractive solution to this problem may be to optimize the efficacy and selectivity of existing antiviral drugs by combining them with agents that strategically block carefully selected metabolic pathways. This approach was used in the rational design of a three-drug combination to increase the apparent potency of acyclovir against herpes simplex virus. Recent advances in analytical techniques have made the evaluation of this complex drug strategy both possible and practical. A modified version of a previously described analytical method was used to identify optimal drug concentrations and to quantitate statistically significant synergy. Concentrations of 0.25 microM 5-fluorodeoxyuridine, 3.6 microM 2-acetylpyridine thiosemicarbazone, and 0.3 microM acyclovir were determined to be optimal in terms of antiviral activity. The volume of synergy produced was nearly 2,000 microM3% at a 95% level of confidence (corresponding to a 186-fold decrease in the apparent 50% inhibitory concentration of acyclovir with the addition of 0.25 microM 5-fluorodeoxyuridine and 3.6 microM 2-acetylpyridine thiosemicarbazone). We anticipate that this strategic approach and the supporting three-dimensional analytical method will prove valuable in designing and understanding multidrug therapies.


Sujet(s)
Antiviraux/pharmacologie , Conception de médicament , Simplexvirus/effets des médicaments et des substances chimiques , Aciclovir/pharmacologie , Animaux , Lignée cellulaire , Chlorocebus aethiops , Association médicamenteuse , Synergie des médicaments , Floxuridine/pharmacologie , Rein , Thiosemicarbazones/pharmacologie
16.
Antiviral Res ; 20(3): 249-59, 1993 Mar.
Article de Anglais | MEDLINE | ID: mdl-8385896

RÉSUMÉ

In cells infected with herpes simplex virus type 1, intracellular dNTP pools increased markedly. Treatment of these cells with 3 microM acyclovir resulted in an additional expansion in pyrimidine deoxyribonucleoside triphosphate pools with dTTP increasing 32-fold and dCTP 8-fold. Both thymidine and deoxycytidine, however, compete with acyclovir for phosphorylation by the viral pyrimidine deoxyribonucleoside kinase and thus reduce the amount of drug that is anabolized to the active form. Theoretically, agents which inhibit thymidylate synthase or dihydrofolate reductase should reduce intracellular pools of thymidine, resulting in the potentiation of the antiviral effects of acyclovir. We explored this strategy by quantitating the synergy produced by combinations of acyclovir and other drugs using three-dimensional dose-response surface methodology (MacSynergy II). Significant synergy was seen with both 5-FdUrd and methotrexate whereas BrVdUrd, 5-CldUrd, 5-IdUrd, and 5-BrdUrd exhibited little to no synergistic activity. It is suggested that inhibitors of thymidylate synthase and dihydrofolate reductase warrant further exploration as potentiators of acyclovir.


Sujet(s)
Aciclovir/pharmacologie , Antiviraux/pharmacologie , Antifoliques , Simplexvirus/effets des médicaments et des substances chimiques , Thymidylate synthase/antagonistes et inhibiteurs , Animaux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Désoxyribonucléosides/métabolisme , Synergie des médicaments , Floxuridine/pharmacologie , Haplorhini , Techniques immunoenzymatiques , Méthotrexate/pharmacologie
17.
Antimicrob Agents Chemother ; 35(6): 1060-5, 1991 Jun.
Article de Anglais | MEDLINE | ID: mdl-1929243

RÉSUMÉ

The combined cytotoxicity of zidovudine and ganciclovir in three cell lines of human origin was examined. The data were generated by a new rapid cell proliferation assay and a more sensitive plating efficiency assay. A three-dimensional analytical approach was used to evaluate the drug-drug interactions, and the results were compared with those obtained by two conventional methods of analysis. Synergistic cytotoxicity was observed in all cell lines examined and by both assays. Moreover, this synergistic cytotoxicity was statistically significant at physiologically relevant concentrations. It is not known whether these drug-drug interactions manifest themselves in vivo as granulocytopenia or other untoward side effects. These results, however, indicate that further investigation is warranted and that the coadministration of zidovudine and ganciclovir may be contraindicated.


Sujet(s)
Survie cellulaire/effets des médicaments et des substances chimiques , Ganciclovir/pharmacologie , Zidovudine/pharmacologie , Division cellulaire/effets des médicaments et des substances chimiques , Évaluation préclinique de médicament , Synergie des médicaments , Humains , Cellules KB/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques
18.
Antiviral Res ; 14(4-5): 181-205, 1990.
Article de Anglais | MEDLINE | ID: mdl-2088205

RÉSUMÉ

Nearly four generations of investigators have studied combined drug effects. Their methods of generating and analyzing data have changed dramatically over the years but the basic problem has not. This review examines the inherent difficulties in analyzing combined drug effects and evaluates modern methods of describing these interactions. Researchers have traditionally used two-dimensional (2-D) methods to approximate the actual three-dimensional (3-D) nature of drug interactions. We conclude that these 2-D methods are often inadequate when used to analyze synergistic and antagonistic drug interactions in antiviral and anticancer chemotherapy. We propose a direct and pragmatic 3-D approach to the problem, made possible by microcomputers and sophisticated graphics programs. This procedure directly elucidates the shape of the dose-response surface, identifies the regions of statistically significant synergy and antagonism, and quantitates these effects. It also greatly simplifies the problem since a 3-D surface presents complete drug interactions in a way that can be easily interpreted. We will show that understanding the shape of the resulting 3-D surface is essential to an understanding of complex drug interactions. This new method facilitates the rigorous analysis of drug-drug interactions and offers investigators powerful new tools to analyze combinations of antiviral and anticancer drugs.


Sujet(s)
Antiviraux , Simulation numérique , Interactions médicamenteuses , Antiviraux/administration et posologie , Relation dose-effet des médicaments , Association médicamenteuse , Modèles biologiques , Conformation moléculaire
19.
J Virol Methods ; 28(1): 101-6, 1990 Apr.
Article de Anglais | MEDLINE | ID: mdl-2161417

RÉSUMÉ

Although the virus yield reduction assay is a powerful technique for evaluating the efficacy of antiviral compounds, it is not routinely utilized due to its labor-intensive nature. This procedure was modified, developed, thereby reducing greatly the time and effort required to perform yield reduction assays. Monolayer cultures of mammalian cells were grown in 96-well microtiter tissue culture plates and infected with virus. Test compounds were added and serially diluted directly with the plates. Following a cycle of virus replication, culture lysates were made and serially diluted in a separate set of uninfected cultures grown in microtiter plates. The cultures were incubated, plaques were enumerated in wells containing 5 to 20 plaques, and virus titers were calculated. To illustrate the use of the assay the known antiviral drugs acyclovir and ganciclovir were evaluated using this procedure. Ninety percent inhibitory concentrations for the respective drugs were 3 microM and 0.7 microM against herpes simplex virus type 1 and 60 microM and 1 microM against human cytomegalovirus.


Sujet(s)
Antiviraux/pharmacologie , Cytomegalovirus/effets des médicaments et des substances chimiques , Simplexvirus/effets des médicaments et des substances chimiques , Aciclovir/pharmacologie , Animaux , Lignée cellulaire , Cytomegalovirus/croissance et développement , Ganciclovir/pharmacologie , Tests de sensibilité microbienne/méthodes , Simplexvirus/croissance et développement , Méthode des plages virales/méthodes
20.
Arch Surg ; 115(5): 597-600, 1980 May.
Article de Anglais | MEDLINE | ID: mdl-7377961

RÉSUMÉ

The effects of isovolemic hemodilution on cardiac output and oxygen transport in 11 patients during elective vascular surgery were evaluated. Mean hemoglobin level was decreased from 12.5 +/- 0.6 to 10.2 +/- 0.5 g/dL by withdrawing blood and replacing it with an equal volume of colloid. Hemodilution increased cardiac output from 4.8 +/- 0.3 to 6.4 +/- 0.4 L/min, increased oxygen delivery from 830 +/- 75 to 900 +/- 95 mL/min and increased oxygen consumption from 190 +/- 20 to 240 +/- 40 mL/min. Systemic vascular resistance and mean arterial blood pressure decreased significantly, but cardiac filling pressure, pulmonary vascular resistance, heart rate, and intrapulmonary shunt did not change. In four of these patients who did not require all their blood during surgery, 1 unit of their withdrawn blood was reinfused after completion of surgery. In all four patients, cardiac output, oxygen delivery, and oxygen consumption decreased from the pretransfusion values. We conclude that, since intraoperative isovolemic hemodilution increased blood flow and systemic oxygen transport, it may be useful in the intraoperative management of patients with atherosclerotic vascular disease.


Sujet(s)
Artériosclérose/chirurgie , Débit cardiaque , Hémodilution , Consommation d'oxygène , Sujet âgé , Pression sanguine , Transfusion sanguine autologue , Colloïdes/administration et posologie , Humains , Adulte d'âge moyen , Résistance vasculaire , Procédures de chirurgie vasculaire/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...