Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Antimicrob Agents Chemother ; 68(1): e0095323, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38047645

RESUMO

Molnupiravir, an oral prodrug of N-hydroxycytidine (NHC), previously demonstrated broad in vitro antiviral activity against multiple RNA viruses and has shown a high barrier to the development of resistance. Here, we present the antiviral activity of NHC against recent SARS-CoV-2 variants and the results of resistance selection studies to better understand the potential for viral resistance to NHC. NHC activity against SARS-CoV-2 variants omicron (BA.1, BA.1.1, BA.2, BA.4, BA.4.6, BA.5, BQ.1.1, XBB.1, and XBB.1.5), alpha (B.1.1.7), beta (B.1.351), gamma (P.1), delta (B.1.617.2), lambda (C.37), and mu (B.1.621) was evaluated in Vero E6 cells using cytopathic effect assays. Resistance selection studies were performed by passaging SARS-CoV-2 (WA1) in the presence of NHC or a 3C-like protease inhibitor (MRK-A) in Vero E6 cells. Supernatants from cultures exhibiting a cytopathic effect score of ≥2 were re-passaged, and IC50 values were estimated. Whole-genome deep sequencing was performed on viral RNA isolated at each passage. NHC demonstrated similar potency against all SARS-CoV-2 variants evaluated. No evidence of SARS-CoV-2 phenotypic or genotypic resistance to NHC was observed following 30 passages. A random pattern of nucleotide changes was observed in NHC cultures, consistent with the drug's mechanism of action. In contrast, resistance was readily selected in all three MRK-A control cultures with the selection of a T21I substitution in the 3C-like protease. In conclusion, molnupiravir maintains antiviral activity across all major SARS-CoV-2 variants. Furthermore, no evidence of viral resistance to NHC was observed, supporting previous reports that NHC has a high barrier to developing resistance.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Antivirais/farmacologia
2.
J Immunotoxicol ; 17(1): 175-185, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33078980

RESUMO

The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint which may be engaged by cells in a tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®) is a potent and highly selective humanized monoclonal antibody (mAb) of the IgG4/κ isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. The current work was focused on developing a mouse T-Dependent Antibody Response (TDAR) model using a murinized rat anti-mouse PD-1 antibody (muDX400; a rodent surrogate for pembrolizumab) to evaluate the potential impact of treatment with a PD-1 inhibitor on immune responses to an antigen challenge (e.g. HBsAg in Hepatitis B vaccine). Despite the lower binding affinity and T1/2 compared to pembrolizumab, ligand blocking data indicated muDX400 had appropriate pharmacological activity and demonstrated efficacy in mouse tumor models, thus was suitable for pharmacodynamic and vaccination studies in mice. In a vaccination study in which mice were concomitantly administered muDX400 and the Hepatitis B vaccine, muDX400 was well-tolerated and did not result in any immune-mediated adverse effects. The treatment with muDX400 was associated with a shift in the ratio between naive and memory cells in both CD4+ and CD8+ T-lymphocytes in the spleen but did not affect anti-HBsAg antibody response profile. The mouse TDAR model using the Hepatitis B vaccine and the surrogate anti-PD1 monoclonal antibody was a useful tool in the evaluation of the potential immune-mediated effects of pembrolizumab following vaccination and appears to be a suitable alternative for the nonhuman primate TDAR models utilized for other checkpoint inhibitors.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Vacinas contra Hepatite B/imunologia , Vírus da Hepatite B/fisiologia , Hepatite B/imunologia , Linfócitos T/imunologia , Animais , Formação de Anticorpos , Humanos , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Primatas , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Ratos , Vacinação
3.
Mol Cancer Ther ; 19(6): 1298-1307, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32229606

RESUMO

The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint, which may be engaged by cells in the tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®, MK-3475) is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This blockade enhances the functional activity of T cells to facilitate tumor regression and ultimately immune rejection. Pembrolizumab binds to human and cynomolgus monkey PD-1 with picomolar affinity and blocks the binding of human and cynomolgus monkey PD-1 to PD-L1 and PD-L2 with comparable potency. Pembrolizumab binds both the C'D and FG loops of PD-1. Pembrolizumab overcomes human and cynomolgus monkey PD-L1-mediated immune suppression in T-cell cultures by enhancing IL2 production following staphylococcal enterotoxin B stimulation of healthy donor and cancer patient cells, and IFNγ production in human primary tumor histoculture. Ex vivo and in vitro studies with human and primate T cells show that pembrolizumab enhances antigen-specific T-cell IFNγ and IL2 production. Pembrolizumab does not mediate FcR or complement-driven effector function against PD-1-expressing cells. Pembrolizumab displays dose-dependent clearance and half-life in cynomolgus monkey pharmacokinetic and toxicokinetic studies typical for human IgG4 antibodies. In nonhuman primate toxicology studies, no findings of toxicologic significance were observed. The preclinical data for pembrolizumab are consistent with the clinical anticancer activity and safety that has been demonstrated in human clinical trials.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Leucócitos Mononucleares/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Animais , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacocinética , Inibidores de Checkpoint Imunológico/farmacologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/patologia , Proteína 2 Ligante de Morte Celular Programada 1/antagonistas & inibidores , Proteína 2 Ligante de Morte Celular Programada 1/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Distribuição Tecidual , Testes de Toxicidade
4.
J Urol ; 190(3): 850-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23507396

RESUMO

PURPOSE: A phase I trial of intravesical recombinant adenovirus mediated interferon-α2b gene therapy (rAd-IFNα) formulated with the excipient SCH Syn3 was conducted in patients with nonmuscle invasive bladder cancer who had disease recurrence after treatment with bacillus Calmette-Guérin. The primary objective was to determine the safety of rAd-IFNα/Syn3. Secondary end points were demonstrated effective rAd-IFNα gene expression and preliminary evidence of clinical activity at 3 months. MATERIALS AND METHODS: A total of 17 patients with recurrent nonmuscle invasive bladder cancer after bacillus Calmette-Guérin treatment were enrolled in the study. A single treatment of rAd-IFNα (3 × 10(9) to 3 × 10(11) particles per ml) formulated with the excipient Syn3 was administered. Patient safety was evaluated for 12 or more weeks. Efficacy of gene transfer was determined by urine IFNα protein concentrations. Preliminary drug efficacy was determined at 3 months. RESULTS: Intravesical rAd-IFNα/Syn3 was well tolerated as no dose limiting toxicity was encountered. Urgency was the most common adverse event and all cases were grade 1 or 2. rAd-IFNα DNA was not detected in the blood. However, transient low serum IFNα and Syn3 levels were measured. High and prolonged dose related urine IFNα levels were achieved with the initial treatment. Of the 14 patients treated at doses of 10(10) or more particles per ml with detectable urine IFNα, 6 (43%) experienced a complete response at 3 months and 2 remained disease-free at 29.0 and 39.2 months, respectively. CONCLUSIONS: Intravesical rAd-IFNα/Syn3 was well tolerated with no dose limiting toxicity encountered. Dose dependent urinary IFNα concentrations confirmed efficient gene transfer and expression. Intravesical rAd-IFNα/Syn3 demonstrated clinical activity in nonmuscle invasive bladder cancer recurring after bacillus Calmette-Guérin.


Assuntos
Carcinoma de Células de Transição/terapia , Terapia Genética/métodos , Interferon-alfa/administração & dosagem , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/terapia , Neoplasias da Bexiga Urinária/terapia , Adenoviridae/genética , Administração Intravesical , Adulto , Idoso , Idoso de 80 Anos ou mais , Vacina BCG/administração & dosagem , Carcinoma de Células de Transição/mortalidade , Carcinoma de Células de Transição/patologia , Ácidos Cólicos/administração & dosagem , Dissacarídeos/administração & dosagem , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Vetores Genéticos , Humanos , Interferon alfa-2 , Interferon-alfa/genética , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Recidiva Local de Neoplasia/patologia , Prognóstico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Medição de Risco , Análise de Sobrevida , Falha de Tratamento , Resultado do Tratamento , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/patologia
5.
Mol Ther ; 12(2): 254-63, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16043097

RESUMO

Intravascular administration of recombinant adenovirus (rAd) in cancer patients has been well tolerated. However, dose-limiting hemodynamic responses associated with suppression of cardiac output have been observed at doses of 7.5 x 10(13) particles. While analysis of hemodynamic responses induced by small-molecule pharmaceuticals is well established, little is known about the cardiovascular effects of rAd. Telemetric cardiovascular (CV) monitoring in mice was utilized to measure hemodynamic events following intravascular rAd administration. Electrocardiogram analysis revealed a block in the SA node 3-4 min postinfusion, resulting in secondary pacemaking initiated at the AV node. This was associated with acute bradycardia, reduced blood pressure, and hypothermia followed by gradual recovery. Adenovirus-primed murine sera with high neutralizing antibody (nAb) titers could inhibit CV responses, whereas human sera with equivalent nAb titers induced by natural infection were, surprisingly, not inhibitory. Interestingly, repeat dosing within 2-4 h of the primary injection resulted in desensitization, resembling tachyphylaxis, for subsequent CV responses. Last, depletion of Kupffer cells prior to rAd infusion precluded induction of CV responses. These inhibitory effects suggest that rAd interactions with certain cells of the reticular endothelial system are associated with induction of CV responses. Significantly, these studies may provide insight into management of acute adverse effects following rAd systemic delivery, enabling a broadening of therapeutic index.


Assuntos
Adenoviridae/genética , Terapia Genética , Vetores Genéticos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , beta-Galactosidase/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas , Débito Cardíaco/efeitos dos fármacos , Sistema Cardiovascular/efeitos dos fármacos , Eletrocardiografia , Vetores Genéticos/uso terapêutico , Humanos , Vírus da Influenza A , Células de Kupffer/fisiologia , Camundongos , Camundongos Endogâmicos BALB C
6.
Shock ; 23(6): 507-15, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15897802

RESUMO

Control of dendritic cell (DC) function is critical for strategies to modulate innate and acquired immune responses. We examined whether transduction of murine DCs with adenoviral vectors (Adv) expressing interleukin (IL)-10 could alter their phenotype and T cell stimulatory function. Murine bone marrow-derived DCs were transduced with AdV encoding human IL-10 or green fluorescent protein (GFP). Whereas transduction of immature DCs with AdV/GFP resulted in dose-dependent maturation, DCs transduced with Adv/IL-10 maintained an immature state with low major histocompatibility complex (MHC) class II, CD86, and IL-12 expression. The Adv/IL-10 transduced DCs were phenotypically unique, characterized by suppression of IL-12 expression, failure to stimulate Th1 or Th2 cytokine responses, and retained capacity to endocytose antigen. Importantly, Adv/IL-10-transduced DCs were biologically active in vivo, in that administration of these DCs into mice before a generalized peritonitis significantly improved survival. We conclude that Adv/IL-10 transduction of DCs provides an efficient means to modulate DC function. The capacity to modify DCs by adenoviral expression of IL-10 may provide a novel ex vivo or in vivo approach to mitigate acute and chronic inflammatory diseases like sepsis.


Assuntos
Adenoviridae/genética , Células Dendríticas/citologia , Interleucina-10/genética , Interleucina-10/uso terapêutico , Sepse/terapia , Adenoviridae/metabolismo , Animais , Antígenos CD/biossíntese , Antígeno B7-2 , Complexo CD3/biossíntese , Antígenos CD40/biossíntese , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Endocitose , Feminino , Citometria de Fluxo , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Interleucina-12/metabolismo , Lipopolissacarídeos/metabolismo , Linfonodos/patologia , Linfócitos/citologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Sepse/metabolismo , Linfócitos T/citologia , Células Th1 , Células Th2/metabolismo , Fatores de Tempo
7.
Hum Gene Ther ; 16(1): 109-25, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15703494

RESUMO

As virus vectors for gene therapy approach the goal of successful clinical treatment, it is increasingly necessary for the product to be fully characterized. Empty capsids are perhaps the main extraneous component of recombinant adenovirus (rAd) products that are purified by column chromatography. Two diverse rAd products, one a replication-defective rAd and the other a conditionally replicating rAd, show different protein compositions of their empty capsids. The empty capsid type from the replication-defective rAd carrying the gene for p53 was previously determined to have approximately 1400 copies per particle of pVIII, the precursor to the hexon-associated protein VIII (Vellekamp et al., Hum. Gene Ther. 2001;12:1923-1936). Quantification of this protein is a useful measure of the amount of empty capsids in preparations of this vector. Here we purify and characterize empty capsids from the conditionally replicating rAd. This empty capsid type lacks any appreciable amount of pVIII but contains pVI and multiple forms of the L1 52/55K protein, mostly as disulfidelinked oligomers. Empty capsid from conditionally replicating rAd present new challenges in terms of its quantification, but sodium dodecyl sulfate-polyacrylamide gel electrophoresis densitometry analysis suggests that the amount of this empty capsid in a preparation, like that of rAd p53 empty capsid, declines with increased time of infection. This empty capsid demonstrates heterogeneity by anion-exchange chromatography, electron microscopy, and CsCl density gradient centrifugation.


Assuntos
Adenoviridae/fisiologia , Capsídeo/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Replicação Viral , Sequência de Aminoácidos , Animais , Capsídeo/química , Capsídeo/ultraestrutura , Linhagem Celular , Separação Celular , Centrifugação com Gradiente de Concentração , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Genes p53/genética , Terapia Genética , Humanos , Espectrometria de Massas , Microscopia Eletrônica , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Coelhos , Proteínas Virais/metabolismo
8.
J Endotoxin Res ; 10(6): 393-401, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15588421

RESUMO

Adenovirus-based gene therapy offers a unique opportunity to target gene expression to the liver by systemic delivery. However, systemic administration of a first generation adenoviral construct elicits an inflammatory response leading to TNF-alpha-dependent liver injury. The aim of this study was to evaluate whether the systemic administration of recombinant adenovirus exacerbates a subsequent TNF-alpha-dependent liver injury induced by D-galactosamine and lipopolysaccharide. Surprisingly, low-dose adenovirus administration (10(5) particles) protects, while high-dose adenovirus (10(10) particles) is associated with an exaggerated hepatic inflammatory response from a subsequent D-galactosamine and lipopolysaccharide challenge. This exacerbation is TNF-alpha dependent, since treatment with a TNF inhibitor fully protects against the liver injury. Moreover, intravenous administration of an adenoviral construct expressing the anti-inflammatory protein interleukin-10 reduces TNF-alpha appearance and attenuates the increased hepatocyte injury. Taken together, this report demonstrates potential additive effects of TNF-alpha responses induced by adenovirus and other inflammatory signals, and suggests that the response can be mitigated by relative adenovirus particle dose or by inhibitors, such as TNF-binding protein or interleukin 10.


Assuntos
Adenoviridae/genética , Terapia Genética , Lipopolissacarídeos/toxicidade , Hepatopatias/terapia , Adenoviridae/imunologia , Animais , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas , Relação Dose-Resposta Imunológica , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Injeções Intraperitoneais , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolissacarídeos/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/patologia , Hepatopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
9.
Hum Immunol ; 65(11): 1344-55, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15556685

RESUMO

Recombinant adenoviruses (rAd) are efficient tools for genetic modification of human dendritic cells (DC) in vitro. Infection of DCs by rAd encoding beta-galactosidase (betagal) results in partial maturation of DCs, as witnessed by the upregulation of major histocompatibility complex and costimulatory molecules. Accordingly, these DCs are more potent stimulators of Th1-type proliferative responses. We now demonstrate that infection of immature DCs with rAd encoding human interleukin (IL)-10 results in the secretion by the DCs of large amounts of IL-10, while not affecting expression of activation markers indicative of partial DC maturation. In contrast to rAd-betagal-infected DCs, rAdIL-10-infected DCs are very poor stimulators of monoclonal and polyclonal Th1-type responses. Instead, stimulation of nonpolarized CD4+ T-cell cultures with rAdIL-10-infected DCs selectively activates and expands an IL-10-producing CD4+ T-cell subset capable of suppressing Th1 responses in vitro. Our data argue that rAd-infected human DCs genetically engineered to produce IL-10 may be exploited for the modulation of harmful Th1-type responses in transplantation and autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Interleucina-10/genética , Ativação Linfocitária/imunologia , Células Th1/imunologia , Adenoviridae/genética , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Proliferação de Células , Citocinas/metabolismo , Células Dendríticas/metabolismo , Vetores Genéticos/genética , Humanos , Interleucina-10/metabolismo , Células Th1/metabolismo , Transfecção
10.
Oncogene ; 22(23): 3645-54, 2003 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12789273

RESUMO

The temporal gene expression profile during the entire process of apoptosis and cell cycle progression in response to p53 in human ovarian cancer cells was explored with cDNA microarrays representing 33 615 individual human genes. A total of 1501 genes (4.4%) were found to respond to p53 (approximately 80% of these were repressed by p53) using 2.5-fold change as a cutoff. It was anticipated that most of p53 responsive genes resulted from the secondary effect of p53 expression at late stage of apoptosis. To delineate potential p53 direct and indirect target genes during the process of apoptosis and cell cycle progression, microarray data were combined with global p53 DNA-binding site analysis. Here we showed that 361 out of 1501 p53 responsive genes contained p53 consensus DNA-binding sequence(s) in their regulatory region, approximately 80% of which were repressed by p53. This is the first time that a large number of p53-repressed genes have been identified to contain p53 consensus DNA-binding sequence(s) in their regulatory region. Hierarchical cluster analysis of these genes revealed distinct temporal expression patterns of transcriptional activation and repression by p53. More genes were activated at early time points, while more repressed genes were found after the onset of apoptosis. A small-scale quantitative chromatin immunoprecipitation analysis indicated that in vivo p53-DNA interaction was detected in eight out of 10 genes, most of which were repressed by p53 at the early onset of apoptosis, suggesting that a portion of p53 target genes in the human genome could be negatively regulated by p53 via sequence-specific DNA binding. The approaches and genes described here should aid the understanding of global gene regulatory network of p53.


Assuntos
Apoptose/genética , Ciclo Celular/genética , Neoplasias Ovarianas/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Sequência de Aminoácidos , Sítios de Ligação , Cromatina/metabolismo , Análise por Conglomerados , Sequência Conservada , Feminino , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/patologia , Testes de Precipitina , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transfecção/métodos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
11.
Cancer Gene Ther ; 10(3): 224-38, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12637944

RESUMO

A cohort study was designed to evaluate the efficiency of gene transfer and whether biological activity from the expressed therapeutic gene resulted after administration of a recombinant adenovirus containing the human wild-type p53 (p53(wt)) gene (rAd-p53 SCH 58500). The cohort study was conducted in five trial subjects with recurrent ovarian cancer. Each trial subject received multiple cycles of rAd-p53 SCH 58500, each cycle comprised of doses of 7.5 x 10(13) particles on each of five consecutive days. Subjects were treated with rAd-p53 SCH 58500 alone during Cycle 1 and in combination with gemcitabine during the subsequent cycles. Both tumor biopsies and peritoneal aspirates were collected and evaluated for gene transfer and evidence of the biological activities of the expressed p53(wt) gene. Using quantitative PCR and RT-PCR, and in situ PCR, gene transfer and expression were documented in tumor biopsies (four of five patients) collected from Cycle 1. Furthermore, upregulation of p21/WAF1, bax and mdm-2, and downregulation of survivin were observed in these same tumor biopsy samples, suggesting that intraperitoneal administration of rAd-p53 SCH 58500 leads to detectable p53 biological activity in target tumor tissue. In addition, gene transfer and its expression were observed in cells obtained from peritoneal aspirates. These fluids were mainly comprised of polymorphonuclear neutrophils, indicating that successful gene transfer can be achieved by multiple cycle intraperitoneal administration of recombinant adenovirus.


Assuntos
Adenoviridae/genética , Genes p53/genética , Recidiva Local de Neoplasia/terapia , Neoplasias Ovarianas/terapia , Adenoviridae/crescimento & desenvolvimento , Apoptose , Líquido Ascítico/metabolismo , Líquido Ascítico/patologia , Biópsia , Estudos de Coortes , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/análise , Ciclinas/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos , Humanos , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/metabolismo , Vírion/crescimento & desenvolvimento
12.
J Immunol ; 168(7): 3412-8, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11907099

RESUMO

The dendritic cell (DC) is the most potent APC of the immune system, capable of stimulating naive T cells to proliferate and differentiate into effector T cells. Recombinant adenovirus (Adv) readily transduces DCs in vitro allowing directed delivery of transgenes that modify DC function and immune responses. In this study we demonstrate that footpad injection of a recombinant Adv readily targets transduction of myeloid and lymphoid DCs in the draining popliteal lymph node, but not in other lymphoid organs. Popliteal DCs transduced with an empty recombinant Adv undergo maturation, as determined by high MHC class II and CD86 expression. However, transduction with vectors expressing human IL-10 limit DC maturation and associated T cell activation in the draining lymph node. The extent of IL-10 expression is dose dependent; transduction with low particle numbers (10(5)) yields only local expression, while transduction with higher particle numbers (10(7) and 10(10)) leads additionally to IL-10 appearance in the circulation. Furthermore, local DC expression of human IL-10 following in vivo transduction with low particle numbers (10(5)) significantly improves survival following cecal ligation and puncture, suggesting that compartmental modulation of DC function profoundly alters the sepsis-induced immune response.


Assuntos
Adenovírus Humanos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interleucina-10/biossíntese , Sepse/imunologia , Sepse/mortalidade , Adenovírus Humanos/genética , Animais , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/virologia , Relação Dose-Resposta Imunológica , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Injeções Subcutâneas , Interleucina-10/sangue , Interleucina-10/genética , Interleucina-6/antagonistas & inibidores , Interleucina-6/sangue , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Sepse/microbiologia , Análise de Sobrevida , Timo/enzimologia , Timo/patologia , Regulação para Cima/genética , Regulação para Cima/imunologia
13.
J Clin Oncol ; 20(4): 957-65, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11844817

RESUMO

PURPOSE: To study safety, feasibility, and biologic activity of adenovirus-mediated p53 gene transfer in patients with bladder cancer. PATIENTS AND METHODS: Twelve patients with histologically confirmed bladder cancer scheduled for cystectomy were treated on day 1 with a single intratumoral injection of SCH 58500 (rAd/p53) at cystoscopy at one dose level (7.5 x 10(11) particles) or a single intravesical instillation of SCH 58500 with a transduction-enhancing agent (Big CHAP) at three dose levels (7.5 x 10(11) to 7.5 x 10(13) particles). Cystectomies were performed in 11 patients on day 3, and transgene expression, vector distribution, and biologic markers of transgene activity were assessed by molecular and immunohistochemical methods in tumors and normal bladder samples. RESULTS: Specific transgene expression was detected in tissues from seven of eight assessable patients treated with intravesical instillation of SCH 58500 but in none of three assessable patients treated with intratumoral injection of SCH 58500. Induction of RNA and protein expression of the p53 target gene p21/WAF1 was demonstrated in samples from patients treated with SCH 58500 instillation at higher dose levels. Distribution studies after intravesical instillation of SCH 58500 revealed both high transduction efficacy and vector penetration throughout the whole urothelium and into submucosal tumor cells. No dose-limiting toxicity was observed, and side effects were local and of transient nature. CONCLUSION: Intravesical instillation of SCH 58500 combined with a transduction-enhancing agent is safe, feasible, and biologically active in patients with bladder cancer. Studies to evaluate the clinical efficacy of this treatment in patients with localized high-risk bladder cancer are warranted.


Assuntos
Técnicas de Transferência de Genes , Genes p53 , Terapia Genética , Vetores Genéticos/uso terapêutico , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/terapia , Adenoviridae/genética , Administração Intravesical , Adulto , Idoso , Idoso de 80 Anos ou mais , Cistectomia , Primers do DNA , Relação Dose-Resposta a Droga , Humanos , Pessoa de Meia-Idade , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA