Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Small Anim Pract ; 62(9): 719-729, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34018618

RESUMO

Hyperbaric oxygen therapy is in wide use in human medicine around the world. Although hyperbaric oxygen therapy is available for veterinary use, it is still significantly underutilised. The physical principles, gas laws and physiologic mechanisms by which hyperbaric oxygen therapy is therapeutic, especially in traumatic injuries and complicated wound care, are discussed. Then, considerations are offered for the implementation of hyperbaric oxygen therapy in veterinary practices. Finally, a review of clinical indications for veterinary practices, including a presentation of select literature, is provided. Applying hyperbaric oxygen therapy in an earlier and more consistent manner could improve short- and long-term outcomes in complicated wounds. The authors also hope this information may stimulate interest in the design of future, prospective studies for the various clinical situations described.


Assuntos
Oxigenoterapia Hiperbárica , Animais , Oxigenoterapia Hiperbárica/veterinária , Estudos Prospectivos
2.
Cancer Gene Ther ; 23(6): 178-87, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27151235

RESUMO

Interleukin-2 (IL-2) has been used clinically for the treatment of some malignancies, but the toxicities associated with systemic IL-2 therapy are a major challenge. Here we have determined whether transcriptional targeting of IL-2 to breast cancer (BrCa) using an engineered human mammaglobin promoter/enhancer (MPE2) is a feasible option for reducing IL-2-associated toxicities while still achieving a meaningful antitumor effect. We have constructed nonreplicating adenovirus vectors encoding either a reporter gene (luciferase) or human IL-2 (hIL-2) complementary DNA under control of the MPE2 sequence, the murine cytomegalovirus immediate early (MCMV) promoter or the human telomerase reverse transcriptase (hTERT) promoter. Luciferase and hIL-2 complementary DNAs under the control of the MPE2 sequence in adenovirus vectors were expressed at high levels in BrCa cells and at lower levels in normal cells of human and murine origin. Cancer specificity of the hTERT promoter was found to be similar to that of the MPE2 promoter in cells of human origin, but reduced specificity in murine cells. The MPE2 regulatory sequence demonstrated excellent tissue specificity in a mouse tumor model. Whereas the MCMV promoter-controlled IL-2 vector generated high liver toxicity in mice, the MPE2-controlled IL-2 vector generated little or no liver toxicity. Both IL-2 vectors exerted significant tumor growth delay; however, attempts to further enhance antitumor activity of the IL-2 vectors by combining with the proapoptotic drug procaspase activating compound 1 (PAC1) were unsuccessful.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Elementos Facilitadores Genéticos , Vetores Genéticos/genética , Interleucina-2/genética , Regiões Promotoras Genéticas , Secretoglobinas/genética , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Expressão Gênica , Genes Reporter , Terapia Genética , Humanos , Camundongos , Telomerase/genética , Carga Tumoral
3.
Cancer Gene Ther ; 19(12): 888-98, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23099884

RESUMO

Despite the tremendous potential of adenovirus (Ad) as a delivery vector for cancer gene therapy, its use in clinical settings has been limited, mainly as a result of the limited infectivity in many tumors and the wide tissue tropism associated with Ad. To modify the tropism of the virus, we have inserted the epidermal growth factor-like domain of the human heregulin-α (HRG) into the HI loop of Ad5 fiber. This insertion had no adverse effect on fiber trimerization nor did it affect incorporation of the modified fiber into infectious viral particles. Virions bearing modified fiber displayed growth characteristics and viral yields indistinguishable from those of wild-type (wt) virus. Most importantly, HRG-tagged virions showed enhanced infection of cells expressing the cognate receptors HER3/ErbB3 and HER4/ErbB4. This was significantly reduced in the presence of soluble HRG. Furthermore, HER3-expressing Chinese hamster ovary (CHO) cells were transduced by the HRG-modified virus, but not by wt virus. In contrast, CHO cells expressing the coxsackie-Ad receptor were transduced with both viruses. However, infection of an in vivo breast cancer xenograft model after intratumoral injection was similar with both viruses, suggesting that the tumor microenvironment and/or the route of delivery have important roles in infection of target cells with fiber-modified Ads.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/genética , Neoplasias da Mama/virologia , Receptor ErbB-3/metabolismo , Adenoviridae/metabolismo , Animais , Neoplasias da Mama/metabolismo , Cricetinae , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Injeções Intralesionais , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Blood Cancer J ; 2: e82, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22885405

RESUMO

Sox2 (sex-determining region Y-Box) is one of the master transcriptional factors that are important in maintaining the pluripotency of embryonic stem cells (ESCs). In line with this function, Sox2 expression is largely restricted to ESCs and somatic stem cells. We report that Sox2 is expressed in cell lines and tumor samples derived from ALK-positive anaplastic large cell lymphoma (ALK(+)ALCL), for which the normal cellular counterpart is believed to be mature T-cells. The expression of Sox2 in ALK(+)ALCL can be attributed to nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), the oncogenic fusion protein carrying a central pathogenetic role in these tumors. By confocal microscopy, Sox2 protein was detectable in virtually all cells in ALK(+)ALCL cell lines. However, the transcriptional activity of Sox2, as assessed using a Sox2-responsive reporter construct, was detectable only in a small proportion of cells. Importantly, downregulation of Sox2 using short interfering RNA in isolated Sox2(active) cells, but not Sox2(inactive) cells, resulted in a significant decrease in cell growth, invasiveness and tumorigenicity. To conclude, ALK(+)ALCL represents the first example of a hematologic malignancy that aberrantly expresses Sox2, which represents a novel mechanism by which NPM-ALK mediates tumorigenesis. We also found that the transcriptional activity and oncogenic effects of Sox2 can be heterogeneous in cancer cells.

5.
J Vet Intern Med ; 26(4): 888-96, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22676337

RESUMO

BACKGROUND: Pancreas-specific lipase is reported to aid in diagnosing acute pancreatitis (AP) in dogs but has not been rigorously evaluated clinically. HYPOTHESIS/OBJECTIVES: To describe variability of disease in dogs with suspected clinical AP, and to evaluate accuracy of 2 pancreatic-specific lipase immunoassays, Spec cPL (SPEC) and SNAP cPL (SNAP), in diagnosing clinical AP. We hypothesized that SPEC and SNAP provide better diagnostic accuracy than serum amylase or total lipase. ANIMALS: A total of 84 dogs; 27 without AP and 57 with clinical signs associated with AP. METHODS: Multicenter study. Dogs were prospectively enrolled based upon initial history and physical examination, then retrospectively classified into groups according to the likelihood of having clinical AP by a consensus of experts blinded to SPEC and SNAP results. Bayesian latent class analyses were used to estimate the diagnostic accuracy of SPEC and SNAP. RESULTS: The estimates for test sensitivities and specificities, respectively, ranged between 91.5-94.1% and 71.1-77.5% for SNAP, 86.5-93.6% and 66.3-77.0% for SPEC (cutoff value of 200 µg/L), 71.7-77.8% and 80.5-88.0% for SPEC (cutoff value of 400 µg/L), and were 52.4-56.0% and 76.7-80.6% for amylase, and 43.4-53.6% and 89.3-92.5% for lipase. CONCLUSIONS AND CLINICAL IMPORTANCE: SNAP and SPEC have higher sensitivity for diagnosing clinical AP than does measurement of serum amylase or lipase activity. A positive SPEC or SNAP has a good positive predictive value (PPV) in populations likely to have AP and a good negative predictive value (NPV) when there is low prevalence of disease.


Assuntos
Doenças do Cão/diagnóstico , Ensaio de Imunoadsorção Enzimática/veterinária , Lipase/sangue , Pancreatite/veterinária , Doença Aguda , Animais , Teorema de Bayes , Estudos de Casos e Controles , Estudos de Coortes , Doenças do Cão/sangue , Doenças do Cão/enzimologia , Cães , Ensaio de Imunoadsorção Enzimática/normas , Cadeias de Markov , Método de Monte Carlo , Pancreatite/sangue , Pancreatite/enzimologia , Valor Preditivo dos Testes , Estudos Prospectivos , Estudos Retrospectivos , Sensibilidade e Especificidade
6.
Mol Ther ; 4(2): 84-91, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11482978

RESUMO

Gene replacement therapy represents an interesting new approach for the treatment of cystic fibrosis (CF) lung disease. Basic research suggests that CF gene therapy is feasible, but major technological challenges must be addressed before clinical applications are likely to succeed. Therapeutic genes can be delivered to and expressed in human airways, but the number of cells expressing the transgene is relatively low. The inefficiency of gene delivery is largely attributable to the remarkable defenses of human airways. Maintaining long-term transgene expression in airway cells is also a significant obstacle. Recent advances have been made in the development of vectors, expression cassettes, and delivery techniques for enhancing airway gene transfer and expression. These advances have the potential to improve the efficiency of lung gene therapy and to achieve clinical benefits for CF patients in the future.


Assuntos
Fibrose Cística/terapia , Terapia Genética/métodos , Pulmão/fisiologia , Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Vetores Genéticos , Humanos , Pulmão/citologia
7.
Cancer Gene Ther ; 8(5): 361-70, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11477456

RESUMO

Synergy between interleukin-12 (IL-12) and B7-1 (CD80) for cancer immunotherapy has previously been demonstrated in animal models of breast cancer, lymphoma, and multiple myeloma. With a view to human clinical application, tricistronic retroviral and adenovirus vectors co-expressing IL-12 (IL-12p40 plus IL-12p35) and CD80 were constructed by utilizing two internal ribosome entry site (IRES) sequences to link the three cDNAs. A murine stem cell virus (MSCV)-based retroviral vector (MSCV-hIL12.B7) utilized distinct IRES sequences from the encephalomyocarditis virus (EMCV) and the foot-and-mouth disease virus (FMCV), whereas Ad5-based adenovirus vectors contained transcriptional units with two EMCV IRES sequences under the control of murine (AdMh12.B7) or human (AdHh12.B7) cytomegalovirus promoters. AdMh12.B7 was found to consistently direct higher levels of IL-12 and CD80 expression than AdHh12.B7 following infection of a number of human tumor cell lines. In preclinical studies, the human myeloma cell line U266 was infected with MSCV-hIL12.B7 and a resulting clonal cell line, U/MSCV-h12.B7, was generated with stable expression of CD80 and secreting IL-12 at 1 ng/24 h/10(6) cells. By comparison, following AdMh12.B7 infection, 81% of infected U266 cells (U/AdMh12.B7) expressed CD80 and secreted IL-12 at 25-50 ng/24 h/10(6) cells. Both engineered myeloma cell lines stimulated enhanced allogeneic mixed lymphocyte proliferation and provoked increases in cytotoxic T-lymphocyte responses and gamma-interferon release from normal donor lymphocytes exposed to parental U266 cells. These results suggest potential clinical utility of AdMh12.B7 in immunotherapy strategies for the treatment of multiple myeloma and other cancers.


Assuntos
Adenoviridae/genética , Antígeno B7-1/genética , Vetores Genéticos , Imunoterapia/métodos , Interleucina-12/genética , Neoplasias/terapia , Retroviridae/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Antígeno B7-1/metabolismo , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica , DNA Complementar , Quimioterapia Combinada , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Interleucina-12/metabolismo , Neoplasias/metabolismo , Neoplasias/virologia , Linfócitos T/imunologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia
8.
J Immunol ; 166(10): 6212-7, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342643

RESUMO

In this study, we used intratumor delivery of adenoviral vectors to induce a selective anti-tumor response by combining the potent angiogenesis inhibitor murine angiostatin (adenovirus (Ad)-angiostatin) with the powerful immune simulator and angiostatic cytokine murine IL-12 (Ad-IL-12). In a murine model of breast carcinoma, intratumor injection of Ad-angiostatin delayed mean tumor growth, as compared with control virus with an initial regression of tumor growth, in 65% of treated animals. However, all treated animals eventually succumbed to the tumors. Mice injected with Ad-IL-12 alone responded with an initial regression in 20% of treated animals, with only 13% developing a total regression. Coinjection of the vectors resulted in 96% of the treated animals developing an initial regression, with 54% undergoing a total regression of the tumor. These mice were resistant to tumor rechallenge and developed a strong CTL response. Frozen tumor sections were stained for microvessel density using an Ab against murine CD31, an endothelial cell marker. Automated image analysis revealed the mean microvessel density following the administration of Ad-angiostatin and Ad-IL-12 alone or in combination was significantly reduced compared with the control-treated tumor. In summary, we have shown that a short-term course of antiangiogenic therapy combined with immunotherapy can effectively shrink a solid tumor and vaccinate the animal against rechallenge. The rationale for this therapy is to limit the tumor size by attacking the vasculature with angiostatin, thereby allowing IL-12 to mount a T cell-specific response against the tumor AG:


Assuntos
Adenovírus Humanos/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia Ativa , Interleucina-12/genética , Neoplasias Mamárias Experimentais/terapia , Fragmentos de Peptídeos/genética , Plasminogênio/genética , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Inibidores da Angiogênese/administração & dosagem , Angiostatinas , Animais , Antígenos Transformantes de Poliomavirus/biossíntese , Antígenos Transformantes de Poliomavirus/genética , Linhagem Celular Transformada , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Soros Imunes/análise , Imuno-Histoquímica , Imunoterapia Ativa/métodos , Injeções Intralesionais , Injeções Subcutâneas , Interleucina-12/administração & dosagem , Interleucina-12/biossíntese , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/química , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Transplante de Neoplasias , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/biossíntese , Plasminogênio/administração & dosagem , Plasminogênio/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Coloração e Rotulagem , Células Tumorais Cultivadas
9.
Leukemia ; 15(5): 846-54, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11368448

RESUMO

Eight multiple myeloma patients participated in a phase I trial evaluating the feasibility and safety of subcutaneous vaccination with adenovirus engineered, autologous plasma cells after high-dose therapy. Plasma cells were concentrated from bone marrow harvests by negative selection and high gradient magnetic separation. The mean plasma cell yield was 2.61 x 10(8). Transgene expression measured 48 h after plasma cell infection with an IL-2 expressing adenovirus averaged 2.95 ng/ml/10(6) cells. Vaccine production was successful for 88% of patients. Two months after high-dose therapy, six patients received from one to five injections of 3.5-9.0 x 10(7) cells/vaccine. Vaccines were well tolerated with only minor systemic symptoms reported. Injection with tumor cells induced a local inflammatory response consisting predominantly of CD8+ and/or TIA-1+ T-lymphocytes. Myeloma specific anti-tumor responses, assessed by interferon-gamma (IFN-gamma) release and cytotoxic T cell killing of autologous tumor cells, were not enhanced after vaccination in one evaluable patient. Clinical response, manifested as a decrease in serum paraprotein, was not observed in the one patient who had measurable disease at the time of vaccination. These results demonstrate that the generation of adenovector modified plasma cell vaccines is technically feasible and can be safely administered post-transplant. Further studies of immunlogic and clinical efficacy are required.


Assuntos
Terapia Genética , Interleucina-2/genética , Mieloma Múltiplo/terapia , Plasmócitos/imunologia , Vacinação , Adenoviridae/genética , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
10.
Gene Ther ; 8(4): 282-90, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313802

RESUMO

It has been shown that intratumor administration of an adenovirus vector expressing IL-12 produces a potent T cell-mediated response that leads to significant tumor regression in a murine breast cancer model. IP-10 and MIG are CXC chemokines that recruit mononuclear cells in vivo. In addition to their chemotactic roles, IP-10 and MIG inhibit angiogenesis. We tested whether the addition of IP-10 or MIG may both enhance the antitumor immune response of IL-12 through T cell recruitment and inhibit tumor growth through angiostasis. Adenovirus vectors expressing IP-10 or MIG and/or IL-12 were administered intratumorally in a murine model of mammary adenocarcinoma and fibrosarcoma. Administration of IP-10 or MIG in combination with IL-12 resulted in considerable tumor regression and increased survival time of tumor-bearing animals as compared with IP-10, MIG, IL-12 alone or control-treated animals, with the IP-10 IL-12 combination being most effective. These results suggest augmenting the antitumor immune response and inhibiting tumor angiogenesis with adenoviral vectors expressing IP-10 in combination with IL-12 is a novel way to enhance tumor regression.


Assuntos
Adenoviridae/genética , Quimiocinas CXC/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Interleucina-12/genética , Neoplasias Mamárias Experimentais/terapia , Animais , Northern Blotting , Complexo CD3/análise , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Colágeno , Testes Imunológicos de Citotoxicidade , Combinação de Medicamentos , Endotélio Vascular/patologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Injeções Intralesionais , Interferon gama/análise , Interleucina-4/análise , Laminina , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Nus , Neovascularização Patológica , Proteoglicanas , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Gene Ther ; 8(4): 316-23, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313806

RESUMO

Overexpression of ErbB-2/neu occurs in 20-30% of patients with breast cancer and indicates a poor prognosis. The presence of a detectable immune response to ErbB-2/neu in some patients suggests that this oncogene may be a useful target for vaccine therapy. We evaluated whether genetic immunization using dendritic cells (DC) transduced ex vivo with an adenovirus expressing the ErbB-2/neu gene (AdNeuTK) could induce protective and therapeutic immunity against a breast tumor cell line overexpressing ErbB-2/neu. Subcutaneous (s.c.) immunization with the DC vaccine elicited protective immunity in an average of 60% of animals. CTL analysis demonstrated specific cytotoxic activity against breast tumor cells, as well as syngeneic fibroblasts transduced with AdNeuTK. In vivo depletion studies demonstrated both CD4+ and CD8+ T cells were required. In a therapeutic setting, immunization with the DC vaccines could cure mice with pre-established tumors and efficacy was further enhanced by cotransducing DCs with a vector expressing murine IL-12 (AdmIL-12). These studies support DC vaccines as a therapeutic strategy for human breast cancer, while emphasizing the importance of optimizing an immune response by combining tumor antigen presentation with immunostimulatory cytokines.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/transplante , Terapia Genética/métodos , Interleucina-12/genética , Receptor ErbB-2/genética , Adenoviridae/genética , Animais , Neoplasias da Mama/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Testes Imunológicos de Citotoxicidade , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Vetores Genéticos/administração & dosagem , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos , Neoplasias Experimentais/terapia , Ratos , Receptor ErbB-2/metabolismo , Transdução Genética/métodos
13.
Curr Pharm Des ; 6(6): 613-32, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10788600

RESUMO

The understanding of cytokine networks and the exploitation of these networks for the treatment of immune and inflammatory diseases as well as cancer depend on in vivo delivery of cytokines. Due to instability of recombinant cytokine proteins, investigators have employed cytokine-encoding gene therapy vectors to induce high levels of cytokine expression in vivo. Numerous gene therapy vectors have been developed recently which are suitable for this purpose. Recent advances in the design of adenovirus, adeno-associated virus, poxvirus, retrovirus, lentivirus, and nonviral vectors are described here. Properties of the various vector systems which determine their usefulness for cytokine gene delivery are compared. The implementations of cytokine-encoding gene therapy vectors for analyzing immune responses and for the therapy of inflammatory disorders, immune disease, infections and cancer are reviewed.


Assuntos
Citocinas/biossíntese , Vetores Genéticos/genética , Adenoviridae/genética , Animais , Citocinas/genética , Humanos , Retroviridae/genética
14.
Cancer Gene Ther ; 6(5): 465-74, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10505857

RESUMO

The toxic effects of tumor necrosis factor alpha (TNFalpha) have greatly limited its use in tumor therapy. Recently, clear evidence has been obtained linking the p55 TNF receptor (TNFR) to the induction of systemic toxicity. We have generated a p75 murine TNFR (mTNFR)-specific mutant of mTNFalpha (D142N-A144R), cloned this gene into a recombinant adenovirus vector (Ad-75), and studied its efficacy for tumor immunotherapy of a murine transgenic breast cancer model. Cell culture supernatants from Ad-75-transduced cells showed no cytotoxic activity on L929 cells, but retained the ability to induce proliferation of a murine T-cell line (CT6); this activity was not blocked by soluble p55 mTNFR. Furthermore, it was shown that the mutant form of mTNFalpha was able to coimmunoprecipitate only with the p75 mTNFR and not with the p55 mTNFR. Tumors injected with Ad-75 became necrotic, and mice injected with < or =1 x 10(9) plaque-forming units showed no mortality, whereas both wild-type murine and human TNF vectors induced lethality at doses of 1 and 5 x 10(8) plaque-forming units. All Ad-TNF vectors induced partial or permanent tumor regressions, with cured mice showing immune memory against the tumor. These results demonstrate that a p75 mTNFR agonist expressed from a recombinant adenovirus vector does not induce mortality at doses that cause tumor regression.


Assuntos
Adenoviridae/genética , Antígenos CD/genética , Vetores Genéticos , Neoplasias Mamárias Experimentais/imunologia , Receptores do Fator de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Animais , Sequência de Bases , Divisão Celular , Primers do DNA , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Mutação , Receptores Tipo II do Fator de Necrose Tumoral
15.
Am J Respir Cell Mol Biol ; 21(3): 317-26, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10460749

RESUMO

Expression of granulocyte macrophage colony-stimulating factor (GM-CSF) in the airway allows allergic sensitization to ovalbumin (OVA) in an experimental protocol that others have shown to induce inhalation tolerance. The ensuing response is characterized by T helper (Th)2 cytokines, marked eosinophilia in the bronchoalveolar lavage fluid (BALF) and the tissue, and goblet-cell hyperplasia. These findings, which underscore the importance of the airway microenvironment in the development of immune responses to airborne antigens, prompted us to investigate whether a Type 1 polarized cytokine milieu in the airway would modulate the allergic sensitization. To this end, we concurrently expressed GM-CSF and interleukin (IL)-12 in the airway, using an adenovirus-mediated gene transfer approach. Coexpression of IL-12 did not prevent the development of an antigen-specific immune inflammatory response, but altered its phenotype. Whereas a similar total cell number was observed in the BALF, airway eosinophilia was abrogated. Histologic evaluation of the tissue corroborated the findings in the BALF and demonstrated that IL-12 coexpression prevented goblet-cell hyperplasia. Expression of IL-12 decreased IL-4 and IL-5 content in the BALF by about 80 and 95%, respectively, and IL-5 in the serum by approximately 80%. In contrast, interferon (IFN)-gamma was increased in both BALF and serum. Similarly, we observed a Th2/Th1 shift in OVA-specific cytokine production in vitro. Recall challenge with OVA in vivo after resolution of the initial inflammatory response demonstrated that the effect of IL-12 was persistent. IL-12-mediated inhibition of airway eosinophilia was mainly IFN-gamma-independent, whereas inhibition of OVA-specific IgE synthesis was IFN-gamma-dependent. Our data underscore the importance of the airway microenvironment in the elicitation of immune responses to environmental antigens.


Assuntos
Brônquios/imunologia , Técnicas de Transferência de Genes , Interleucina-12/genética , Adenoviridae/genética , Administração por Inalação , Aerossóis , Animais , Brônquios/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Células Cultivadas , Citocinas/imunologia , Citocinas/farmacologia , Relação Dose-Resposta a Droga , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunoglobulina E/biossíntese , Imunoglobulina E/imunologia , Memória Imunológica/imunologia , Interleucina-12/imunologia , Interleucina-12/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa/imunologia , Mucosa/metabolismo , Ovalbumina/administração & dosagem , Ovalbumina/farmacologia
16.
Hum Gene Ther ; 10(5): 697-709, 1999 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10210138

RESUMO

We have previously demonstrated that intratumoral injection with Ad vectors expressing IL-2 or IL-12 can induce regression in a murine breast cancer model. These IL-2- or IL-12-induced antitumor responses were mainly mediated by Ag-specific T cells. Lymphotactin is a novel lymphocyte chemokine that can cause local accumulation of CD4+, CD8+, and NK cells. We hypothesized that addition of lymphotactin may enhance the antitumor immune responses induced by locally produced IL-2 and IL-12 as we have previously shown. To this end we constructed two double-recombinant adenoviral vectors expressing lymphotactin along with either IL-2 (Ad5 Lym/IL-2) or IL-12 (Ad5 Lym/IL-12). Subcutaneous injection of polyoma middle T (PyMT) or Neu (8142) transgenically derived breast adenocarcinoma cells, in the hind flank of FVB/n mice, results in the formation of tumor nodules in 14-21 days. We show that these constructs elicit potent antitumor responses when administered intratumorally. The antitumor responses are long lasting as determined by rechallenge experiments and hence demonstrate a protective immunity. These observations indicate that by augmenting the antitumor response with adenoviral vectors expressing lymphotactin in combination with IL-2 or IL-12 is a novel way to enhance immunotherapeutic approaches.


Assuntos
Adenoviridae/genética , Quimiocinas C , Terapia Genética , Interleucina-12/genética , Interleucina-2/genética , Linfocinas/genética , Neoplasias Mamárias Experimentais/terapia , Sialoglicoproteínas/genética , Adenoviridae/metabolismo , Animais , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Testes Imunológicos de Citotoxicidade , Quimioterapia Combinada , Vetores Genéticos , Imuno-Histoquímica , Interferon gama/metabolismo , Interleucina-12/uso terapêutico , Interleucina-2/uso terapêutico , Interleucina-4/metabolismo , Linfocinas/uso terapêutico , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Sialoglicoproteínas/uso terapêutico , Fatores de Tempo
17.
J Immunol ; 162(2): 753-60, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9916695

RESUMO

Experimental infection of the susceptible BALB/c (H-2d) mouse with the intracellular parasite Leishmania major induces a predominant Th2-type T cell response that eventually leads to death. In contrast, the resistant B10.D2 (H-2d) strain develops Th1 cells that control parasite replication and disease. In this study, we tested the ability of a recombinant adenovirus vector-expressing IL-12 to skew the immune response in a Th1 direction and prevent leishmaniasis in susceptible mice. We report that BALB/c mice treated with the Ad5IL-12 vector on the same day as parasitic challenge are significantly protected against leishmaniasis and acquired long-lasting immunity, because upon rechallenge with L. major parasites they were resistant to disease. The vector-derived IL-12 expression was transient and highly localized to the tissue after i.m. injection; it caused an increase in the number of Ag-specific IFN-gamma-secreting lymphocytes and enhanced NK cell activity in the draining popliteal node. In contrast, resistant B10.D2 mice given i.m. injections with a recombinant adenovirus-expressing IL-4 displayed greater susceptibility to disease, and severe lesions were produced in some of the infected animals. These results suggest the potential use of recombinant adenoviruses expressing cytokines as potent immunomodulatory agents for the generation of protective immune responses against intracellular pathogens.


Assuntos
Adenoviridae/imunologia , Vetores Genéticos/imunologia , Interleucina-12/imunologia , Interleucina-4/imunologia , Leishmania major/imunologia , Leishmaniose Cutânea/prevenção & controle , Vacinas Sintéticas/imunologia , Adenoviridae/genética , Animais , Suscetibilidade a Doenças , Extremidades , Vetores Genéticos/administração & dosagem , Interações Hospedeiro-Parasita , Injeções Intramusculares , Interleucina-12/administração & dosagem , Interleucina-12/biossíntese , Interleucina-12/genética , Interleucina-4/administração & dosagem , Interleucina-4/biossíntese , Interleucina-4/genética , Leishmania major/crescimento & desenvolvimento , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/parasitologia , Antígenos Comuns de Leucócito/biossíntese , Antígenos Comuns de Leucócito/genética , Linfonodos/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Esplenopatias/parasitologia , Esplenopatias/prevenção & controle
18.
J Immunol ; 160(9): 4615-9, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9574569

RESUMO

Murine collagen-induced arthritis (CIA) is characterized by pannus formation, cell infiltration, and cartilage erosion, and shares histologic and immunologic features with rheumatoid arthritis. Numerous cytokines are reportedly associated with RA and/or CIA; however, their mechanistic role is not clear. To determine the role of IL-12 in CIA, DBA/1 LacJ mice were administered 3 x 10(8) plaque-forming units of mIL-12 i.p. in a nonreplicating adenoviral vector (AdIL-12) on day 25 following primary type II collagen immunization. Our studies demonstrated that systemic transient overexpression of IL-12 accelerated disease progression and augmented the arthritis severity relative to mice expressing a replication-deficient, E1-deleted Ad5 construct. A likely mechanism for this increase in pathology was the increase in the expression of cytokines and chemokines known to play a proinflammatory role in disease. In particular, levels of murine IFN-gamma were significantly increased in mice overexpressing AdIL-12 relative to the replication-deficient, E1-deleted Ad5 construct. Interestingly, the C-X-C chemokine murine macrophage inflammatory protein-2, as well as the C-C chemokines murine monocyte chemoattractant protein-1 and murine macrophage inflammatory protein-1alpha were up-regulated by AdIL-12 relative to controls. In an additional set of studies, neutralization of endogenous IL-12 in CIA mice was shown to delay disease onset and attenuate disease severity. IFN-gamma levels in the mice receiving anti-IL-12 were significantly decreased in joint homogenates. These studies demonstrate that IL-12 is an important cytokine involved in controlling the production of chemokines/cytokines leading to the evolution of experimental arthritis.


Assuntos
Artrite/genética , Artrite/imunologia , Quimiocinas/imunologia , Técnicas de Transferência de Genes , Terapia Genética , Interleucina-12/genética , Adenoviridae , Animais , Artrite/induzido quimicamente , Artrite/terapia , Quimiocinas/biossíntese , Colágeno , Vetores Genéticos , Camundongos , Camundongos Endogâmicos DBA
19.
Int J Oncol ; 12(3): 509-15, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9472086

RESUMO

Induction of systemic toxicity is a major factor limiting the use of TNFa in tumour therapy. The local expression of TNFa from Ad vector infected tumour cells, might result in reduced systemic toxicity and induce an enhanced local antitumour response. Two adenoviral vectors expressing human TNFa were constructed for use in tumour immunotherapy, one utilizing the HCMV promoter (Ad-HCMV-TNF) and the second utilizing the MCMV promoter (Ad-MCMV-TNF). Both vectors induced the secretion of hTNFa from transduced cells in vitro, however the MCMV promoter directed stronger expression in murine cells. Expression from the two vectors was also kinetically different with the MCMV promoter inducing earlier expression, from murine tumour cells in vitro and in vivo. Both vectors induced intratumoural necrosis, however only the Ad-MCMV-TNF vector induced systemic toxicity and significant antitumour activity when directly injected into tumours, killing 8 of 20 mice and inducing partial (2 of 12) and permanent (1 of 12) tumour regressions at a dose of 5x108 pfu/mouse. These data indicate that hTNFa expressed from an Ad vector is considerably toxic to mice while inducing a moderate antitumour response.


Assuntos
Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/terapia , Fator de Necrose Tumoral alfa/biossíntese , Adenoviridae , Animais , Antígenos Transformantes de Poliomavirus/biossíntese , Bioensaio , Linhagem Celular , Sobrevivência Celular , Citomegalovirus , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Imunoterapia , Rim , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Transgênicos , Necrose , Regiões Promotoras Genéticas , Proteínas Recombinantes/biossíntese , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/uso terapêutico
20.
Hum Gene Ther ; 9(5): 707-18, 1998 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-9551618

RESUMO

Although cytokine gene transfer for cancer treatment can stimulate immune recognition and tumor regression in animal models, there is still a need for improvements to these strategies. In this study, we examined the efficacy of a combination gene therapy using adenovirus (Ad) 5 vectors expressing human interleukin-2 and the wild-type (wt) human p53 gene under control of the human cytomegalovirus immediate early promoter (AdIL-2 and Adp53wt, respectively). Infected murine cell lines and primary mouse tumor cells secreted high levels of IL-2 and over expressed the p53 protein for at least 9 days. After infection of cells with Adp53wt, DNA synthesis was significantly inhibited and apoptosis was induced within 3-5 days. Both vectors were tested in a transgenic mouse mammary adenocarcinoma model for antitumor response. Following a single intratumoral injection of mice bearing PyMT induced tumors, the combination of Adp53wt (1 x 10(9) pfu) plus a relatively low dose of AdIL-2 (1.5 x 10(8) pfu) caused regressions in 65% of the treated tumors without toxicity. Fifty percent of the treated mice remained tumor free and were immune to rechallenge with fresh tumor cells. In contrast, injection of either vector alone at this does resulted in only a delay in tumor growth. Only mice co-injected with Adp53wt and AdIL-2 showed specific antitumor cytolytic T lymphocyte (CTL) activity, indicating that the immune response involved in tumor regression was promoted by the combination therapy. These results suggest that cancer treatment strategies involving combined delivery of immunomodulatory and antiproliferative genes may be highly effective.


Assuntos
Adenoviridae/genética , Genes p53 , Terapia Genética , Vetores Genéticos , Interleucina-2/genética , Neoplasias Mamárias Experimentais/terapia , Animais , Apoptose , Western Blotting , Terapia Combinada , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/imunologia , Humanos , Interleucina-2/uso terapêutico , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Transgênicos , Indução de Remissão , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...