Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biochem ; 105(4): 1092-108, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18816594

RESUMO

mrtl (myc-related translation/localization regulatory factor) is a previously uncharacterized protein synthesized from the first open reading frame contained within the human c-myc P0 transcript, approximately 800 nucleotides upstream of the Myc coding sequence. The mrtl protein, 114 amino acids in length, is projected to contain an N-terminal transmembrane domain and a highly charged C-terminal interaction domain with homology to numerous RNA-binding proteins. Using monoclonal antibodies raised against the hydrophilic C-terminal domain, endogenous mrtl was visualized in human breast tumor cell lines and primary mammary epithelial cells at the nuclear envelope and contiguous endoplasmic/nucleoplasmic reticulum. mrtl colocalizes and coimmunoprecipitates with translation initiation factor eIF2alpha and the 40S ribosomal protein RACK1, and appears capable of binding specifically to the c-myc RNA. Inducible ectopic overexpression of wild-type mrtl interferes with the function of endogenous mrtl, which results in loss of Myc from the nucleus. Furthermore, treatment of cells with a peptide derived from the C-terminal domain displaces endogenous mrtl and causes a dramatic reduction in total cellular Myc protein levels. Together with our previous work demonstrating complete loss of tumorigenicity in association with ectopic expression of the c-myc P0 5'-UTR (containing the mrtl coding sequence), these results suggest that mrtl may serve an important function in regulating Myc translation and localization to the nucleus, perhaps ultimately contributing to the role of the c-myc locus in oncogenesis.


Assuntos
Proteínas de Transporte , Retículo Endoplasmático/química , Matriz Nuclear/química , Proteínas Proto-Oncogênicas c-myc/genética , Transporte Ativo do Núcleo Celular , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Fases de Leitura Aberta , Biossíntese de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA
2.
Front Biosci ; 11: 1189-98, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146806

RESUMO

DNA vaccines, also referred to as genetic, plasmid or polynucleotide vaccines, represent a relatively simple and economical method to exploit gene transfer for immunization against tumor associated antigens. This review discusses the potential advantages of DNA vaccines for cancer immunotherapy as compared to conventional protein vaccines and viral vectors. The proposed mechanisms responsible for induction of immune responses following DNA-based immunization are summarized. The preclinical development of DNA vaccines and the clinical experience with DNA immunization for cancer to date are reviewed. The low toxicity associated with DNA vaccines favors its further development, but additional strategies to improve the potency of this approach are needed if it is to be successfully integrated into the clinical setting.


Assuntos
Vacinas Anticâncer/uso terapêutico , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Vacinas de DNA/uso terapêutico , Animais , Antígenos de Neoplasias , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Sistema Imunitário , Neoplasias/metabolismo , Neoplasias/patologia , Plasmídeos/metabolismo , Resultado do Tratamento
3.
Hum Gene Ther ; 16(9): 1047-57, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16149903

RESUMO

Studies have demonstrated that packaging of recombinant adeno-associated virus 2 (rAAV) as self-complementary duplex strand (sc) results in early transgene expression, possibly eliminating rate-limiting second-strand synthesis. In the present study, we evaluated the molecular organization, stability of the sc AAV genome, and transgene expression in the quadriceps muscle of C57BL/6J mice in vivo as compared with single-stranded (ss) AAV. Studies were carried out with rAAV encoding green fluorescent protein (GFP) or human carcinoembryonic antigen (CEA) either as single-stranded or self-complementary duplex strand structures, encapsidated in AAV-2 capsids. Mice were injected with 10(11) particles of the respective viruses and the vector-injected muscles were harvested 1 week, 2 weeks, 3 weeks, or 2 months later. Tissues were processed for total DNA isolation for the analyses of vector genomic configuration and copy number, and for immunostaining of transgene expression. ELISA was done on serum samples to quantitate CEA-specific humoral immune response as a correlate of transgene expression. Results of Southern blot and PCR analyses indicated more disintegration of the monomeric ss AAV DNA in vivo compared with linear sc AAV DNA. The results also indicated efficient conversion of the self-complementary duplex-stranded vector genome to dimer during early time points. As expected, transgene expression was detected at early time points with self-complementary duplex-stranded vector and persisted stably. However, the advantage of higher transgene expression from sc AAV was balanced over time by the single-stranded vector. These data demonstrate that sc AAV provides better stability for transgene structure during the initial stages of transduction and may have better utility in AAV gene therapy in situations, which mandate early transgene expression.


Assuntos
DNA Complementar/genética , Dependovirus/genética , Vetores Genéticos/genética , Instabilidade Genômica , Músculo Esquelético/efeitos dos fármacos , Transdução Genética/métodos , Transgenes/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , DNA Complementar/administração & dosagem , DNA Complementar/análise , Ensaio de Imunoadsorção Enzimática , Terapia Genética , Humanos , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase , Transgenes/efeitos dos fármacos
4.
Vaccine ; 23(10): 1273-83, 2005 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-15652670

RESUMO

The anti-tumor immunologic effects of plasmid DNA vaccines encoding human carcinoembryonic antigen (CEA) fused to mouse granulocyte/macrophage colony-stimulating factor (GM-CSF) were examined. Immunization of C57BL/6 mice with the CEA-GMCSF fusion plasmids in a three injection, high-dose immunization schedule led to T cell and antibody responses specific for CEA. Mice injected with CEA-GMCSF fusion plasmids also developed IgG autoantibodies to GM-CSF. Tumor challenge with the CEA-expressing syngeneic mouse adenocarcinoma line, MC38-CEA-2, showed delayed tumor growth in mice immunized with the CEA-GMCSF fusion plasmids but complete protection in mice immunized with plasmid encoding CEA alone. In contrast, a single low-dose immunization with CEA-GMCSF fusion plasmids provided better tumor protection than low-dose CEA plasmid alone and resulted in lower titers of GM-CSF antibodies.


Assuntos
Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/prevenção & controle , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Western Blotting , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Linhagem Celular Tumoral , Citocinas/metabolismo , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunização , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Plasmídeos/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Vacinas de DNA
5.
Int J Cancer ; 113(3): 406-14, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15455351

RESUMO

Deficiency in costimulatory molecule expression has been implicated in the ability of tumors to escape immune effectors. The activity of the intratumoral administration of recombinant fowlpox vectors expressing a triad of costimulatory molecules (rF-TRICOM) was evaluated in the asbestos-induced AB12 and AC29 mouse models of mesothelioma. Mesothelioma cell infected with rF-TRICOM expressed high levels of the costimulatory molecules. Prolongation of survival was observed in mice receiving rF-TRICOM in AB12 and AC29 intraperitoneal models. Complete tumor regressions were observed in mice receiving intratumoral rF-TRICOM in the AB12 subcutaneous tumor model. Tumor regressions were associated with the development of serum IgG reactivities to mesothelioma-associated determinants and specific systemic cytolytic activity, and responding mice were capable of rejecting tumors upon re-challenge. Antitumor activity was also observed in mice with established AB12 tumor vaccinated with irradiated rF-TRICOM-infected AB12 cells. The antitumor activity of intratumoral rF-TRICOM was superior to that of the intratumoral injection of a fowlpox vector expressing granulocyte-macrophage colony stimulating factor (rF-GM-CSF). AB12 and AC29 tumors were found to produce GM-CSF and to have substantial macrophage infiltration. Production of GM-CSF decreased in vivo in tumors injected with rF-TRICOM. rF-TRICOM and wild-type fowlpox inhibited the growth of AB12 and AC29 cells in vitro; less inhibition was observed with rF-GM-CSF. These results indicate that the intratumoral injection of rF-TRICOM has significant activity in mouse models of mesothelioma and can elicit a systemic antitumor immune response. The results also suggest potential limitations to the intratumoral administration of cytokines, such as GM-CSF, in mesothelioma.


Assuntos
Varíola Aviária/genética , Vetores Genéticos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Mesotelioma/terapia , Vacinas Sintéticas/uso terapêutico , Animais , Amianto/toxicidade , Aves , Vacinas Anticâncer , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , DNA Recombinante , Feminino , Varíola Aviária/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunoglobulina G/sangue , Mesotelioma/induzido quimicamente , Mesotelioma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Taxa de Sobrevida , Vacinação , Vacinas Sintéticas/genética
7.
Hum Gene Ther ; 15(9): 856-64, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15353040

RESUMO

Recombinant adeno-associated virus 2 (rAAV) vectors have been successfully used for sustained expression of therapeutic genes. The potential of using rAAV as a cancer vaccine vector and the impact of a bacterial plasmid adjuvant on this activity were investigated. C57BL/6 mice received a single intramuscular injection of rAAV expressing the human tumor-associated antigen, carcinoembryonic antigen (CEA). Three weeks later, when CEA expression was optimal, a bacterial plasmid containing methylated DNA motifs was injected into the same muscle. Mice were challenged 1 week later with syngeneic MC38 tumor cells stably expressing CEA. Immunization with rAAV-CEA alone resulted in sustained transgene expression and the elicitation of a humoral immune response to CEA. Cellular immune response, however, was weak, and tumor protection was not significant. In contrast, immunization with rAAV-CEA and the plasmid adjuvant resulted in stronger cellular immune response to CEA and tumor protection. The addition of plasmid adjuvant increased both myeloid dendritic cell recruitment in situ and CEA-specific T-helper-1-associated immune response. These data indicate that robust rAAV transgene expression of a tumor antigen followed by transient plasmid delivery to recruit and activate dendritic cells is an effective method of eliciting antitumor cellular immune responses.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Vacinas Anticâncer , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Dependovirus/genética , Neoplasias Experimentais/prevenção & controle , Plasmídeos/uso terapêutico , Animais , Anticorpos Antineoplásicos/sangue , Anticorpos Antivirais/sangue , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Clonagem Molecular , Terapia Combinada , Citocinas/genética , Citocinas/imunologia , Citomegalovirus/genética , DNA Recombinante/genética , Dependovirus/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Transplante de Neoplasias/imunologia , Neoplasias Experimentais/imunologia , Regiões Promotoras Genéticas , Células Th1/imunologia
8.
Int J Cancer ; 111(2): 303-9, 2004 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-15197787

RESUMO

Treatment options for disseminated cervical cancer remain inadequate. Here, we investigated a strategy featuring Ad5-Delta 24 RGD, an oncolytic adenovirus replication-competent selectively in cells defective in the Rb-p16 pathway, such as most cervical cancer cells. The viral fiber contains an alpha(v)beta(3) and alpha(v)beta(5) integrin-binding RGD-4C motif, allowing coxsackie-adenovirus receptor-independent infection. These integrins have been reported to be frequently upregulated in cervical cancer. Oncolysis of cervical cancer cells was similar to a wild-type control in vitro. In an animal model of cervical cancer, the therapeutic efficacy of Ad5-Delta 24 RGD could be demonstrated for both intratumoral and intravenous application routes. Biodistribution was determined following intravenous administration to mice. Further preclinical safety data were obtained by demonstrating lack of replication of the agent in human peripheral blood mononuclear cells. These results suggest that Ad5-Delta 24 RGD could be useful for local or systemic treatment of cervical cancer in patients with disease resistant to currently available modalities.


Assuntos
Adenoviridae , Neoplasias do Colo do Útero/terapia , Animais , Terapia Biológica/métodos , Modelos Animais de Doenças , Feminino , Humanos , Infusões Intravenosas , Leucócitos Mononucleares , Camundongos , Distribuição Tecidual , Células Tumorais Cultivadas , Replicação Viral
9.
BMC Cancer ; 4: 19, 2004 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15140265

RESUMO

BACKGROUND: Mesothelin is an attractive target for cancer immunotherapy due to its restricted expression in normal tissues and high level expression in several tumor types including ovarian and pancreatic adenocarcinomas. Three mesothelin transcript variants have been reported, but their relative expression in normal tissues and tumors has been poorly characterized. The goal of the present study was to clarify which mesothelin transcript variants are commonly expressed in human tumors. METHODS: Human genomic and EST nucleotide sequences in the public databases were used to evaluate sequences reported for the three mesothelin transcript variants in silico. Subsequently, RNA samples from normal ovary, ovarian and pancreatic carcinoma cell lines, and primary ovarian tumors were analyzed by reverse transcription-polymerase chain reaction (RT-PCR) and nucleotide sequencing to directly identify expressed transcripts. RESULTS: In silico comparisons of genomic DNA sequences with available EST sequences supported expression of mesothelin transcript variants 1 and 3, but there were no sequence matches for transcript variant 2. Newly-derived nucleotide sequences of RT-PCR products from tissues and cell lines corresponded to mesothelin transcript variant 1. Mesothelin transcript variant 2 was not detected. Transcript variant 3 was observed as a small percentage of total mesothelin amplification products from all studied cell lines and tissues. Fractionation of nuclear and cytoplasmic RNA indicated that variant 3 was present primarily in the nuclear fraction. Thus, mesothelin transcript variant 3 may represent incompletely processed hnRNA. CONCLUSION: Mesothelin transcript variant 1 represents the predominant mature mRNA species expressed by both normal and tumor cells. This conclusion should be important for future development of cancer immunotherapies, diagnostic tests, and gene microarray studies targeting mesothelin.


Assuntos
Antígenos de Neoplasias/análise , Glicoproteínas de Membrana/análise , Neoplasias Ovarianas/química , Pâncreas/química , Neoplasias Pancreáticas/química , Biomarcadores Tumorais/análise , Carcinoma Endometrioide/química , Linhagem Celular Tumoral , Cistadenocarcinoma Papilar/química , Feminino , Proteínas Ligadas por GPI , Humanos , Masculino , Mesotelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
10.
Cancer Res ; 64(5): 1781-7, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14996740

RESUMO

Angiogenesis is characteristic of solid tumor growth and a surrogate marker for metastasis in many human cancers. Inhibition of tumor angiogenesis using antiangiogenic drugs and gene transfer approaches has suggested the potential of this form of therapy in controlling tumor growth. However, for long-term tumor-free survival by antiangiogenic therapy, the factors controlling tumor neovasculature need to be systemically maintained at stable therapeutic levels. Here we show sustained expression of the antiangiogenic factors angiostatin and endostatin as secretory proteins by recombinant adeno-associated virus 2 (rAAV)-mediated gene transfer. Both vectors provided significant protective efficacy in a mouse tumor xenograft model. Stable transgene persistence and systemic levels of both angiostatin and endostatin were confirmed by in situ hybridization of the vector-injected tissues and by serum ELISA measurements, respectively. Whereas treatment with rAAV containing either endostatin or angiostatin alone resulted in moderate to significant protection, the combination of endostatin and angiostatin gene transfer from a single vector resulted in a complete protection. These data suggest that AAV-mediated long-term expression of both endostatin and angiostatin may have clinical utility against recurrence of cancers after primary therapies and may represent rational adjuvant therapies in combination with radiation or chemotherapy.


Assuntos
Inibidores da Angiogênese/genética , Angiostatinas/genética , Dependovirus/genética , Endostatinas/genética , Terapia Genética , Vetores Genéticos/genética , Neoplasias/terapia , Angiostatinas/sangue , Animais , Linhagem Celular , Endostatinas/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo
11.
Int J Cancer ; 108(6): 805-11, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14712480

RESUMO

Sperm protein 17 (Sp17) is an antigenic protein highly expressed in spermatozoa. Sp17 expression was demonstrated recently in multiple myeloma, suggesting that it may be a novel cancer-testis antigen. Expression of Sp17 mRNA and protein was examined in human ovarian tumors. Sp17 mRNA was evaluated by reverse transcription-polymerase chain reaction (RT-PCR) and Northern blot analysis of RNA derived from epithelial ovarian tumors and normal tissues. RT-PCR analysis detected Sp17 transcripts in 15 of 18 (83%) primary ovarian tumors. The transcript was not detected in RNA derived from normal uterus or cervix, whereas weak expression was noted in some normal ovarian tissue samples. Northern blot analysis showed no detectable Sp17 mRNA expression in normal tissues, including normal ovary, but showed Sp17 expression in 17 of 25 ovarian tumors (68%). To evaluate protein expression, mouse monoclonal antibodies were produced against recombinant Sp17 protein and used in Western blot and immunohistochemical analyses of normal reproductive tissue and primary ovarian tumor samples. Sp17 protein was detected by Western blot analysis in normal spermatozoa and in 8 of 19 ovarian tumor samples. Immunohistochemical studies showed Sp17 expression in spermatozoa, ciliated cells of the female reproductive tract, and most ovarian tumors evaluated. Tumors showed a predominantly nuclear localization of Sp17 expression, with some cytoplasmic staining. These results demonstrate that Sp17, a protein with restricted expression in somatic tissues, is expressed in ovarian tumors. Because Sp17 is immunogenic, it may represent a novel target for immunotherapeutic interventions for ovarian cancer patients.


Assuntos
Proteínas de Transporte/biossíntese , Neoplasias Ovarianas/metabolismo , Anticorpos Monoclonais/metabolismo , Antígenos de Superfície , Northern Blotting , Western Blotting , Proteínas de Ligação a Calmodulina , Proteínas de Transporte/genética , Colo do Útero/metabolismo , Clonagem Molecular , Citoplasma/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Proteínas de Membrana , Ovário/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual , Útero/metabolismo
12.
Vaccine ; 22(5-6): 773-80, 2004 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-14741172

RESUMO

KSA (Ep-CAM) is highly expressed by colorectal cancers. The safety and immunologic effects of a vaccine consisting of recombinant baculovirus-derived KSA formulated with monophosphoryl lipid A (MPL) in liposomes and emulsified in mineral oil were evaluated, with and without co-administration of granulocyte-macrophage colony-stimulating factor (GM-CSF). Eleven patients with metastatic colorectal cancer received three subcutaneous (s.c.) injections of the vaccine at 4-week intervals. Six patients were randomized to also receive human recombinant GM-CSF (rGM-CSF) by subcutaneous injection daily for 4 days with each vaccination. Immunizations with and without rGM-CSF were well tolerated. Seven of the 11 patients developed significant KSA-specific cellular immune responses as assessed by lymphoproliferation and interferon-gamma (IFN-gamma) ELISPOT assays. All nine tested patients developed positive delayed type hypersensitivity reactions. Eight of the 11 patients developed KSA-specific antibody responses. The highest levels of cellular immune responses were observed in patients who received GM-CSF. Immunization with baculovirus-derived KSA formulated with monophosphoryl lipid A in liposomal emulsion is safe and can elicit KSA-specific immune responses. Co-administration of GM-CSF with this formulation is an effective method of generating KSA-specific T-helper (Th) 1-associated cellular immune responses.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos de Neoplasias/imunologia , Baculoviridae/imunologia , Vacinas Anticâncer/imunologia , Moléculas de Adesão Celular/imunologia , Neoplasias Colorretais/imunologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Lipídeo A/química , Adulto , Idoso , Anticorpos Antineoplásicos/análise , Anticorpos Antineoplásicos/biossíntese , Formação de Anticorpos/imunologia , Reações Antígeno-Anticorpo , Vacinas Anticâncer/efeitos adversos , Divisão Celular , Química Farmacêutica , Emulsões , Ensaio de Imunoadsorção Enzimática , Molécula de Adesão da Célula Epitelial , Escherichia coli/imunologia , Feminino , Humanos , Imunidade Celular/imunologia , Imunização , Interferon gama/biossíntese , Lipossomos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Óleo Mineral , Projetos Piloto , Proteínas Recombinantes , Testes Cutâneos , Vacinas Sintéticas/imunologia
13.
AIDS Res Hum Retroviruses ; 19(9): 817-23, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14585212

RESUMO

Codon usage optimization of human immunodeficiency virus type 1 (HIV-1) structural genes has been shown to increase protein expression in vitro as well as in the context of DNA vaccines in vivo; however, all optimized genes reported thus far are derived from HIV-1 (group M) subtype B viruses. Here, we report the generation and biological characterization of codon usage-optimized gag, pol, env (gp160, gp140, gp120), and nef genes from a primary (nonrecombinant) HIV-1 subtype C isolate. After transfection into 293T cells, optimized subtype C genes expressed one to two orders of magnitude more protein (as determined by immunoblot densitometry) than the corresponding wild-type constructs. This effect was most pronounced for gp160, gp140, Gag, and Pol (>250-fold), but was also observed for gp120 and Nef (45- and 20-fold, respectively). Optimized gp160- and gp140-derived glycoproteins were processed, incorporated into virus particles, and mediated virus entry when expressed in trans to complement an env-minus HIV-1 provirus. Mice immunized with optimized gp140 DNA developed antibody as well as CD4+ and CD8+ T cell immune responses that were orders of magnitude greater than those of mice immunized with wild-type gp140 DNA. These data confirm and extend previous studies of codon usage optimization of HIV-1 genes to the most prevalent group M subtype. Our panel of matched optimized and wild-type subtype C genes should prove valuable for studies of protein expression and function, the generation of subtype-specific immunological reagents, and the production of DNA-based sub-unit vaccines directed against a broader spectrum of viruses.


Assuntos
Vacinas contra a AIDS/imunologia , Códon , Genes env , Genes gag , Genes nef , Genes pol , HIV-1/genética , Vacinas de DNA/imunologia , Animais , Feminino , HIV-1/classificação , HIV-1/imunologia , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C
14.
Clin Cancer Res ; 8(9): 2782-7, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12231517

RESUMO

Despite an abundance of preclinical data, relatively little is known regarding the efficacy of DNA vaccination in humans. Here, we present results from a dose-escalation clinical trial of a dual expression plasmid encoding carcinoembryonic antigen (CEA) and hepatitis B surface antigen (HBsAg) in 17 patients with metastatic colorectal carcinoma. CEA was selected as a prototypic tumor-associated self-antigen, and the HBsAg cDNA was included as a positive control for immune response to the DNA vaccine without relying upon breaking tolerance to a self-antigen. Groups of 3 patients received escalating single i.m. doses of the DNA vaccine at 0.1, 0.3, and 1.0 mg. Subsequent groups of 3 patients received three repetitive 0.3- or 1.0-mg doses at 3-week intervals. A final group of 2 patients received three repetitive 2.0 mg doses at 3-week intervals. Toxicity was limited to transient grade 1 injection site tenderness, fatigue, and creatine kinase elevations, each affecting a minority of patients in a non-dose-related manner. Repetitive dosing of the DNA vaccine induced HBsAg antibodies in 6 of 8 patients, with protective antibody levels achieved in four of these patients. CEA-specific antibody responses were not observed, but 4 of 17 patients developed lymphoproliferative responses to CEA after vaccination. No objective clinical responses to the DNA vaccine were observed among this population of patients with widely metastatic colorectal carcinoma. Nevertheless, this pilot trial has provided encouraging human immune response data in support of this vaccine technology.


Assuntos
Adenocarcinoma/terapia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Antígenos de Superfície da Hepatite B/imunologia , Imunoterapia Ativa , Vacinas de DNA/uso terapêutico , Adenocarcinoma/imunologia , Idoso , Anemia/induzido quimicamente , Antígenos de Neoplasias/genética , Proteína C-Reativa/análise , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/genética , Neoplasias Colorretais/imunologia , Creatina Quinase/sangue , Relação Dose-Resposta Imunológica , Feminino , Anticorpos Anti-Hepatite B/biossíntese , Antígenos de Superfície da Hepatite B/genética , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Segurança , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...