Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Reprod Toxicol ; 114: 9-21, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36206921

RESUMO

UV-4 (N-(9-methoxynonyl)-1-deoxynojirimycin) is a host-targeted antiviral agent, which targets mammalian proteins (endoplasmic reticulum glucosidases) rather than virally encoded proteins. This mechanism confers both broad-spectrum activity and low potential for generation of viral drug resistance mutations. Reproductive and developmental studies of UV-4 evaluated effects on fertility and early embryonic development in rats, embryo-fetal development in rats and rabbits, and pre- and postnatal development including maternal function in rats. All reproductive and developmental studies conducted achieved dose levels where parental toxicity (generally decreased body weight, decreased food consumption and adverse clinical signs) were observed. Toxicokinetic evaluations confirmed UV-4 crossed the placenta exposing fetal rats and rabbits in utero. Adverse findings in reproductive and developmental studies included decreases in sperm motility with histopathology correlates, visceral and skeletal malformations, changes in eye opening, air drop reflex, vaginal opening and preputial separation. The combined results of the fertility and early embryonic developmental study and pre- and postnatal study suggested that there may be an increased risk for male fertility. These effects are similar to those reported in pre-clinical studies of the structurally related drug Miglustat (N-butyl-1-deoxynojirimycin), therefore UV-4 may have risk of developmental or reproductive adverse outcomes in humans similar to existing approved agents in this drug class.


Assuntos
Reprodução , Motilidade dos Espermatozoides , Gravidez , Feminino , Humanos , Masculino , Ratos , Coelhos , Animais , Ratos Sprague-Dawley , Relação Dose-Resposta a Droga , Fertilidade , Peso Corporal , Mamíferos
2.
PLoS Negl Trop Dis ; 16(8): e0010636, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35939501

RESUMO

BACKGROUND: UV-4 (N-(9'-methoxynonyl)-1-deoxynojirimycin, also called MON-DNJ) is an iminosugar small-molecule oral drug candidate with in vitro antiviral activity against diverse viruses including dengue, influenza, and filoviruses and demonstrated in vivo efficacy against both dengue and influenza viruses. The antiviral mechanism of action of UV-4 is through inhibition of the host endoplasmic reticulum-resident α-glucosidase 1 and α-glucosidase 2 enzymes. This inhibition prevents proper glycan processing and folding of virus glycoproteins, thereby impacting virus assembly, secretion, and the fitness of nascent virions. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a first-in-human, single ascending dose Phase 1a study to evaluate the safety, tolerability, and pharmacokinetics of UV-4 hydrochloride (UV-4B) in healthy subjects (ClinicalTrials.gov Identifier NCT02061358). Sixty-four subjects received single oral doses of UV-4 as the hydrochloride salt equivalent to 3, 10, 30, 90, 180, 360, 720, or 1000 mg of UV-4 (6 subjects per cohort), or placebo (2 subjects per cohort). Single doses of UV-4 hydrochloride were well tolerated with no serious adverse events or dose-dependent increases in adverse events observed. Clinical laboratory results, vital signs, and physical examination data did not reveal any safety signals. Dose-limiting toxicity was not observed; the maximum tolerated dose of UV-4 hydrochloride in humans has not yet been determined (>1000 mg). UV-4 was rapidly absorbed and distributed after dosing with the oral solution formulation used in this study. Median time to reach maximum plasma concentration ranged from 0.5-1 hour and appeared to be independent of dose. Exposure increased approximately in proportion with dose over the 333-fold dose range. UV-4 was quantifiable in pooled urine over the entire collection interval for all doses. CONCLUSIONS/SIGNIFICANCE: UV-4 is a host-targeted broad-spectrum antiviral drug candidate. At doses in humans up to 1000 mg there were no serious adverse events reported and no subjects were withdrawn from the study due to treatment-emergent adverse events. These data suggest that therapeutically relevant drug levels of UV-4 can be safely administered to humans and support further clinical development of UV-4 hydrochloride or other candidate antivirals in the iminosugar class. TRIAL REGISTRATION: ClinicalTrials.gov NCT02061358 https://clinicaltrials.gov/ct2/show/NCT02061358.


Assuntos
Dengue , alfa-Glucosidases , 1-Desoxinojirimicina/efeitos adversos , Antivirais/farmacologia , Área Sob a Curva , Dengue/tratamento farmacológico , Relação Dose-Resposta a Droga , Método Duplo-Cego , Voluntários Saudáveis , Humanos , alfa-Glucosidases/metabolismo , alfa-Glucosidases/uso terapêutico
3.
Int J Toxicol ; 41(3): 182-200, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35426748

RESUMO

The iminosugar UV-4 is a broad-spectrum antiviral drug candidate with activity in vitro and in vivo against multiple, diverse viruses. The toxicological profile of UV-4, dosed as the hydrochloride salt, was evaluated in single-dose and repeat-dose oral toxicity studies in mice, rats, dogs, and non-human primates (NHP). No moribundity or deaths were associated with the drug up to the maximum tolerated dose. No treatment-related adverse effects were observed following single oral doses in dogs, rats, and mice up to 250, 400, 1000 mg/kg, respectively, and in NHP up to 180 mg/kg administered three times daily for 10 days. UV-4-related findings were generally seen at higher doses after 7- or 14-day exposure. The most common clinical pathology findings (increase in aspartate aminotransferase and decreased platelet count) were consistently found across species and each appeared dose related. The kidney, mesenteric lymph nodes, stomach including gastrointestinal tract, and thymus were identified as target organs in mice, rats, and dogs. In 14-day repeat-dose toxicology studies in mice and dogs conducted in compliance with Good Laboratory Practice regulations, the dog was considered to be the most sensitive species to UV-4 exposure based on the treatment-related adverse effects noted in the identified target organs. The results of these studies demonstrate the safety profile of UV-4 hydrochloride and supported the selection of starting and maximal doses for a single ascending dose first-in-human clinical study.


Assuntos
Antivirais , Drogas em Investigação , Administração Oral , Animais , Antivirais/uso terapêutico , Antivirais/toxicidade , Cães , Drogas em Investigação/toxicidade , Dose Máxima Tolerável , Camundongos , Ratos
4.
Int J Toxicol ; 41(3): 201-211, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35227115

RESUMO

UV-4 (N-(9-methoxynonyl)-1-deoxynojirimycin) is a broad-spectrum antiviral drug candidate with demonstrated activity in vitro and in vivo against multiple, diverse viruses. Nonclinical safety pharmacology studies were conducted to support the filing of an Investigational New Drug (IND) application. Preliminary in vitro pharmacology testing evaluating potential for binding to "off-target" receptors and enzymes indicated no significant liability for advanced development of UV-4. The safety pharmacology of UV-4 was evaluated in the in vitro human ether-à-go-go-related gene (hERG) assay, in a central nervous system (CNS) study in the mouse (modified Irwin test), in a respiratory safety study in conscious mice using whole body plethysmography, and in a cardiovascular safety study in conscious, radiotelemetry-instrumented beagle dogs. There were no observed adverse treatment-related effects following administration of UV-4 as the hydrochloride salt in the hERG potassium channel assay, on respiratory function, in the CNS study, or in the cardiovascular assessment. Treatment-related cardiovascular effect of decreased arterial pulse pressure after 50 or 200 mg of UV-4/kg was the only change outside the normal range, and all hemodynamic parameters returned to control levels by the end of the telemetry recording period. These nonclinical safety pharmacology assessments support the evaluation of this host-targeted broad-spectrum antiviral drug candidate in clinical studies.


Assuntos
Sistema Cardiovascular , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Animais , Antivirais/toxicidade , Cães , Avaliação Pré-Clínica de Medicamentos , Drogas em Investigação , Camundongos , Telemetria
5.
Sci Rep ; 9(1): 7484, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31097731

RESUMO

Development of antiviral drug resistance is a continuous concern for viruses with high mutation rates such as influenza. The use of antiviral drugs targeting host proteins required for viral replication is less likely to result in the selection of resistant viruses than treating with direct-acting antivirals. The iminosugar UV-4B is a host-targeted glucomimetic that inhibits endoplasmic reticulum α-glucosidase I and II enzymes resulting in improper glycosylation and misfolding of viral glycoproteins. UV-4B has broad-spectrum antiviral activity against diverse viruses including dengue and influenza. To examine the ability of influenza virus to develop resistance against UV-4B, mouse-adapted influenza virus was passaged in mice in the presence or absence of UV-4B and virus isolated from lungs was used to infect the next cohort of mice, for five successive passages. Deep sequencing was performed to identify changes in the viral genome during passaging in the presence or absence of UV-4B. Relatively few minor variants were identified within each virus and the ratio of nonsynonymous to synonymous (dN/dS) substitutions of minor variants confirmed no apparent positive selection following sustained exposure to UV-4B. Three substitutions (one synonymous in PB2, one nonsynonymous in M and PA each) were specifically enriched (>3%) in UV-4B-treated groups at passage five. Recombinant viruses containing each individual or combinations of these nonsynonymous mutations remained sensitive to UV-4B treatment in mice. Overall, these data provide evidence that there is a high genetic barrier to the generation and selection of escape mutants following exposure to host-targeted iminosugar antivirals.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/genética , Inibidores de Glicosídeo Hidrolases/farmacologia , Vírus da Influenza A/genética , Infecções por Orthomyxoviridae/virologia , Animais , Feminino , Genoma Viral , Vírus da Influenza A/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Recombinação Genética , Seleção Genética
6.
Antiviral Res ; 138: 22-31, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27908828

RESUMO

Iminosugars are host-directed antivirals with broad-spectrum activity. The iminosugar, N-butyl-deoxynojirimycin (NB-DNJ or Miglustat®), is used in humans for treatment of Gaucher's disease and has mild antiviral properties. More potent analogs of NB-DNJ have been generated and have demonstrated activity against a variety of viruses including flaviviruses, influenza, herpesviruses and filoviruses. In the current study, a panel of analogs based on NB-DNJ was analyzed for activity against Ebola (EBOV) and Marburg viruses (MARV). The antiviral activity of NB-DNJ (UV-1), UV-2, UV-3, UV-4 and UV-5 against both EBOV and MARV was demonstrated in Vero cells. Subsequent studies to examine the activity of UV-4 and UV-5 using rodent models of EBOV and MARV were performed. In vivo efficacy studies provided inconsistent data following treatment with iminosugars using filovirus mouse models. A tolerability study in nonhuman primates demonstrated that UV-4 could be administered at much higher dose levels than rodents. Since UV-4 was active in vitro, had been demonstrated to be active against influenza and dengue in vivo, and was being tested in a Phase 1 clinical trial, a small proof-of-concept nonhuman primate trial was performed to determine whether this antiviral candidate could provide clinical benefit to EBOV-infected individuals. Administration of UV-4B did not provide a clinical or survival benefit to macaques infected with EBOV-Makona; however, dosing of animals was not optimal in this study. Efficacy may be improved by thrice daily dosing (e.g. by nasogastric tube feeding) to match the efficacious dosing regimens demonstrated against dengue and influenza viruses.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Ebolavirus/efeitos dos fármacos , Imino Açúcares/farmacologia , Imino Açúcares/uso terapêutico , Marburgvirus/efeitos dos fármacos , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/agonistas , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/uso terapêutico , Animais , Antivirais/administração & dosagem , Antivirais/química , Chlorocebus aethiops , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Imino Açúcares/administração & dosagem , Imino Açúcares/química , Macaca , Camundongos , Modelos Animais , Células Vero
7.
Viruses ; 8(3): 71, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-27072420

RESUMO

Iminosugars that are competitive inhibitors of endoplasmic reticulum (ER) α-glucosidases have been demonstrated to have antiviral activity against a diverse set of viruses. A novel iminosugar, UV-4B, has recently been shown to provide protection against lethal infections with dengue and influenza A (H1N1) viruses in mice. In the current study, the breadth of activity of UV-4B against influenza was examined ex vivo and in vivo. Efficacy of UV-4B against influenza A and B viruses was shown in primary human bronchial epithelial cells, a principal target tissue for influenza. Efficacy of UV-4B against influenza A (H1N1 and H3N2 subtypes) and influenza B was demonstrated using multiple lethal mouse models with readouts including mortality and weight loss. Clinical trials are ongoing to demonstrate safety of UV-4B and future studies to evaluate antiviral activity against influenza in humans are planned.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Antivirais/administração & dosagem , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , Animais , Antivirais/farmacologia , Peso Corporal , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/virologia , Humanos , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Análise de Sobrevida , Resultado do Tratamento
8.
Antiviral Res ; 129: 93-98, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26946111

RESUMO

The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10-20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Inibidores de Glicosídeo Hidrolases/farmacologia , Dengue Grave/tratamento farmacológico , alfa-Glucosidases/metabolismo , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Anticorpos Facilitadores/efeitos dos fármacos , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Células Cultivadas , Chlorocebus aethiops , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Drogas em Investigação , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Inibidores de Glicosídeo Hidrolases/administração & dosagem , Inibidores de Glicosídeo Hidrolases/química , Inibidores de Glicosídeo Hidrolases/uso terapêutico , Humanos , Concentração Inibidora 50 , Camundongos , Monócitos/virologia , Receptores de Interferon/deficiência , Sorogrupo , Dengue Grave/virologia , Células Vero
9.
Viruses ; 7(5): 2404-27, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25984714

RESUMO

Iminosugars are capable of targeting the life cycles of multiple viruses by blocking host endoplasmic reticulum α-glucosidase enzymes that are required for competent replication of a variety of enveloped, glycosylated viruses. Iminosugars as a class are approved for use in humans with diseases such as diabetes and Gaucher's disease, providing evidence for safety of this class of compounds. The in vitro antiviral activity of iminosugars has been described in several publications with a subset of these demonstrating in vivo activity against flaviviruses, herpesviruses, retroviruses and filoviruses. Although there is compelling non-clinical in vivo evidence of antiviral efficacy, the efficacy of iminosugars as antivirals has yet to be demonstrated in humans. In the current study, we report a novel iminosugar, UV-12, which has efficacy against dengue and influenza in mouse models. UV-12 exhibits drug-like properties including oral bioavailability and good safety profile in mice and guinea pigs. UV-12 is an example of an iminosugar with activity against multiple virus families that should be investigated in further safety and efficacy studies and demonstrates potential value of this drug class as antiviral therapeutics.


Assuntos
Antivirais/uso terapêutico , Dengue/tratamento farmacológico , Imino Açúcares/uso terapêutico , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Cobaias , Imino Açúcares/farmacologia , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Orthomyxoviridae/efeitos dos fármacos , Resultado do Tratamento
10.
PLoS One ; 10(3): e0121662, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25786028

RESUMO

Our lead iminosugar analog called UV-4 or N-(9-methoxynonyl)-1-deoxynojirimycin inhibits activity of endoplasmic reticulum (ER) α-glucosidases I and II and is a potent, host-targeted antiviral candidate. The mechanism of action for the antiviral activity of iminosugars is proposed to be inhibition of ER α-glucosidases leading to misfolding of critical viral glycoproteins. These misfolded glycoproteins would then be incorporated into defective virus particles or targeted for degradation resulting in a reduction of infectious progeny virions. UV-4, and its hydrochloride salt known as UV-4B, is highly potent against dengue virus in vitro and promotes complete survival in a lethal dengue virus mouse model. In the current studies, UV-4 was shown to be highly efficacious via oral gavage against both oseltamivir-sensitive and -resistant influenza A (H1N1) infections in mice even if treatment was initiated as late as 48-72 hours after infection. The minimal effective dose was found to be 80-100 mg/kg when administered orally thrice daily. UV-4 treatment did not affect the development of protective antibody responses after either influenza infection or vaccination. Therefore, UV-4 is a promising candidate for further development as a therapeutic intervention against influenza.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Antivirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Oseltamivir/farmacologia , 1-Desoxinojirimicina/efeitos adversos , 1-Desoxinojirimicina/farmacologia , Animais , Anticorpos Antivirais/biossíntese , Antivirais/efeitos adversos , Feminino , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Segurança , Fatores de Tempo , Carga Viral/efeitos dos fármacos
11.
J Virol ; 89(10): 5592-601, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762732

RESUMO

UNLABELLED: The host-targeted antiviral drug UV-4B reduces viral replication and promotes survival in a mouse model of experimental dengue virus (DENV) infection. UV-4B is an iminosugar that inhibits the α-glucosidase family of enzymes and subsequently the folding of glycosylated proteins, both viral and host. Here, we utilized next-generation sequencing to investigate evolution of a flavivirus under selective pressure by a host-targeted antiviral in vivo. In viral populations recovered from UV-4B-treated mice, there was a significant increase in the number of single-nucleotide polymorphisms (SNPs) and the ratio of nonsynonymous to synonymous SNPs compared to findings in viral populations from vehicle-treated mice. The strongest evidence of positive selection was in the glycosylated membrane protein, thereby providing in vivo validation of the mechanism of action of an iminosugar. In addition, mutations in glycosylated proteins were present only in drug-treated mice after a single passage. However, the bulk of the other mutations were present in both populations, indicating nonspecific selective pressure. Together with the continued control of viremia by UV-4B, these findings are consistent with the previously predicted high genetic barrier to escape mutations in host-targeted antivirals. IMPORTANCE: Although hundreds of millions of people are infected with DENV every year, there is currently no approved vaccine or antiviral therapy. UV-4B has demonstrated antiviral activity against DENV and is expected to enter clinical trials soon. Therefore, it is important to understand the mechanisms of DENV resistance to UV-4B. Host-targeted antivirals are thought to have a higher genetic barrier to escape mutants than directly acting antivirals, yet there are very few published studies of viral evolution under host-targeted antivirals. No study to date has described flavivirus evolution in vivo under selective pressure by a host-based antiviral drug. We present the first in vivo study of the sequential progression of viral evolution under selective pressure by a host-targeted antiviral compound. This study bolsters support for the clinical development of UV-4B as an antiviral drug against DENV, and it provides a framework to compare how treatment with other host-targeted antiflaviviral drugs in humans and different animal models influence viral genetic diversity.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Dengue/tratamento farmacológico , Dengue/virologia , Animais , Vírus da Dengue/fisiologia , Modelos Animais de Doenças , Evolução Molecular , Variação Genética , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imino Açúcares/farmacologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mutação , Polimorfismo de Nucleotídeo Único , Seleção Genética , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
12.
Antiviral Res ; 98(1): 35-43, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23376501

RESUMO

The aim of the present study was to evaluate the ability of the iminosugar drug UV-4 to provide in vivo protection from lethal dengue virus (DENV) challenge. This study utilized a well-described model of dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS)-like lethal disease in AG129 mice lacking the type I and II interferon receptors. Herein, we present UV-4 as a potent iminosugar for controlling DENV infection and disease in this mouse model. Specifically, administration of UV-4 reduced mortality, as well as viremia and viral RNA in key tissues, and cytokine storm. In addition, UV-4 treatment can be delayed, and it does not alter the anti-DENV antibody response. These results have set the foundation for development of UV-4 as a DENV-specific antiviral in phase I human clinical trials.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Dengue/virologia , Imino Açúcares/farmacologia , Animais , Antivirais/química , Citocinas , Dengue/imunologia , Vírus da Dengue/fisiologia , Humanos , Imino Açúcares/química , Camundongos , Camundongos Endogâmicos , Relação Estrutura-Atividade
13.
Vaccine ; 28(50): 7852-64, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-20943208

RESUMO

Immunization with plasmid DNA that has been encapsulated in poly lactide-co-glycolide (PLG) microparticles targets the plasmid DNA to antigen presenting cells and elicits immune responses to the encoded antigen(s). Application of a series of electrical pulses (EPT) immediately following unformulated DNA injection enhances expression of the encoded antigen and increases immune responses. The combination of using EPT before or after PLG-encapsulated plasmid DNA immunization was tested to determine if enhanced immune responses would be generated. The results show that the combination lead to both enhanced expression of antigen and more robust T cell responses, even if EPT was applied prior to immunization. The data also demonstrate that recruitment of phagocytes to the injection site was markedly enhanced by EPT, and this resulted in an increase of the antigen expression levels in these cells. Co-administration of microparticles and EPT also effected localized necrosis of muscle fibers, caused persistent Th-1-modulated cytokine production, and lead to the release of two endogenous adjuvants, uric acid and HMGB1. In all, we describe that increased immunogenicity observed with the combination of PLG-encapsulated plasmid DNA microparticle with EPT was caused by an increase in the recruitment of antigen presenting cells which mediated a more robust T cell response than observed with immunization alone.


Assuntos
Eletroporação , Ácido Láctico/farmacologia , Ácido Poliglicólico/farmacologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Células Apresentadoras de Antígenos/imunologia , Hidrocarboneto de Aril Hidroxilases/imunologia , Linhagem Celular Tumoral , Citocromo P-450 CYP1B1 , Citocinas/imunologia , Feminino , Imunidade Inata , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Microesferas , Fagócitos/imunologia , Plasmídeos/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Linfócitos T/imunologia
14.
Clin Immunol ; 112(1): 45-53, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15207781

RESUMO

Injection of microparticle-encapsulated DNA elicits immune responses to plasmid-encoded antigens in mice and humans. Cytochrome P450 CYP1B1 (CYP1B1) is a member of the CYP1 P450 enzyme family that is overexpressed in a variety of solid tumors. The work described herein was performed to study the kinetics of stimulating T cell responsiveness with an encapsulated DNA encoding CYP1B1 and provides support for the clinical development of this formulation. Immunization of HLA-A2/Kb transgenic mice with human CYP1B1 encoding plasmid DNA formulated in poly(lactide-co-glycolide) (PLG) microparticles elicits CD8+ T cells that respond to human CYP1B1-positive target cells. The duration of the immune response, the effect on the immune response of multiple injections, and the safety of repeated injections were studied. These results show that the PLG-encapsulated DNA therapeutic elicits durable immune responses to CYP1B1, the responses are dependent on repeat immunization, and that the formulation is well tolerated.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Sistema Enzimático do Citocromo P-450/imunologia , Imunização/métodos , Vacinas de DNA/administração & dosagem , Animais , Hidrocarboneto de Aril Hidroxilases , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Citocromo P-450 CYP1B1 , Sistema Enzimático do Citocromo P-450/genética , Feminino , Glicolatos/farmacologia , Antígeno HLA-A2/imunologia , Ácido Láctico , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microesferas , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Testes de Toxicidade , Vacinas de DNA/efeitos adversos , Vacinas de DNA/genética , Vacinas de DNA/imunologia
15.
Blood ; 102(9): 3287-94, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12869499

RESUMO

Cytochrome P450 1B1 (CYP1B1), a drug-metabolizing extrahepatic enzyme, was recently shown to be overexpressed in multiple types of cancer. Such tumor-associated genes may be useful targets for anticancer therapy, particularly cancer immunotherapeutics. We identified HLA-A*0201-binding peptides and a naturally processed and presented T-cell epitope capable of inducing CYP1B1-specific cytotoxic T lymphocytes (CTLs) in HLA-A2 transgenic mice. Furthermore, the induction of CYP1B1-specific T cells was demonstrated in healthy donors and cancer patients. These T cells efficiently lysed target cells pulsed with the cognate peptide. More important, HLA-A2-matched tumor cell lines and primary malignant cells were also recognized by CYP1B1-specific CTLs. These findings form the basis of a phase 1 clinical trial exploring a DNA-based vector encoding CYP1B1 for widely applicable cancer immunotherapy conducted at the Dana-Farber Cancer Institute.


Assuntos
Antígenos de Neoplasias/imunologia , Hidrocarboneto de Aril Hidroxilases/imunologia , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Células Sanguíneas , Linhagem Celular Tumoral , Citocromo P-450 CYP1B1 , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Antígenos HLA-A , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias/sangue , Neoplasias/patologia , Peptídeos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...