Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int J Toxicol ; 35(3): 309-16, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26700136

RESUMO

SYN-004 is a first in class, recombinant ß-lactamase that degrades ß-lactam antibiotics and has been formulated to be administered orally to patients receiving intravenous ß-lactam antibiotics including cephalosporins. SYN-004 is intended to degrade unmetabolized antibiotics excreted into the intestines and thus has the potential to protect the gut microbiome from disruption by these antibiotics. Protection of the gut microbiome is expected to protect against opportunistic enteric infections such as Clostridium difficile infection as well as antibiotic-associated diarrhea. In order to demonstrate that oral SYN-004 is safe for human clinical trials, 2 Good Laboratory Practice-compliant toxicity studies were conducted in Beagle dogs. In both studies, SYN-004 was administered orally 3 times per day up to the maximum tolerated dose of the formulation. In the first study, doses of SYN-004 administered over 28 days were safe and well tolerated in dogs with the no-observed-adverse-effect level at the high dose of 57 mg/kg/day. Systemic absorption of SYN-004 was minimal and sporadic and showed no accumulation during the study. In the second study, doses up to 57 mg/kg/day were administered to dogs in combination with an intravenous dose of ceftriaxone (300 mg/kg) given once per day for 14 days. Coadministration of oral SYN-004 with intravenous ceftriaxone was safe and well tolerated, with SYN-004 having no noticeable effect on the plasma pharmacokinetics of ceftriaxone. These preclinical studies demonstrate that SYN-004 is well tolerated and, when coadministered with ceftriaxone, does not interfere with its systemic pharmacokinetics. These data supported advancing SYN-004 into human clinical trials.


Assuntos
Antibacterianos , Ceftriaxona , Substâncias Protetoras , Proteínas Recombinantes , beta-Lactamases , Administração Intravenosa , Administração Oral , Animais , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Antibacterianos/farmacocinética , Ductos Biliares/metabolismo , Ceftriaxona/administração & dosagem , Ceftriaxona/sangue , Ceftriaxona/farmacocinética , Cães , Interações Medicamentosas , Feminino , Microbioma Gastrointestinal , Masculino , Substâncias Protetoras/administração & dosagem , Substâncias Protetoras/farmacocinética , Substâncias Protetoras/farmacologia , Substâncias Protetoras/toxicidade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/toxicidade , Comprimidos com Revestimento Entérico , Testes de Toxicidade Subaguda , beta-Lactamases/administração & dosagem , beta-Lactamases/farmacocinética , beta-Lactamases/farmacologia , beta-Lactamases/toxicidade
2.
Anaerobe ; 41: 58-67, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27262694

RESUMO

The gut microbiome, composed of the microflora that inhabit the gastrointestinal tract and their genomes, make up a complex ecosystem that can be disrupted by antibiotic use. The ensuing dysbiosis is conducive to the emergence of opportunistic pathogens such as Clostridium difficile. A novel approach to protect the microbiome from antibiotic-mediated dysbiosis is the use of beta-lactamase enzymes to degrade residual antibiotics in the gastrointestinal tract before the microflora are harmed. Here we present the preclinical development and early clinical studies of the beta-lactamase enzymes, P3A, currently referred to as SYN-004, and its precursor, P1A. Both P1A and SYN-004 were designed as orally-delivered, non-systemically available therapeutics for use with intravenous beta-lactam antibiotics. SYN-004 was engineered from P1A, a beta-lactamase isolated from Bacillus licheniformis, to broaden its antibiotic degradation profile. SYN-004 efficiently hydrolyses penicillins and cephalosporins, the most widely used IV beta-lactam antibiotics. In animal studies, SYN-004 degraded ceftriaxone in the GI tract of dogs and protected the microbiome of pigs from ceftriaxone-induced changes. Phase I clinical studies demonstrated SYN-004 safety and tolerability. Phase 2 studies are in progress to assess the utility of SYN-004 for the prevention of antibiotic-associated diarrhea and Clostridium difficile disease.


Assuntos
Antibacterianos/farmacologia , Enterocolite Pseudomembranosa/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , beta-Lactamases/farmacologia , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Clostridioides difficile/efeitos dos fármacos , Cães , Farmacorresistência Bacteriana , Estabilidade de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Cinética , Testes de Sensibilidade Microbiana , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Sus scrofa , beta-Lactamases/química , beta-Lactamases/uso terapêutico
3.
Clin Drug Investig ; 36(9): 725-734, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27283946

RESUMO

BACKGROUND: SYN-004 is an orally administered ß-lactamase enzyme, designed to be given concurrently with certain intravenous ß-lactam antibiotics like cephalosporins. SYN-004 is intended to degrade residual antibiotics excreted into the intestine as a result of hepatobiliary excretion and to prevent the disruption of the gut microbiome by these excess antibiotics. Preserving the gut microbiome is expected to prevent secondary infections by pathogens like Clostridium difficile and protect against other antibiotic-associated diarrheas. METHODS: Two, randomized, double blind, placebo-controlled Phase 1 clinical studies were conducted in normal healthy adult volunteers to assess the tolerability and systemic absorption of single and multiple doses of SYN-004. A single-ascending dose study investigated single oral doses of 75-750 mg SYN-004 and was conducted in 40 subjects (five cohorts of six active and two placebo subjects). A multiple-ascending dose study investigated doses of 75-300 mg SYN-004, administered every 6 h for 7 days and was conducted in 24 subjects (three cohorts of six active and two placebo subjects). The safety and tolerability of SYN-004 was assessed and serial plasma and serum samples were collected to assess the pharmacokinetics and potential immunogenicity of SYN-004. RESULTS: Minimal and mild adverse events were reported in ~30 % of the subjects who received active drug and placebo and no antidrug antibodies were detected in any subject. Analysis of serial plasma samples demonstrated negligible systemic bioavailability of SYN-004 with most plasma concentrations being below the lower limit of quantitation (0.8 ng/mL) for the assay. SYN-004 was well tolerated in the 48 subjects who received active drug, and adverse events in those subjects were comparable to the 16 subjects who received placebo, up to the maximum doses administered in each study. CONCLUSION: SYN-004 was well tolerated up to a single oral dose of 750 mg and multiple doses of 300 mg every 6 h for 7 days. The pharmacokinetic results support that SYN-004 remained localized in the intestine.


Assuntos
Clostridioides difficile , Infecções por Clostridium/tratamento farmacológico , Diarreia/prevenção & controle , Proteínas Recombinantes/uso terapêutico , beta-Lactamases/uso terapêutico , Adolescente , Adulto , Idoso , Disponibilidade Biológica , Infecções por Clostridium/complicações , Diarreia/induzido quimicamente , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética , Adulto Jovem , beta-Lactamases/efeitos adversos , beta-Lactamases/farmacocinética
4.
Cancer Gene Ther ; 11(2): 92-102, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14730331

RESUMO

Oncolytic adenoviral vectors selectively replicate in and lyse human tumor cells, providing a promising means for targeted tumor destruction. However, oncolytic vectors have limited capacity for incorporation of additional genetic material that could encode therapeutic transgenes and/or transcriptional regulatory control elements to augment the efficacy and/or safety of the vector. Therefore, we hypothesized that coadministration of an oncolytic vector with a replication-defective, gutless adenoviral vector encoding a therapeutic transgene would result in replication of both vectors within a tumor and potentiate antitumor efficacy relative to the use of either vector alone. We constructed gutless vectors encoding the murine granulocyte-macrophage colony-stimulating factor (AGVmGMF) or human tumor necrosis factor alpha-related apoptosis-inducing ligand (AGVhTRAIL) gene and tested the ability of these vectors to augment the efficacy of an oncolytic vector (Ar6pAE2fE3F) in a potentiating vector strategy. In Hep3B cells in vitro, cotreatment with Ar6pAE2fE3F increased transgene expression from AGVhTRAIL and permitted replication of AGVhTRAIL, suggesting that an oncolytic vector can propagate gutless vector spread in vivo. In pre-established Hep3B xenograft tumors, neither gutless vector alone inhibited tumor growth; however, coadministration of AGVmGMF or AGVhTRAIL with Ar6pAE2fE3F significantly reduced tumor growth relative to Ar6pAE2fE3F alone. Additionally, use of AGVhTRAIL with Ar6pAE2fE3F increased the number of complete or partial tumor regressions observed at study end. These data provide evidence that coadministration of an oncolytic vector with a gutless vector holds promise for potentiating tumor ablation efficacy.


Assuntos
Adenoviridae/genética , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Glicoproteínas de Membrana/genética , Neoplasias Experimentais/terapia , Fator de Necrose Tumoral alfa/genética , Animais , Antineoplásicos/uso terapêutico , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Vírus Auxiliares/genética , Humanos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
5.
J Virol ; 79(9): 5455-65, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15827160

RESUMO

Historically, the adenoviral E3 region was found to be nonessential for viral replication in vitro. In addition, adenoviruses whose genome was more than approximately 105% the size of the native genome were inefficiently packaged. These profound observations were used experimentally to insert transgenes into the adenoviral backbone. More recently, however, the reintroduction of the E3 region into oncolytic adenoviruses has been found to positively influence antitumor efficacy in preclinical models and clinical trials. In the studies reported here, the granulocyte-macrophage colony-stimulating factor (GM-CSF) cDNA sequence has been substituted for the E3-gp19 gene in oncolytic adenoviruses that otherwise retained the E3 region. Five viruses that differed slightly in the method of transgene insertion were generated and compared to Ar6pAE2fGmF (E2F/GM/DeltaE3), a previously described E3-deleted oncolytic adenovirus encoding GM-CSF. In all of the viruses, the human E2F-1 promoter regulated E1A expression and GM-CSF expression was under the control of the adenoviral E3 promoter and the packaging signal was relocated immediately upstream from the right terminal repeat. The E3-gp19-deleted viruses had similar cytolytic properties, as measured in vitro by cytotoxicity assays, but differed markedly in their capacity to express and secrete GM-CSF. Ar15pAE2fGmF (E2F/GM/E3b), the virus that produced the highest levels of GM-CSF and retained the native GM-CSF leader sequence, was selected for further analysis. The E2F/GM/E3b and E2F/GM/DeltaE3 viruses exhibited similar cytotoxic activity and GM-CSF production in several tumor cell lines in vitro. However, when compared in vivo in nude mouse xenograft tumor models, E2F/GM/E3b spread through tumors to a greater extent, resulted in higher peak GM-CSF and total exposure levels in both tumor and serum, and was more efficacious than the E3-deleted virus. Using the matched WI-38 (parental) and WI-38-VA13 (simian virus 40 large T antigen transformed) cell pair, GM-CSF was shown to be selectively produced in cells expressing high levels of E2F, indicating that the tumor-selective E2F promoter controlled E1A and GM-CSF expression.


Assuntos
Adenoviridae/genética , Proteínas E3 de Adenovirus/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias Experimentais/terapia , Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E2 de Adenovirus/genética , Proteínas E3 de Adenovirus/análise , Animais , Proteínas do Capsídeo/análise , Feminino , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Transgenes , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Ther ; 7(6): 755-64, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12788649

RESUMO

Oncolytic adenoviruses are being tested as biological cancer therapeutics. Ar6pAE2fF (E2F vector) contains the E2F-1 promoter to regulate the expression of the E1a gene in cells with a disregulated retinoblastoma pathway. Ar6pAE2fmGmF (E2F-GM vector) includes the murine granulocyte-macrophage colony-stimulating factor (GM-CSF) transgene to enhance anti-tumor activity. Both vectors selectively killed human tumor cells in vitro. The E2F-GM vector expressed biologically active murine GM-CSF in vitro and GM-CSF was detected for several days in serum and tumor extracts following injections of established human xenograft tumors. In vivo, both vectors showed significant dose-dependent anti-tumor responses. The E2F-GM vector elicited greater efficacy compared to the E2F vector, demonstrating that GM-CSF enhanced the anti-tumor activity, even in immunodeficient nude mice. Histological analysis showed that both vectors induced necrosis and mononuclear cell infiltration, but only the E2F-GM vector resulted in eosinophil infiltration. Vector replication in vivo was demonstrated. The data showed that intratumoral injection of a GM-CSF-armed oncolytic vector induced potent anti-tumor responses in xenograft tumor models, likely as the result of both oncolytic vector activity and the induction of GM-CSF-mediated inflammation and innate immunity.


Assuntos
Adenoviridae/fisiologia , Carcinoma Hepatocelular/virologia , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Vetores Genéticos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias Hepáticas/virologia , Fatores de Transcrição/genética , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Ensaio de Imunoadsorção Enzimática , Feminino , Vetores Genéticos/genética , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA