Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antiviral Res ; 211: 105521, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36596323

RESUMO

The 35th International Conference on Antiviral Research (ICAR), sponsored by the International Society for Antiviral Research (ISAR), was held in Seattle, Washington, USA, on March 21-25, 2022 and concurrently through an interactive remote meeting platform. This report gives an overview of the conference on behalf of the society. It provides a general review of the meeting and awardees, summarizing the presentations and their main conclusions from the perspective of researchers active in many different areas of antiviral research and development. Through ICAR, leaders in the field of antiviral research were able to showcase their efforts, as participants learned about key advances in the field. The impact of these efforts was exemplified by many presentations on SARS-CoV-2 demonstrating the remarkable response to the ongoing pandemic, as well as future pandemic preparedness, by members of the antiviral research community. As we address ongoing outbreaks and seek to mitigate those in the future, this meeting continues to support outstanding opportunities for the exchange of knowledge and expertise while fostering cross-disciplinary collaborations in therapeutic and vaccine development. The 36th ICAR will be held in Lyon, France, March 13-17, 2023.


Assuntos
Antivirais , COVID-19 , Humanos , Antivirais/uso terapêutico , Washington , Complexo Ferro-Dextran , SARS-CoV-2
2.
Antivir Chem Chemother ; 30: 20402066221130853, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36305015

RESUMO

As a result of the multiple gathering and travels restrictions during the SARS-CoV-2 pandemic, the annual meeting of the International Society for Antiviral Research (ISAR), the International Conference on Antiviral Research (ICAR), could not be held in person in 2021. Nonetheless, ISAR successfully organized a remote conference, retaining the most critical aspects of all ICARs, a collegiate gathering of researchers in academia, industry, government and non-governmental institutions working to develop, identify, and evaluate effective antiviral therapy for the benefit of all human beings. This article highlights the 2021 remote meeting, which presented the advances and objectives of antiviral and vaccine discovery, research, and development. The meeting resulted in a dynamic and effective exchange of ideas and information, positively impacting the prompt progress towards new and effective prophylaxis and therapeutics.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , SARS-CoV-2 , Pandemias
3.
Antiviral Res ; 187: 105018, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33476709

RESUMO

MBX-2168 has a mechanism of action similar to that of acyclovir (ACV) and ganciclovir (GCV), but two unique steps differentiate this drug from ACV/GCV. First, MBX-2168 is, at least partially, phosphorylated by the endogenous cellular kinase TAOK3 to a monophosphate. The second involves the removal of a moiety at the 6-position of MBX-2168-MP by adenosine deaminase like protein-1 (ADAL-1). It has been previously demonstrated that co-incubation with pentostatin (dCF), an ADAL-1 inhibitor, antagonizes the anti-viral activity of MBX-2168. We therefore hypothesize that inhibiting ADAL-1 results in a reduction of active compound produced in virus-infected cells. To test this, we examined the effect dCF has on the conversion of MBX-2168 to a triphosphate in HSV-1 and HCMV-infected cells. Our results demonstrate incubation of MBX-2168 alone or with dCF in HCMV-infected cells resulted in 53.1 ± 0.7 and 39.4 ± 1.5 pmol triphosphate/106 cells at 120 h, respectively. Incubation of MBX-2168 alone or with dCF in Vero cells resulted in 12.8 ± 0.1 and 6.7 ± 0.7 pmol triphosphate/106 cells at 24 h, respectively. HSV-1-infected Vero cells demonstrated no statistical difference in triphosphate accumulation at 24 h (13.1 ± 0.3 pmol triphosphate/106 cells). As expected, incubation with dCF resulted in the accumulation of MBX-2168-MP in both HFF (9.8 ± 0.9 pmol MBX-2168-MP/106 cells at 120 h) and Vero cells (4.7 ± 0.3 pmol MBX-2168-MP/106 cells at 24 h) while no detectable levels of monophosphate were observed in cultures not incubated with dCF. We conclude that dCF antagonizes the anti-viral effect of MBX-2168 by inhibiting the production of triphosphate, the active compound.


Assuntos
Antivirais/antagonistas & inibidores , Antivirais/farmacologia , Ciclopropanos/antagonistas & inibidores , Citomegalovirus/efeitos dos fármacos , Guanina/análogos & derivados , Herpesvirus Humano 1/efeitos dos fármacos , Pentostatina/farmacologia , Polifosfatos/metabolismo , Aciclovir/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Ciclopropanos/farmacologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/virologia , Fibroblastos/virologia , Prepúcio do Pênis/citologia , Ganciclovir/farmacologia , Guanina/antagonistas & inibidores , Guanina/farmacologia , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Mutação com Perda de Função , Masculino , Fosforilação , Células Vero , Replicação Viral/efeitos dos fármacos
4.
Antiviral Res ; 175: 104713, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31968222

RESUMO

The third generation of methylenecyclopropane nucleoside analogs (MCPNAs) elicit an anti-viral effect against all three sub-classes of herpes viruses without inducing cytotoxicity in vitro. It has been previously established that the mechanism of action of MCPNAs is similar to that of ganciclovir (GCV) or acyclovir (ACV). However, the activation of MBX-2168, a third generation MCPNA, involves additional and unique enzymatic steps and this process has not been examined in virus-infected cells. To that end, herpes virus-infected cells were incubated with MBX-2168, synguanol, GCV, or ACV. Incubation of HCMV-infected cells with five times the EC50 of MBX-2168 (4.0 µM), synguanol (10.5 µM), or GCV (25 µM) resulted in a time-dependent increase in triphosphate accumulation reaching a maximum of 48.1 ± 5.5, 45.5 ± 2.5, and 42.6 ± 3.7 pmol/106 cells at 120 h, respectively. Additionally, half-lives of these compounds were similar in HCMV-infected cells (GCV-TP = 25.5 ± 2.7 h; MBX-2168-TP/synguanol-TP = 23.0 ± 1.4 h). HSV-1-infected cells incubated with five times the EC50 of MBX-2168 (33.5 µM) or ACV (5.0 µM) demonstrated a time-dependent increase in triphosphate levels reaching a maximum of 12.3 ± 1.5 and 11.6 ± 0.7 pmol/106 cells at 24 h, respectively. ACV-TP and MBX-2168-TP also had similar half-lives under these conditions (27.3 ± 4.8 h and 22.2 ± 2.2 h, respectively). We therefore conclude that although MBX-2168 does not follow the classical route of nucleoside analog activation, the metabolic profile of MBX-2168 is similar to other nucleoside analogs such as GCV and ACV that do.


Assuntos
Antivirais/metabolismo , Ciclopropanos/metabolismo , Guanina/análogos & derivados , Herpesvirus Humano 1/efeitos dos fármacos , Polifosfatos/análise , Aciclovir/farmacologia , Animais , Chlorocebus aethiops , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Fibroblastos/virologia , Ganciclovir/farmacologia , Guanina/biossíntese , Guanina/metabolismo , Meia-Vida , Herpesvirus Humano 1/fisiologia , Humanos , Cinética , Masculino , Nucleosídeos/biossíntese , Nucleosídeos/metabolismo , Polifosfatos/metabolismo , Células Vero
5.
Artigo em Inglês | MEDLINE | ID: mdl-31332074

RESUMO

To determine the mechanism of action of third-generation methylenecyclopropane nucleoside analogs (MCPNAs), DNA sequencing of herpes simplex virus 1 (HSV-1) isolates resistant to third-generation MCPNAs resulted in the discovery of G841S and N815S mutations in HSV-1 UL30. Purified HSV-1 UL30 or human cytomegalovirus (HCMV) UL54 was then subjected to increasing concentrations of MBX-2168-triphosphate (TP), with results demonstrating a 50% inhibitory concentration (IC50) of ∼200 µM, indicating that MBX-2168-TP does not inhibit the viral DNA polymerase. Further metabolic studies showed the removal of a moiety on the guanine ring of MBX-2168. Therefore, we hypothesized that enzymatic removal of a moiety at the 6-position of the guanine ring of third-generation MCPNAs is an essential step in activation. To test this hypothesis, pentostatin (deoxycoformycin [dCF]), an adenosine deaminase-like protein 1 (ADAL-1) inhibitor, was coincubated with MBX-2168. The results showed that dCF antagonized the effect of MBX-2168, with a >40-fold increase in the 50% effective concentration (EC50) at 50 µM dCF (EC50 of 63.1 ± 8.7 µM), compared with MBX-2168 alone (EC50 of 0.2 ± 0.1 µM). Purified ADAL-1 demonstrated time-dependent removal of the moiety on the guanine ring of MBX-2168-monophosphate (MP), with a Km of 17.5 ± 2.4 µM and a Vmax of 0.12 ± 0.04 nmol min-1 Finally, synguanol-TP demonstrated concentration-dependent inhibition of HSV-1 UL30 and HCMV UL54, with IC50s of 0.33 ± 0.16 and 0.38 ± 0.11 µM, respectively. We conclude that ADAL-1 is the enzyme responsible for removing the moiety from the guanine ring of MBX-2168-MP prior to conversion to a TP, the active compound that inhibits the viral DNA polymerase.


Assuntos
Adenosina Desaminase/metabolismo , Ciclopropanos/química , Ciclopropanos/farmacologia , Nucleosídeos/análogos & derivados , Nucleosídeos/farmacologia , Adenosina Desaminase/genética , Animais , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/patogenicidade , DNA Viral/genética , Guanina/análogos & derivados , Guanina/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/patogenicidade , Humanos , Análise de Sequência de DNA/métodos , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética , Replicação Viral/fisiologia
6.
Antiviral Res ; 161: 116-124, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472161

RESUMO

A key step in the replication of human cytomegalovirus (HCMV) in the host cell is the generation and packaging of unit-length genomes into preformed capsids. Enzymes required for this process are so-called terminases, first described for double-stranded DNA bacteriophages. The HCMV terminase consists of the two subunits, the ATPase pUL56 and the nuclease pUL89, and a potential third component pUL51. The terminase subunits are essential for virus replication and are highly conserved throughout the Herpesviridae family. Together with the portal protein pUL104 they form a powerful biological nanomotor. It has been shown for tailed dsDNA bacteriophages that DNA translocation into preformed capsid needs an extraordinary amount of energy. The HCMV terminase subunit pUL56 provides the required ATP hydrolyzing activity. The necessary nuclease activity to cleave the concatemers into unit-length genomes is mediated by the terminase subunit pUL89. Whether this cleavage is mediated by site-specific duplex nicking has not been demonstrated, however, it is required for packaging. Binding to the portal is a prerequisite for DNA translocation. To date, it is a common view that during translocation the terminase moves along some domains of the DNA by a binding and release mechanism. These critical structures have proven to be outstanding targets for drugs to treat HCMV infections because corresponding structures do not exist in mammalian cells. Herein we examine the HCMV terminase as a target for drugs and review several inhibitors discovered by both lead-directed medicinal chemistry and by target-specific design. In addition to producing clinically active compounds the research also has furthered the understanding of the role and function of the terminase itself.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/enzimologia , Endodesoxirribonucleases/antagonistas & inibidores , Acetatos/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Citomegalovirus/genética , Infecções por Citomegalovirus/tratamento farmacológico , Humanos , Quinazolinas/uso terapêutico , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
7.
Antiviral Res ; 158: 255-263, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30153445

RESUMO

Human cytomegalovirus (HCMV) can cause severe disease in patients with compromised or immature immune systems. Currently approved pharmacotherapies for the treatment of systemic HCMV infections [ganciclovir (GCV), cidofovir (CDV), foscarnet] are limited by a high incidence of adverse effects and/or the development of drug resistance. Given that many of these drugs have the same viral target (HCMV-encoded DNA polymerase), cross-resistance is relatively common. The primary means to combat drug resistance is combination pharmacotherapy using therapeutics with different molecular mechanisms of action with the expectation that those combinations result in an additive or synergistic enhancement of effect; combinations that result in antagonism can, in many cases, be detrimental to the outcome of the patient. We therefore tested select combinations of approved (GCV, CDV, letermovir (LMV)) and experimental (brincidofovir (BCV), cyclopropavir (CPV), maribavir (MBV), BDCRB) drugs with the hypothesis that combinations of drugs with different and distinct molecular mechanisms of action will produce an additive and/or synergistic enhancement of antiviral effect against HCMV in vitro. Using MacSynergy II (a statistical package that measures enhancement or lessening of effect relative to zero/additive), select drug combination studies demonstrated combination indices ranging from 160 to 372 with 95% confidence intervals greater than zero indicating that these combinations elicit a synergistic enhancement of effect against HCMV in vitro. These data suggest that administration of a viral DNA polymerase inhibitor, MBV, and/or a viral terminase inhibitor in combination has the potential to address the resistance/cross-resistance problems associated with currently available therapeutics.


Assuntos
Antivirais/administração & dosagem , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Benzimidazóis/farmacologia , Linhagem Celular , Cidofovir/farmacologia , Ciclopropanos/farmacologia , Citosina/análogos & derivados , Citosina/farmacologia , DNA Polimerase Dirigida por DNA/efeitos dos fármacos , Antagonismo de Drogas , Combinação de Medicamentos , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Sinergismo Farmacológico , Quimioterapia Combinada , Endodesoxirribonucleases/antagonistas & inibidores , Fibroblastos , Foscarnet/farmacologia , Ganciclovir/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Organofosfonatos/farmacologia , Ribonucleosídeos/farmacologia , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
8.
Ann Surg Oncol ; 24(13): 3837-3841, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29019106

RESUMO

BACKGROUND: Infectious postoperative complications often delay systemic chemotherapy after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CS-HIPEC). Because the authors have empirically observed fewer incisional infectious complications than expected after CS-HIPEC with mitomycin C (MMC), they investigated the antimicrobial properties of HIPEC perfusate fluid. METHODS: This study prospectively measured in vitro bacterial growth inhibition by HIPEC perfusate (n = 18). After 10 µL of perfusate had been plated on agar plate inoculated by standard strains of either Escherichia coli (strain 25922) or Staphylococcus aureus (strain 25923), it was incubated at 37 °C for 24 h. Antimicrobial activity evidenced by a zone of complete growth inhibition was measured in millimeters. These were compared against growth inhibition produced by control groups represented by MMC solution in normal saline (MMC concentrations of 2, 4, 6, 8, and 8.75 µg/mL), 7 per group. RESULTS: Bacterial inhibition by HIPEC perfusate was stronger against E. coli than against S. aureus (13.1 ± 6.8 vs 8.3 ± 7.7 mm; p = 0.005). No E. coli inhibition was observed for MMC saline in concentrations of 2 through 8 µg/mL (p < 0.001 each), and inhibition of 4.5 ± 5.7 mm was observed for an MMC saline concentration of 8.75 µg/mL (p = 0.007). The S. aureus inhibition zones by MMC saline solutions were 2.2 ± 2.1 (p = 0.002), 5.1 ± 2.3 (p = 0.135), 7.5 ± 1.0 (p = 0.654), 9.6 ± 0.9 (p = 0.058), and 10.2 ± 0.4 mm (p = 0.021). CONCLUSION: The antimicrobial properties of HIPEC perfusate are considerable but variable between patients and stronger against E. coli than against S. aureus. Further studies of HIPEC carrier solutions and chemotherapy agents may result in reduction of surgical-site infection and thus enhanced patient recovery.


Assuntos
Antibacterianos/administração & dosagem , Quimioterapia do Câncer por Perfusão Regional , Procedimentos Cirúrgicos de Citorredução , Hipertermia Induzida , Mitomicina/uso terapêutico , Neoplasias Peritoneais/terapia , Antibióticos Antineoplásicos/uso terapêutico , Terapia Combinada , Escherichia coli/efeitos dos fármacos , Seguimentos , Humanos , Infusões Parenterais , Perfusão , Neoplasias Peritoneais/microbiologia , Neoplasias Peritoneais/patologia , Prognóstico , Estudos Prospectivos , Staphylococcus aureus/efeitos dos fármacos
9.
Antimicrob Agents Chemother ; 60(7): 4176-82, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27139481

RESUMO

Cyclopropavir (CPV) is a promising antiviral drug against human cytomegalovirus (HCMV). As with ganciclovir (GCV), the current standard for HCMV treatment, activation of CPV requires multiple steps of phosphorylation and is enantioselective. We hypothesized that the resulting CPV triphosphate (CPV-TP) would stereoselectively target HCMV DNA polymerase and terminate DNA synthesis. To test this hypothesis, we synthesized both enantiomers of CPV-TP [(+) and (-)] and investigated their action on HCMV polymerase. Both enantiomers inhibited HCMV polymerase competitively with dGTP, with (+)-CPV-TP exhibiting a more than 20-fold lower apparent Ki than (-)-CPV-TP. Moreover, (+)-CPV-TP was a more potent inhibitor than GCV-TP. (+)-CPV-TP also exhibited substantially lower apparent Km and somewhat higher apparent kcat values than (-)-CPV-TP and GCV-TP for incorporation into DNA by the viral polymerase. As is the case for GCV-TP, both CPV-TP enantiomers behaved as nonobligate chain terminators, with the polymerase terminating DNA synthesis after incorporation of one additional nucleotide. These results elucidate how CPV-TP acts on HCMV DNA polymerase and help explain why CPV is more potent against HCMV replication than GCV.


Assuntos
Antivirais/farmacologia , Ciclopropanos/farmacologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/enzimologia , DNA Polimerase Dirigida por DNA/metabolismo , Guanina/análogos & derivados , Replicação do DNA/efeitos dos fármacos , DNA Viral/genética , Farmacorresistência Viral/genética , Guanina/farmacologia , Cinética , Estrutura Molecular , Fosforilação/efeitos dos fármacos
10.
Antimicrob Agents Chemother ; 59(1): 226-32, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25348532

RESUMO

Human cytomegalovirus (HCMV) infection can cause severe illnesses, including encephalopathy and mental retardation, in immunocompromised and immunologically immature patients. Current pharmacotherapies for treating systemic HCMV infections include ganciclovir, cidofovir, and foscarnet. However, long-term administration of these agents can result in serious adverse effects (myelosuppression and/or nephrotoxicity) and the development of viral strains with reduced susceptibility to drugs. The deoxyribosylindole (indole) nucleosides demonstrate a 20-fold greater activity in vitro (the drug concentration at which 50% of the number of plaques was reduced with the presence of drug compared to the number in the absence of drug [EC50] = 0.34 µM) than ganciclovir (EC50 = 7.4 µM) without any observed increase in cytotoxicity. Based on structural similarity to the benzimidazole nucleosides, we hypothesize that the indole nucleosides target the HCMV terminase, an enzyme responsible for packaging viral DNA into capsids and cleaving the DNA into genome-length units. To test this hypothesis, an indole nucleoside-resistant HCMV strain was isolated, the open reading frames of the genes that encode the viral terminase were sequenced, and a G766C mutation in exon 1 of UL89 was identified; this mutation resulted in an E256Q change in the amino acid sequence of the corresponding protein. An HCMV wild-type strain, engineered with this mutation to confirm resistance, demonstrated an 18-fold decrease in susceptibility to the indole nucleosides (EC50 = 3.1 ± 0.7 µM) compared to that of wild-type virus (EC50 = 0.17 ± 0.04 µM). Interestingly, this mutation did not confer resistance to the benzimidazole nucleosides (EC50 for wild-type HCMV = 0.25 ± 0.04 µM, EC50 for HCMV pUL89 E256Q = 0.23 ± 0.04 µM). We conclude, therefore, that the G766C mutation that results in the E256Q substitution is unique for indole nucleoside resistance and distinct from previously discovered substitutions that confer both indole and benzimidazole nucleoside resistance (D344E and A355T).


Assuntos
Benzimidazóis/farmacologia , Citomegalovirus/efeitos dos fármacos , Desoxirribonucleosídeos/farmacologia , Farmacorresistência Viral/genética , Indóis/farmacologia , Ribonucleosídeos/farmacologia , Proteínas Virais/genética , Sequência de Aminoácidos , Antivirais/farmacologia , Sequência de Bases , Citomegalovirus/genética , Citomegalovirus/isolamento & purificação , Dados de Sequência Molecular , Mutação
11.
Antimicrob Agents Chemother ; 58(4): 2329-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24514084

RESUMO

Human cytomegalovirus (HCMV) is a widespread pathogen that can cause severe disease in immunologically immature and immunocompromised patients. The current standard of therapy for the treatment of HCMV infections is ganciclovir (GCV). However, high incidence rates of adverse effects are prevalent and limit the use of this drug. Cyclopropavir (CPV) is 10-fold more effective against HCMV in vitro than GCV (50% effective concentrations [EC50s]=0.46 and 4.1 µM, respectively) without any observed increase in cytotoxicity (S. Zhou, J. M. Breitenbach, K. Z. Borysko, J. C. Drach, E. R. Kern, E. Gullen, Y. C. Cheng, and J. Zemlicka, J. Med. Chem. 47:566-575, 2004, doi:10.1021/jm030316s). We have previously determined that the viral protein kinase pUL97 and endogenous cellular kinases are responsible for the conversion of CPV to a triphosphate (TP), the active compound responsible for inhibiting viral DNA synthesis and viral replication. However, this conversion has not been observed in HCMV-infected cells. To that end, we subjected HCMV-infected cells to equivalently effective concentrations (∼5 times the EC50) of either CPV or GCV and observed a time-dependent increase in triphosphate levels for both compounds (CPV-TP=121±11 pmol/10(6) cells; GCV-TP=43.7±0.4 pmol/10(6) cells). A longer half-life was observed for GCV-TP (48.2±5.7 h) than for CPV-TP (23.8±5.1 h). The area under the curve for CPV-TP produced from incubation with 2.5 µM CPV was 8,680±930 pmol·h/10(6) cells, approximately 2-fold greater than the area under the curve for GCV-TP of 4,520±420 pmol·h/10(6) cells produced from incubation with 25 µM GCV. We therefore conclude that the exposure of HCMV-infected cells to CPV-TP is greater than that of GCV-TP under these experimental conditions.


Assuntos
Ciclopropanos/metabolismo , Citomegalovirus/metabolismo , Ganciclovir/metabolismo , Guanina/análogos & derivados , Linhagem Celular , Ciclopropanos/farmacologia , Citomegalovirus/efeitos dos fármacos , Ganciclovir/farmacologia , Guanina/metabolismo , Guanina/farmacologia , Humanos , Estrutura Molecular
12.
Antimicrob Agents Chemother ; 57(9): 4343-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23817384

RESUMO

Human cytomegalovirus (HCMV) is a widespread pathogen in the human population, affecting many immunologically immature and immunocompromised patients, and can result in severe complications, such as interstitial pneumonia and mental retardation. Current chemotherapies for the treatment of HCMV infections include ganciclovir (GCV), foscarnet, and cidofovir. However, the high incidences of adverse effects (neutropenia and nephrotoxicity) limit the use of these drugs. Cyclopropavir (CPV), a guanosine nucleoside analog, is 10-fold more active against HCMV than GCV (50% effective concentrations [EC50s] = 0.46 and 4.1 µM, respectively). We hypothesize that the mechanism of action of CPV is similar to that of GCV: phosphorylation to a monophosphate by viral pUL97 protein kinase with further phosphorylation to a triphosphate by endogenous kinases, resulting in inhibition of viral DNA synthesis. To test this hypothesis, we isolated a CPV-resistant virus, sequenced its genome, and discovered that bp 498 of UL97 was deleted. This mutation caused a frameshift in UL97 resulting in a truncated protein that lacks a kinase domain. To determine if this base pair deletion was responsible for drug resistance, the mutation was engineered into the wild-type viral genome, which was then exposed to increasing concentrations of CPV. The results demonstrate that the engineered virus was approximately 72-fold more resistant to CPV (EC50 = 25.8 ± 3.1 µM) than the wild-type virus (EC50 = 0.36 ± 0.11 µM). We conclude, therefore, that this mutation is sufficient for drug resistance and that pUL97 is involved in the mechanism of action of CPV.


Assuntos
Citomegalovirus/genética , Mutação da Fase de Leitura , Fases de Leitura Aberta , Proteínas Quinases/genética , Proteínas Virais/genética , Antivirais/farmacologia , Sequência de Bases , Células Cultivadas , Ciclopropanos/farmacologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/crescimento & desenvolvimento , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Ganciclovir/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Dados de Sequência Molecular , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Virais/metabolismo
13.
Biochem Pharmacol ; 81(1): 43-9, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20846508

RESUMO

Many fraudulent nucleosides including the antivirals acyclovir (ACV) and ganciclovir (GCV) must be metabolized to triphosphates to be active. Cyclopropavir (CPV) is a newer, related guanosine nucleoside analog that is active against human cytomegalovirus (HCMV) in vitro and in vivo. We have previously demonstrated that CPV is phosphorylated to its monophosphate (CPV-MP) by the HCMV pUL97 kinase. Consequently, like other nucleoside analogs phosphorylated by viral kinases, CPV most likely must be converted to a triphosphate (CPV-TP) in order to elicit antiviral activity. Once formed by pUL97, we hypothesized that guanosine monophosphate kinase (GMPK) is the enzyme responsible for the conversion of CPV-MP to CPV-DP. Incubation of CPV-MP with GMPK resulted in the formation of CPV-DP and, surprisingly, CPV-TP. When CPV-DP was incubated with GMPK, a time-dependent increase in CPV-TP occurred corresponding to a decrease in CPV-DP thereby demonstrating that CPV-DP is a substrate for GMPK. Substrate specificity experiments revealed that GMP, dGMP, GDP, and dGDP are substrates for GMPK. In contrast, GMPK recognized only acyclovir and ganciclovir monophosphates as substrates, not their diphosphates. Kinetic studies demonstrated that CPV-DP has a K(M) value of 45±15µM. We were, however, unable to determine the K(M) value for CPV-MP directly, but a mathematical model of experimental data gave a theoretical K(M) value for CPV-MP of 332±60µM. We conclude that unlike many other antivirals, cyclopropavir can be converted to its active triphosphate by a single cellular enzyme once the monophosphate is formed by a virally encoded kinase.


Assuntos
Antivirais/química , Antivirais/metabolismo , Guanilato Quinases/metabolismo , Nucleotídeos/química , Nucleotídeos/metabolismo , Antivirais/farmacologia , Cinética , Modelos Químicos , Estrutura Molecular , Relação Estrutura-Atividade , Especificidade por Substrato
14.
Antimicrob Agents Chemother ; 54(8): 3093-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20547817

RESUMO

Human cytomegalovirus (HCMV) is a widespread pathogen that can cause severe disease in immunologically immature and immunocompromised individuals. Cyclopropavir (CPV) is a guanine nucleoside analog active against human and murine cytomegaloviruses in cell culture and efficacious in mice by oral administration. Previous studies established that the mechanism of action of CPV involves inhibition of viral DNA synthesis. Based upon this action and the structural similarity of CPV to ganciclovir (GCV), we hypothesized that CPV must be phosphorylated to a triphosphate to inhibit HCMV DNA synthesis and that pUL97 is the enzyme responsible for the initial phosphorylation of CPV to a monophosphate (CPV-MP). We found that purified pUL97 phosphorylated CPV 45-fold more extensively than GCV, a known pUL97 substrate and the current standard of treatment for HCMV infections. Kinetic studies with CPV as the substrate for pUL97 demonstrated a Km of 1,750+/-210 microM. Introduction of 1.0 or 10 nM maribavir, a known pUL97 inhibitor, and subsequent Lineweaver-Burk analysis demonstrated competitive inhibition of CPV phosphorylation, with a Ki of 3.0+/-0.3 nM. Incubation of CPV with pUL97 combined with GMP kinase [known to preferentially phosphorylate the (+)-enantiomer of CPV-MP] established that pUL97 stereoselectively phosphorylates CPV to its (+)-monophosphate. These results elucidate the mechanism of CPV phosphorylation and help explain its selective antiviral action.


Assuntos
Antivirais/metabolismo , Ciclopropanos/metabolismo , Guanina/análogos & derivados , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Benzimidazóis/metabolismo , Citomegalovirus/metabolismo , Ganciclovir/metabolismo , Guanina/metabolismo , Guanilato Quinases/metabolismo , Humanos , Cinética , Fosforilação , Ribonucleosídeos/metabolismo , Estereoisomerismo
15.
Antimicrob Agents Chemother ; 54(4): 1512-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20086149

RESUMO

Triciribine (TCN) is a tricyclic nucleoside that inhibits human immunodeficiency virus type 1 (HIV-1) replication by a unique mechanism not involving the inhibition of enzymes directly involved in viral replication. This activity requires the phosphorylation of TCN to its 5' monophosphate by intracellular adenosine kinase. New testing with a panel of HIV and simian immunodeficiency virus isolates, including low-passage-number clinical isolates and selected subgroups of HIV-1, multidrug resistant HIV-1, and HIV-2, has demonstrated that TCN has broad antiretroviral activity. It was active in cell lines chronically infected with HIV-1 in which the provirus was integrated into chromosomal DNA, thereby indicating that TCN inhibits a late process in virus replication. The selection of TCN-resistant HIV-1 isolates resulted in up to a 750-fold increase in the level of resistance to the drug. DNA sequence analysis of highly resistant isolate HIV-1(H10) found five point mutations in the HIV-1 gene nef, resulting in five different amino acid changes. DNA sequencing of the other TCN-resistant isolates identified at least one and up to three of the same mutations observed in isolate HIV-1(H10). Transfer of the mutations from TCN-resistant isolate HIV-1(H10) to wild-type virus and subsequent viral growth experiments with increasing concentrations of TCN demonstrated resistance to the drug. We conclude that TCN is a late-phase inhibitor of HIV-1 replication and that mutations in nef are necessary and sufficient for TCN resistance.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Ribonucleosídeos/farmacologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Linhagem Celular , Farmacorresistência Viral/genética , Genes nef , Transcriptase Reversa do HIV/antagonistas & inibidores , HIV-1/genética , HIV-2/efeitos dos fármacos , Humanos , Técnicas In Vitro , Testes de Sensibilidade Microbiana , Mutação Puntual , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Replicação Viral/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
16.
Bioorg Med Chem Lett ; 19(3): 792-6, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19097789

RESUMO

5'-O-D- and L-amino acid derivatives and 5'-O-(D- and L-amino acid methyl ester phosphoramidate) derivatives of vidarabine (ara-A) were synthesized as vidarabine prodrugs. Some compounds were equi- or more potent in vitro than vidarabine against two pox viruses and their uptake by cultured cells was improved compared to the parent drug.


Assuntos
Antivirais/síntese química , Química Farmacêutica/métodos , Pró-Fármacos/síntese química , Vidarabina/síntese química , Administração Oral , Antivirais/farmacologia , Arabinonucleosídeos/química , Células CACO-2 , Células Cultivadas , Desenho de Fármacos , Ésteres , Células HeLa , Humanos , Ácidos Levulínicos/química , Nucleosídeos/química , Poxviridae/metabolismo , Pró-Fármacos/farmacologia , Vidarabina/farmacologia
17.
Nucleosides Nucleotides Nucleic Acids ; 28(9): 795-808, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20183619

RESUMO

Enantiomeric cyclopropavir phosphates (+)-9 and (-)-9 were synthesized and investigated as substrates for GMP kinase. N(2)-Isobutyryl-di-O-acetylcyclopropavir (11) was converted to (+)-monoacetate 12 using hydrolysis catalyzed by porcine liver esterase. Phosphorylation via phosphite 13 gave after deacylation, phosphate (+)-9. Acid-catalyzed tetrahydropyranylation of (+)-monoacetate 12 gave, after deacylation, tetrahydropyranyl derivative 14. Phosphorylation via phosphite 15 furnished, after deprotection, enantiomeric phosphate (-)-9. Racemic diphosphate 16 was also synthesized. The phosphate (+)-9 is a relatively good substrate for GMP kinase with a K(M) value of 57 microM that is similar to that of the natural substrates GMP (61 microM) and dGMP (82 microM). In contrast, the enantiomer (-)-9 is not a good substrate (K(M) 1200 microM) indicating a significant enantioselectivity for the GMP kinase catalyzed reaction of monophosphate to diphosphate.


Assuntos
Ciclopropanos/química , Guanina/análogos & derivados , Guanilato Quinases/metabolismo , Compostos Organofosforados/síntese química , Compostos Organofosforados/metabolismo , Acilação , Esterases/metabolismo , Guanina/química , Cinética , Estrutura Molecular , Compostos Organofosforados/química , Fosforilação , Especificidade por Substrato
18.
Cancer Res ; 66(7): 3845-51, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585212

RESUMO

Suicide gene therapy with the herpes simplex virus thymidine kinase (HSV-TK) cDNA and ganciclovir can elicit cytotoxicity to transgene-expressing and nonexpressing bystander cells via transfer of ganciclovir phosphates through gap junctions. HeLa cells do not exhibit bystander cytotoxicity, although we showed recently that they transfer low levels of ganciclovir phosphates to bystander cells. Here, we attempted to induce bystander cytotoxicity using hydroxyurea, an inhibitor of ribonucleotide reductase, to decrease the endogenous dGTP pool, which should lessen competition with ganciclovir triphosphate for DNA incorporation. Addition of hydroxyurea to cocultures of HSV-TK-expressing and bystander cells synergistically increased ganciclovir-mediated cytotoxicity to both cell populations while producing primarily an additive effect in cultures of 100% HSV-TK-expressing cells. Whereas HSV-TK-expressing cells in coculture were approximately 50-fold less sensitive to ganciclovir compared with cultures of 100% HSV-TK-expressing cells, addition of hydroxyurea restored ganciclovir sensitivity. Quantification of deoxynucleoside triphosphate pools showed that hydroxyurea decreased dGTP pools without significantly affecting ganciclovir triphosphate levels. Although hydroxyurea significantly increased the ganciclovir triphosphate:dGTP value for 12 to 24 hours in HSV-TK-expressing and bystander cells from coculture (1.4- to 4.9-fold), this value was increased for <12 hours (2.5-fold) in 100% HSV-TK-expressing cells. These data suggest that the prolonged increase in the ganciclovir triphosphate:dGTP value in cells in coculture resulted in synergistic cytotoxicity. Compared with enhancement of bystander cytotoxicity through modulation of gap junction intercellular communication, this strategy is superior because it increased cytotoxicity to both HSV-TK-expressing and bystander cells in coculture. This approach may improve clinical efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ganciclovir/farmacologia , Hidroxiureia/farmacologia , Simplexvirus/genética , Timidina Quinase/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Técnicas de Cocultura , Nucleotídeos de Desoxiguanina/metabolismo , Sinergismo Farmacológico , Ganciclovir/efeitos adversos , Ganciclovir/análogos & derivados , Ganciclovir/metabolismo , Ganciclovir/farmacocinética , Terapia Genética/métodos , Células HeLa , Humanos , Hidroxiureia/efeitos adversos , Nucleotídeos/metabolismo , Simplexvirus/enzimologia , Timidina Quinase/biossíntese , Timidina Quinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...