Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gene Ther ; 18(10): 986-95, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21490686

RESUMO

Recent investigations have demonstrated that adenoviral and lentiviral vectors encoding HER-2 can be utilized in cancer immunotherapy. However, it is not known whether both viral systems elicit a similar immune response. Here, we compare the immune response in mice induced by dendritic cells (DCs) infected with either recombinant adenovirus or lentivirus encoding rat HER-2 (rHER-2). Both vaccine types yielded similar control of tumor growth, but we found clear differences in their immune responses 10 days after DC immunization. Adenovirus rHER-2-transduced DCs elicited locally and systemically high frequencies of CD4+ and CD8+ T cells, while lentivirus rHER-2-transduced DCs predominantly led to CD4+ T-cell infiltration at the tumor site. Splenocytes from mice immunized with lentivirus rHER-2-transduced DCs secreted higher levels of interferon (IFN)-γ, mainly by CD4+ T cells, following stimulation by RM-1-mHER-2 tumors. In contrast, the adenovirus vaccinated group exhibited CD4+ and CD8+ T cells that both contributed to IFN-γ production. Besides an established cellular immune response, the rHER-2/DC vaccine elicited a significant humoral response that was highest in the adenovirus group. DC subsets and regulatory T cells in the spleen were also differentially modulated in the two vaccine systems. Finally, adoptive transfer of splenocytes from both groups of immunized mice strongly inhibited in vivo tumor growth. Our results suggest that not only the target antigen but also the virus system may determine the nature and magnitude of antitumor immunity by DC vaccination.


Assuntos
Adenoviridae , Células Dendríticas/transplante , Genes erbB-2/genética , Vetores Genéticos/administração & dosagem , Imunoterapia/métodos , Lentivirus , Neoplasias/terapia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/metabolismo , Camundongos , Neoplasias/imunologia , Ratos
2.
Curr Opin Biotechnol ; 6(5): 590-5, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7579670

RESUMO

Adenoviral vectors have proven to be excellent vehicles for gene delivery in vivo to a wide range of cell types. These vectors have been used to transfer genes such as CFTR to correct the defect in cystic fibrosis and, more recently, to supply serum blood factors and genetically modify tumors to enhance therapy.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Humanos
3.
Hum Gene Ther ; 7(16): 1995-2002, 1996 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-8930660

RESUMO

Mice bearing breast tumors were treated with a single dose of an adenovirus expressing interleukin-12 (AdmIL-12.1) injected intratumorally, which produced regressions in greater than 75% of the treated tumors; approximately one-third of the animals remained tumor free. Complete regression was associated with immunity to secondary challenge with fresh tumor cells. Analysis of local cytokine expression demonstrated maximum expression of IL-12 within the tumor between 24 and 72 hr post-injection, reaching 600-800 ng per tumor, with elevated local levels of IL-12 detectable for at least 9 days. This expression was highly localized as serum IL-12 peaked at 40-60 ng/ml at 24 hr and was less than 10 ng/ml from day 3 onward. Interferon-gamma (IFN-gamma) concentrations were markedly increased within the tumor following AdmIL-12.1 administration, demonstrating that IL-12 was acting locally. Tumor-draining lymph node cells spontaneously produced IFN-gamma following AdmIL-12.1 treatment, suggesting these cells were activated by IL-12. These data demonstrate that AdmIL-12.1 can be used to deliver very high levels of localized cytokine production. Moreover, we have confirmed that the IL-12 produced from our vector actually affects the local cytokine environment of the tumor and activates responder cells present within the tumor.


Assuntos
Adenovírus Humanos/genética , Terapia Genética , Vetores Genéticos , Interleucina-12/genética , Interleucina-12/uso terapêutico , Neoplasias Mamárias Animais/terapia , Animais , Feminino , Expressão Gênica , Interferon gama/biossíntese , Interleucina-12/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos
4.
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
5.
Cancer Gene Ther ; 7(3): 353-9, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10766341

RESUMO

A model of lipoplex-induced peritonitis was used to characterize the inflammatory response to cationic lipid:DNA lipoplexes with respect to activation of host antitumoral effector mechanisms. Three different cationic lipids were used in these studies: N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N-(1-[2,3-dioleoyloxylpropyl)-N,N,N-trimethylammonium chloride (DOTAP), and N-(1-[2,3-dioleyloxy]propyl)-N,N,N-trimethylammonium chloride (DOTMA). The DODAC and DOTMA lipoplexes exhibited similar transfection properties in vitro, whereas the DOTAP lipoplexes transfected quite poorly in all cell lines tested. Intraperitoneal injection of cationic lipoplexes into immunocompetent mice resulted in a profound infiltration of inflammatory cells, secretion of interferon-gamma, and increased natural killer activity within the peritoneal cavity. Both DODAC and DOTMA lipoplexes produced similar inflammatory responses, lasting at least 5 days. The inflammation induced by DOTAP lipoplexes peaked by day 3 and resolved to near-control levels by day 5. These data indicate that although cationic lipid DNA complexes may differ in their inflammatory properties, the natural killer activation and interferon-gamma secretion that follow lipoplex administration should provide a functional adjuvant for cancer gene therapies that benefit from immunostimulation.


Assuntos
Antineoplásicos/farmacologia , DNA/farmacologia , Lipídeos/farmacologia , Animais , Antineoplásicos/imunologia , Líquido Ascítico/imunologia , Líquido Ascítico/metabolismo , Cátions , Citocinas/metabolismo , DNA/genética , Ácidos Graxos Monoinsaturados/farmacologia , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Lipídeos/genética , Lipossomos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Lavagem Peritoneal , Peritonite/induzido quimicamente , Peritonite/imunologia , Compostos de Amônio Quaternário/farmacologia , Transfecção , Células Tumorais Cultivadas
6.
Cancer Chemother Pharmacol ; 31(2): 146-50, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1280536

RESUMO

We have previously demonstrated that uptake of the amino acid amide sarcosinamide by the glioma cell line SK-MG-1 occurs via the catecholamine carrier that accommodates epinephrine (Km = 0.284 mM; Vmax = 0.154 nmol/10(6) cells/min). Sarcosinamide chloroethylnitrosourea (SarCNU), a new anticancer agent that exerts increased in vitro antitumor activity against gliomas as compared with BCNU (bis-chloroethylnitrosourea), the standard agent of choice, competitively inhibits sarcosinamide uptake by SK-MG-1 cells [inhibition constant (Ki) = 3.26 mM]. Using radiolabeled N-[3H]-sarcosinamide, we determined the transport of sarcosinamide in HT-29 colon-cancer cells, in Calu-1 lung-cancer cells, and in normal foreskin DHF fibroblasts. Sarcosinamide transport was linear for up to 1 min at 22 degrees C. In HT-29 cells and DHF fibroblasts, the uptake of sarcosinamide followed Michaelis-Menten kinetics of carrier-mediated transport. In HT-29 cells the Michaelis constant (Km) was 2.76 +/- 0.1 mM and the maximal velocity (Vmax) was 2.03 +/- 0.1 nmol/10(6) cells/min, whereas in DHF fibroblasts the respective values were 6.58 +/- 3.90 mM and 12.08 +/- 8.20 nmol/10(6) cells/min. In these two cell lines, neither epinephrine nor leucine significantly reduced sarcosinamide transport. In Calu-1 cells there was no evidence of carrier-mediated transport of either sarcosinamide or epinephrine. These nonglial cell lines lack a high-affinity catecholamine carrier. The increased cytotoxicity of SarCNU in gliomas may correlate with the presence of a high-affinity catecholamine carrier.


Assuntos
Epinefrina/farmacocinética , Sarcosina/análogos & derivados , Transporte Biológico , Neoplasias do Colo/metabolismo , Portadores de Fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Sarcosina/farmacocinética , Células Tumorais Cultivadas
7.
Mucosal Immunol ; 7(6): 1395-404, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24781052

RESUMO

Intestinal T helper type 2 (Th2) immunity in food allergy results in IgG1 and IgE production, and antigen re-exposure elicits responses such as anaphylaxis and eosinophilic inflammation. Although interleukin-4 (IL-4) is critically required for allergic sensitization, the source and control of IL-4 during the initiation of Th2 immunity in vivo remains unclear. Non-intestinal and non-food allergy systems have suggested that natural killer-like T (NKT) or γδ T-cell innate lymphocytes can supply the IL-4 required to induce Th2 polarization. Group 2 innate lymphoid cells (ILCs) are a novel IL-4-competent population, but their contribution to initiating adaptive Th2 immunity is unclear. There are also reports of IL-4-independent Th2 responses. Here, we show that IL-4-dependent peanut allergic Th2 responses are completely intact in NKT-deficient, γδ T-deficient or ILC-deficient mice, including antigen-specific IgG1/IgE production, anaphylaxis, and cytokine production. Instead, IL-4 solely from CD4(+) Th cells induces full Th2 immunity. Further, CD4(+) Th cell production of IL-4 in vivo is dependent on OX40L, a costimulatory molecule on dendritic cells (DCs) required for intestinal allergic priming. However, both Th2 cells and ILCs orchestrated IL-13-dependent eosinophilic inflammation. Thus, intestinal Th2 priming is initiated by an autocrine/paracrine acting CD4(+) Th cell-intrinsic IL-4 program that is controlled by DC OX40L, and not by NKT, γδ T, or ILC cells.


Assuntos
Alérgenos/imunologia , Arachis/química , Interleucina-4/imunologia , Intestinos/imunologia , Glicoproteínas de Membrana/imunologia , Hipersensibilidade a Amendoim/imunologia , Células Th2/imunologia , Fatores de Necrose Tumoral/imunologia , Alérgenos/química , Animais , Eosinófilos/imunologia , Eosinófilos/patologia , Imunidade Inata , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-4/genética , Intestinos/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligante OX40 , Hipersensibilidade a Amendoim/patologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Células Th2/patologia , Fatores de Necrose Tumoral/genética
8.
Cancer Gene Ther ; 16(4): 338-50, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18989352

RESUMO

We have investigated the therapeutic potential of a prototypic melanoma vaccine based on recombinant adenovirus expressing human dopachrome tautomerase in the B16F10 murine melanoma model. We found that in the presence of a tumor, the magnitude of T-cell immunity evoked by the vaccine was significantly reduced. This impairment was compounded by defects in cytokine production and degranulation within the tumor-infiltrating lymphocytes (TILs). We showed that the combination of vaccination with high-dose cyclophosphamide was able to skew the response toward the target antigen and enhanced both the quantity and quality of antigen-specific CD8+ and CD4+ T-cell responses in tumor-bearing mice, which resulted in the inhibition of tumor growth. Furthermore, when tumor-specific antigens were targeted by the vaccine, the combination therapy could actually produce tumor regression, which appeared to result from the high frequency of antigen-specific T cells. These data show that recombinant adenovirus vaccines are compatible with conventional high-dose chemotherapy and that the combined treatment results in improved therapeutic outcomes relative to either agent individually.


Assuntos
Adenoviridae/genética , Vacinas Anticâncer/uso terapêutico , Ciclofosfamida/administração & dosagem , Melanoma Experimental/terapia , Vacinas de DNA/uso terapêutico , Animais , Antineoplásicos Alquilantes/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Vetores Genéticos , Humanos , Imunidade Celular/efeitos dos fármacos , Oxirredutases Intramoleculares/biossíntese , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Melanoma Experimental/imunologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Transplante de Neoplasias , Oxirredutases/biossíntese , Oxirredutases/genética , Oxirredutases/imunologia , Resultado do Tratamento
9.
Antisense Nucleic Acid Drug Dev ; 10(3): 217-24, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10905558

RESUMO

Stabilized antisense lipid particles (SALP) have been developed for the systemic delivery of oligonucleotides. The impact of intravenous SALP administration was measured with respect to activation of natural killer (NK) and NK1.1+ T (NKT) cells in the livers of immunocompetent mice. Treatment with a SALP containing a highly mitogenic oligonucleotide (INX-6295) generated an increase in NK cytolytic activity and cell number within the liver but did not appear to affect the number of hepatic NKT cells or their cytolytic activity. The same results were observed after intravenous administration of the mitogenic oligonucleotide alone. Interestingly, treatment with a SALP containing a weakly mitogenic oligonucleotide (INX-6300) also activated the liver NK cells, whereas the oligonucleotide alone was unable to elicit these effects. The NK stimulatory activity of a SALP containing INX-6300 required both lipid and oligonucleotide components. These results demonstrate that in addition to modifying the pharmacokinetics and biodistribution of intravenously administered oligonucleotides, SALP possess immunostimulatory activity independent of oligonucleotide mitogenicity, which can serve as an adjuvant to antisense therapies for cancer.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Células Matadoras Naturais/efeitos dos fármacos , Lipídeos/administração & dosagem , Fígado/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Mitógenos/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Linhagem Celular , Ilhas de CpG/efeitos dos fármacos , Ilhas de CpG/imunologia , Portadores de Fármacos , Feminino , Injeções Intravenosas , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Fígado/citologia , Fígado/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes
10.
J Immunol ; 160(5): 2531-8, 1998 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9498799

RESUMO

Tumors that express tumor-specific antigens can maintain growth in an immunocompetent organism. Current hypotheses tend toward T cell anergy as a key component for the inhibition of immunoreactivity against such tumors. Anergy is thought to occur from hyperactive stimulation of the TCR in the absence of costimulation (costimulation leads to proliferation via IL-2 production). Subcutaneous injection of transgenic polyoma middle T transformed breast adenocarcinoma tumor cells (PyMT) in the hind flank of FVB/n mice results in the formation of tumor nodules at this site. We determined the MHC class I and class II, B7-1, and B7-2 expression in the tumor cells by flow cytometry and showed positive staining for only MHC class I. We show that a single E1-deleted adenovirus constructed to express both the costimulatory molecule B7-1 (murine) and human IL-2 genes (Ad5E1 mB7-1/human IL-2) elicits a very potent antitumor response when administered intratumorally. Ad5E1 mB7-1/human IL-2 induced rapid and complete regression (100%) of all tumors compared with Ad5 E1 mB7-1 (38%), Ad CAIL-2 (42%), and Ad5E1 dl70-3 (control vector) (0%). All mice that exhibited complete tumor regression were fully protected in tumor cell challenge experiments. The systemic immunity generated by intratumoral administration of the Ad vectors was associated with a strong anti-PyMT CTL response. These observations indicate that augmenting the immunogenicity of the tumor with coincident expression of B7-1 in combination with IL-2 may prove beneficial in direct tumor immunotherapy.


Assuntos
Adenocarcinoma/terapia , Adenoviridae/genética , Antígeno B7-1/genética , Vetores Genéticos/imunologia , Interleucina-2/genética , Neoplasias Mamárias Experimentais/terapia , Proteínas Recombinantes de Fusão/imunologia , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenoviridae/imunologia , Animais , Antígenos Transformantes de Poliomavirus/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-1/uso terapêutico , Linhagem Celular , Citotoxicidade Imunológica , Vetores Genéticos/síntese química , Vetores Genéticos/uso terapêutico , Humanos , Injeções Intralesionais , Interleucina-2/biossíntese , Interleucina-2/uso terapêutico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Evasão Tumoral/genética
11.
J Virol ; 73(10): 8027-34, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10482551

RESUMO

We have analyzed transgene (lacZ) expression from a first-generation adenovirus (Ad) vector in comparison to helper-dependent (hd) Ads deleted for various portions of the viral coding sequences and generated by using the Cre/loxP helper-dependent system (R. J. Parks et al., Proc. Natl. Acad. Sci. USA 93:13565-13570, 1996). An hd vector deleted for approximately 70% of the Ad genome (AdRP1001) provided levels and durations of transgene expression similar to those of a control first generation Ad vector containing an identical expression cassette. Deletion of all Ad sequences from the hdAd and replacement with a approximately 22-kb fragment of lambda DNA resulted in a decrease in the level and duration of lacZ expression which could not be reversed by the inclusion of a matrix attachment region. However, substitution of the lambda stuffer in the fully deleted hdAd with sequences from the human hypoxanthine-guanine phosphoribosyltransferase gene resulted in significantly improved transgene expression. In vitro assays for cytotoxic T lymphocytes (CTL) directed against putative peptides encoded by the vector backbone showed that, although CTL were generated against the vector containing the lambda DNA, no such CTL were generated against the vector containing the hypoxanthine-guanine phosphoribosyltransferase (HPRT) sequences. Surprisingly, the rate of loss of the HPRT- and lambda-containing vectors from mouse liver was similar, despite the differences in expression kinetics, indicating that the lambda stuffer-directed CTL were inefficient at eliminating the transduced cells. Thus, the nature of the DNA backbone of hdAds can have important effects on the functioning of the vector. Since most fully deleted vectors require "stuffer" DNA as part of the vector backbone to maintain optimum vector size, these observations must be taken into account in the design of hdAd vectors.


Assuntos
Adenoviridae , DNA/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Linfócitos T Citotóxicos/virologia , Animais , Linhagem Celular , Citotoxicidade Imunológica , Expressão Gênica/imunologia , Genes Reporter , Humanos , Óperon Lac , Camundongos , Linfócitos T Citotóxicos/imunologia
12.
Gene Ther ; 4(10): 1069-76, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9415313

RESUMO

Adenovirus (Ad) vectors are being intensively studied as vehicles for cancer gene therapy. We have been exploring the benefits of direct intratumoral injection of Ads expressing cytokines for immunotherapy. Our previous work demonstrated that therapy using a vector expressing interleukin-12 (AdmIL-12.1) produced regressions in approximately 80% of treated tumors supporting further preclinical investigations. Recent reports have shown that immunity to Ad can be a major limiting factor in Ad-mediated gene transfer. As most animal studies with Ad vectors have involved nonimmune hosts, it remains difficult to predict how effective these treatments will be in humans, where the majority of individuals have had previous exposure to Ad. To address this question, we compared the effectiveness of the AdmIL-12.1 cancer therapy in naive and Ad-immune mice. We found that both groups responded equally well to treatment and that the response to AdmIL-12.1 in both groups resulted in the generation of CTL reactive against tumor antigen indicating that antitumor immunity was achieved. Peak transgene expression in the tumor was only reduced by 2.4 fold in Ad-immune animals compared with nonimmune mice. It was also observed that in naive animals, the virus disseminated from the site of the tumor following injection and by 72 h substantial transgene expression was detected in peripheral organs, most notably the liver. Transgene expression in the liver of Ad-immune animals was reduced by greater than 1000-fold relative to that in naive mice. These results strongly support the clinical utility of Ad-based cancer gene therapy and suggest that Ad immunity may be advantageous in that it is not a complete block to gene transfer in the tumor and it greatly reduces virus dissemination.


Assuntos
Adenoviridae/imunologia , Vetores Genéticos/uso terapêutico , Interleucina-12/biossíntese , Neoplasias Mamárias Experimentais/terapia , Adenoviridae/genética , Adenoviridae/crescimento & desenvolvimento , Infecções por Adenoviridae/imunologia , Animais , Anticorpos Antivirais/sangue , Feminino , Imunidade Celular , Interleucina-12/genética , Interleucina-12/uso terapêutico , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos , Linfócitos T Citotóxicos/imunologia
13.
Gene Ther ; 5(10): 1400-9, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9930346

RESUMO

We have studied the ability of adenoviral (Ad) vectors expressing the cytokines IL-2 or IL-12 to mediate regression of established tumors in a mouse model of mammary adenocarcinoma. Previous results indicated that intratumoral injection of vectors expressing IL-2 (AdCAIL-2), or IL-12 (AdmIL-12.1) induced complete tumor regression in approximately 30-40% of treated animals. In the current studies, we investigated the mechanism of tumor killing in responding animals and the efficacy of AdIL-2 and AdIL-12 vector administration in combination compared with the use of either vector alone. Animals bearing subcutaneous mammary tumors were injected intratumorally with Ad vectors expressing IL-2 or IL-12 or were coinjected with both vectors. Animals receiving the combination treatment responded substantially better than animals which had received either vector alone, with 65% of animals treated with both vectors undergoing complete tumor regression. In all three treatment regimens, tumor regression was associated with the presence of specific antitumor antigen cytotoxic T-lymphocytes (CTLs), which secreted elevated levels of IFN-gamma. Consistent with circulating CTLs being involved in regression, when animals bearing bilateral tumors were inoculated in a single tumor with IL-2 or IL-12 expressing vectors, both tumors regressed in many cases. Again, treatment with both AdCAIL-2 and AdmIL-12.1 was most effective, with 63% of animals undergoing complete regression of both treated and untreated tumors, compared to 18 or 22% of animals injected with either AdCAIL-2 or AdmIL-12.1 alone. These data indicate that the combination of IL-2 and IL-12 is a more effective inducer of antitumor immune responses than either one alone, and that the resulting antitumor responses are effective in mediating the regression of distal untreated tumors, a property which may aid in the treatment of metastatic disease.


Assuntos
Adenocarcinoma/terapia , Terapia Genética/métodos , Imunoterapia/métodos , Interleucinas/genética , Neoplasias Mamárias Experimentais/terapia , Adenocarcinoma/imunologia , Adenoviridae , Animais , Feminino , Expressão Gênica , Vetores Genéticos , Injeções Intralesionais , Interleucina-12/genética , Interleucina-2/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Transgênicos , Linfócitos T Citotóxicos/imunologia
14.
J Immunol ; 167(9): 5027-33, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11673511

RESUMO

By using adoptive transfer of Ag-loaded bone marrow-derived dendritic cells (BMDC), we have established an in vivo model of CTL priming. Activation of CTL in these experiments required both CD4(+) T cells and CD154, demonstrating that this model reflects CD4(+) T cell-dependent dendritic cell (DC) licensing. Because IL-12 has been suggested to play an important role in CTL activation by DC, we examined the ability of BMDC to prime CTL in the complete absence of IL-12 using p40-deficient mice. We observed that the absence of IL-12 does not affect the phenotype or allostimulatory function of BMDC after in vitro maturation. Moreover, there was no difference in the ability of Ag-loaded DC to elicit CTL cytotoxicity, whether the Ag was delivered by virus infection or peptide pulsing. Equal frequencies of Ag-specific, IFN-gamma-secreting CD8(+) T cells developed in both wild-type and IL-12-deficient backgrounds. Finally, CTL generated in the IL-12-deficient environment were capable of protecting immunized mice against tumor challenge, demonstrating that these CTL were fully functional, despite the absence of IL-12 during the maturation process in vivo. These results indicate that IL-12 is not critical for the development of IFN-gamma secreting, CD8(+) T cells and that another mechanism must be used by licensed DC to prime and activate CTL.


Assuntos
Antígenos CD8/análise , Células Dendríticas/fisiologia , Interferon gama/biossíntese , Interleucina-12/fisiologia , Linfócitos T Citotóxicos/fisiologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Ligante de CD40/fisiologia , Feminino , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA