Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 813
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharm ; 16(4): 1714-1722, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30860846

RESUMO

Tumor associated macrophages (TAMs) are important components residing in the tumor microenvironment. They are immunosuppressive and promote tumor progression. Targeting TAMs and reprogramming their phenotype may be a promising strategy that can restore antitumor immune responses. In this study, we developed a microRNA delivery system based on lipid-coated calcium phosphonate nanoparticles (CaP/miR@pMNPs) containing conjugated mannose and sterically shielded with a pH-responsive material. The nanocarrier could respond to the low pH in the tumor microenvironment and expose mannose to promote cellular internalization in TAMs. The carrier could reactivate TAMs and reprogram their functions, reverse the immunosuppressive tumor microenvironment, and inhibit tumor growth in a tumor-bearing mouse model. In summary, redirecting the polarization of TAMs is a potential therapeutic strategy for tumor immunotherapy.


Assuntos
Imunoterapia , Macrófagos/imunologia , MicroRNAs/administração & dosagem , Nanopartículas/administração & dosagem , Sarcoma Experimental/terapia , Microambiente Tumoral/imunologia , Animais , Apoptose , Fosfatos de Cálcio/química , Proliferação de Células , Humanos , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , MicroRNAs/química , MicroRNAs/genética , Nanopartículas/química , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nanomedicine ; 14(2): 289-301, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28993266

RESUMO

In order to prevent the aggregation of ICG and enhance its stability, a novel nanoplatform (TiO2:Yb,Ho,F-ß-CD@ICG/HA) was designed for NIR-induced phototherapy along with multi-mode imaging(UCL/MRI/Flu). In this nanosysytem: TiO2:Yb,Ho,F was used as upconversion materials and applied in vivo for the first time; ß-CD acted as a "protective umbrella" to load separated ICG and avoid the low phototherapy efficiency because of its aggregation; HA was the capping agent of ß-CD to prevent ICG unexpected leaking and a target to recognize CD44 receptor. The nanosystem exhibited excellent size (~200 nm) and photo- and thermal-stability, preferable reactive oxygen yield and temperature response (50.4 °C) under 808 nm laser. It could efficiently target and suppress tumor growth. The imaging ability (UCL/MRI) of TiO2:Yb,Ho,F could facilitate diagnosis of the tumor, especially for deep tissues. Altogether, our work successfully improved the phototherapy efficacy through incorporating the ICG into the cavity of ß-CD and applied TiO2:Yb,Ho,F for upconversion imaging in vivo.


Assuntos
Verde de Indocianina/metabolismo , Neoplasias Mamárias Experimentais/terapia , Imagem Multimodal/métodos , Nanopartículas/administração & dosagem , Fototerapia , Sarcoma Experimental/terapia , Animais , Apoptose , Ciclo Celular , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Nanopartículas/química , Espécies Reativas de Oxigênio/metabolismo , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
3.
Int J Cancer ; 136(4): 965-76, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24978211

RESUMO

Isolated limb perfusion (ILP) is a treatment for advanced extremity sarcoma and in-transit melanoma. Advancing this procedure by investigating the addition of novel agents, such as cancer-selective oncolytic viruses, may improve both the therapeutic efficacy of ILP and the tumour-targeted delivery of oncolytic virotherapy. Standard in vitro assays were used to characterise single agent and combinatorial activities of melphalan, tumour necrosis factor-alpha (TNF-α) and Lister strain vaccinia virus (GLV-1h68) against BN175 rat sarcoma cells. An orthotopic model of advanced extremity sarcoma was used to evaluate survival of animals after ILP with combinations of TNF-α, melphalan and GLV-1h68. We investigated the efficiency of viral tumour delivery by ILP compared to intravenous therapy, the locoregional and systemic biodistribution of virus after ILP, and the effect of mode of administration on antibody response. The combination of melphalan and GLV-1h68 was synergistic in vitro. The addition of virus to standard ILP regimens was well tolerated and demonstrated superior tumour targeting compared to intravenous administration. Triple therapy (melphalan/TNF-α/GLV-1h68) resulted in increased tumour growth delay and enhanced survival compared to other treatment regimens. Live virus was recovered in large amounts from perfused regions, but in smaller amounts from systemic organs. The addition of oncolytic vaccinia virus to existing TNF-α/melphalan-based ILP strategies results in survival advantage in an immunocompetent rat model of advanced extremity sarcoma. Virus administered by ILP has superior tumour targeting compared to intravenous delivery. Further evaluation and clinical translation of this approach is warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Membro Posterior/patologia , Vírus Oncolíticos/fisiologia , Sarcoma Experimental/terapia , Vaccinia virus/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Quimioterapia do Câncer por Perfusão Regional , Terapia Combinada , Membro Posterior/efeitos dos fármacos , Humanos , Masculino , Melfalan/administração & dosagem , Transplante de Neoplasias , Ratos Endogâmicos , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/patologia , Fator de Necrose Tumoral alfa/administração & dosagem
4.
J Immunol ; 187(10): 4935-46, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22013118

RESUMO

Optimum efficacy of therapeutic cancer vaccines may require combinations that generate effective antitumor immune responses, as well as overcome immune evasion and tolerance mechanisms mediated by progressing tumor. Previous studies showed that IL-13Rα2, a unique tumor-associated Ag, is a promising target for cancer immunotherapy. A targeted cytotoxin composed of IL-13 and mutated Pseudomonas exotoxin induced specific killing of IL-13Rα2(+) tumor cells. When combined with IL-13Rα2 DNA cancer vaccine, surprisingly, it mediated synergistic antitumor effects on tumor growth and metastasis in established murine breast carcinoma and sarcoma tumor models. The mechanism of synergistic activity involved direct killing of tumor cells and cell-mediated immune responses, as well as elimination of myeloid-derived suppressor cells and, consequently, regulatory T cells. These novel results provide a strong rationale for combining immunotoxins with cancer vaccines for the treatment of patients with advanced cancer.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Exotoxinas/uso terapêutico , Marcação de Genes/métodos , Imunoterapia/métodos , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Neoplasias Mamárias Experimentais/terapia , Sarcoma Experimental/terapia , Vacinas de DNA/uso terapêutico , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Exotoxinas/genética , Exotoxinas/imunologia , Feminino , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Subunidade alfa2 de Receptor de Interleucina-13/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Vacinas de DNA/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
5.
J Immunol ; 185(5): 2895-902, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675595

RESUMO

T-bet (Tbx21), a T-box transcription factor, has been previously identified as a master regulator of type 1 T cell polarization. We have also recently shown that the genetic engineering of human dendritic cells (DCs) to express human T-bet cDNA yields type 1-polarizing APCs in vitro (1). In the present study, murine CD11c(+) DCs were transduced with a recombinant adenovirus encoding full-length murine T-bets (DC.mTbets) and analyzed for their immunomodulatory functions in vitro and in vivo. Within the range of markers analyzed, DC.mTbets exhibited a control DC phenotype and were indistinguishable from control DCs in their ability to promote allogenic T cell proliferation in MLR in vitro. However, DC.mTbets were superior to control DCs in promoting Th1 and Tc1 responses in vitro via a mechanism requiring DC-T cell interaction or the close proximity of these two cell types and that can only partially be explained by the action of DC-elaborated IL-12p70. When injected into day 7 s.c. CMS4 sarcoma lesions growing in syngenic BALB/c mice, DC.mTbets dramatically slowed tumor progression (versus control DCs) and extended overall survival via a mechanism dependent on both CD4(+) and CD8(+) T cells and, to a lesser extent, asialoGM1(+) NK cells. DC.mTbet-based therapy also promoted superior tumor-specific Tc1 responses in the spleens and tumor-draining lymph nodes of treated animals, and within the tumor microenvironment it inhibited the accumulation of CD11b(+)Gr1(+) myeloid-derived suppressor cells and normalized CD31(+) vascular structures. These findings support the potential translational utility of DC.Tbets as a therapeutic modality in the cancer setting.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Engenharia Genética/métodos , Imunidade Inata/genética , Injeções Intralesionais/métodos , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Proteínas com Domínio T/administração & dosagem , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica/imunologia , Antígenos H-2/administração & dosagem , Antígenos H-2/genética , Humanos , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sarcoma Experimental/mortalidade , Sarcoma Experimental/virologia , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Transdução Genética
6.
Nat Med ; 1(12): 1297-302, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7489412

RESUMO

Dendritic cells, the most potent 'professional' antigen-presenting cells, hold promise for improving the immunotherapy of cancer. In three different well-characterized tumour models, naive mice injected with bone marrow-derived dendritic cells prepulsed with tumour-associated peptides previously characterized as being recognized by class I major histocompatibility complex-restricted cytotoxic T lymphocytes, developed a specific T-lymphocyte response and were protected against a subsequent lethal tumour challenge. Moreover, in the C3 sarcoma and the 3LL lung carcinoma murine models, treatment of animals bearing established macroscopic tumours (up to 1 cm2 in size) with tumour peptide-pulsed dendritic cells resulted in sustained tumour regression and tumour-free status in more than 80% of cases. These results support the clinical use of tumour peptide-pulsed dendritic cells as components in developing effective cancer vaccines and therapies.


Assuntos
Células da Medula Óssea , Células Dendríticas/transplante , Neoplasias Pulmonares/terapia , Sarcoma Experimental/terapia , Vacinas Sintéticas/imunologia , Animais , Apresentação de Antígeno , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas
7.
Nat Med ; 3(6): 682-5, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9176498

RESUMO

The 4-1BB glycoprotein is a member of the tumor necrosis factor receptor superfamily and binds to a high-affinity ligand (4-1BBL) expressed on several antigen-presenting cells such as macrophages and activated B cells. Expression of 4-1BB is restricted to primed CD4+ and CD8+ T cells, and 4-1BB signaling either by binding to 4-1BBL or by antibody ligation delivers a dual mitogenic signal for T-cell activation and growth. These observations suggest an important role for 4-1BB in the amplification of T cell-mediated immune responses. We now show that administration of anti-4-1BB monoclonal antibodies can eradicate established large tumors in mice, including the poorly immunogenic Ag104A sarcoma and the highly tumorigenic P815 masto cytoma. The immune response induced by anti-4- 1BB monoclonal antibodies is mediated by both CD8+ and CD4+ T cells and is accompanied by a marked augmentation of tumor-selective cytolytic T-cell activity. Our data suggest that a similar approach may be efficacious for immunotherapy of human cancer.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Sarcoma de Mastócitos/terapia , Receptores de Fator de Crescimento Neural/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Sarcoma Experimental/terapia , Animais , Antígenos CD , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Sarcoma de Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos DBA , Transplante de Neoplasias , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral
8.
J Exp Med ; 168(6): 2031-43, 1988 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-3143799

RESUMO

Intraperitoneal injection of human rIL-1 in a dose of 0.5 microgram daily for 5 d, or 1 microgram daily for 3 d, was capable of causing complete regression of immunogenic SA1 sarcoma growing subcutaneously in syngeneic or semisyngeneic mice. Higher doses of IL-1 were not more therapeutic against the SA1 sarcoma, but needed to be given to cause complete regression of the immunogenic L5178Y lymphoma. On the other hand, the P815 mastocytoma was much less responsive to IL-1 therapy, in that it failed to undergo complete regression in response to doses of IL-1 capable of causing regression of the L5178Y lymphoma. IL-1 caused regression of the SA1 sarcoma when given on days 6-8 of tumor growth, but not when given on days 1-3. This refractoriness of a small tumor to IL-1 therapy suggests that the antitumor action of IL-1 is based on an underlying host-immune response that is not generated until after day 3 of tumor growth. Direct evidence for the participation of host immunity in IL-1-induced tumor regression was supplied by results showing that IL-1 was not therapeutic against the SA1 sarcoma growing in T cell-deficient (TXB) mice, unless these mice were first infused with Ly-2+ and L3T4+ T cells from donor mice bearing an established SA1 sarcoma. In contrast, normal T cells, or T cells from donor mice bearing a YAC-1 lymphoma, failed to provide TXB recipients with the ability to cause regression of their SA-1 sarcoma in response to IL-1 treatment. The results are in keeping with the interpretation that exogenous IL-1, by augmenting the production of tumor-sensitized T cells, converts a subtherapeutic level of host immunity to a therapeutic level. The results suggest, in addition, that IL-1 only stimulates the replication of T cells that are already engaged in the antitumor immune response.


Assuntos
Interleucina-1/farmacologia , Sarcoma Experimental/terapia , Linfócitos T/imunologia , Animais , Síndromes de Imunodeficiência/imunologia , Injeções Intraperitoneais , Leucemia L5178/terapia , Sarcoma de Mastócitos/terapia , Camundongos , Camundongos Endogâmicos , Sarcoma Experimental/imunologia
9.
J Exp Med ; 174(6): 1511-5, 1991 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-1744582

RESUMO

Interleukin 7 (IL-7) is a 25-kD cytokine that was initially described as a pre-B cell growth factor. This cytokine has also been shown to have T cell proliferative and differentiation effects. In this report, we demonstrate that antitumor cytotoxic T lymphocytes (CTL) generated by secondary in vitro sensitization of draining lymph node cells in IL-7 are effective in treating 3-day syngeneic methylcholanthrene (MCA) sarcoma pulmonary metastases in mice. In vivo titrations comparing IL-7 to IL-2 antitumor CTL show that they have equivalent potency in adoptive immunotherapy. IL-7 antitumor CTL generated against MCA sarcomas of weak immunogeneity are also tumor specific in their in vivo efficacy. This study represents the first successful use of a cytokine other than IL-2 for the generation of cells with in vivo efficacy in cellular adoptive transfer.


Assuntos
Imunoterapia Adotiva , Interleucina-7/farmacologia , Sarcoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Feminino , Interleucina-2/farmacologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos C57BL , Sarcoma Experimental/induzido quimicamente
10.
J Exp Med ; 167(3): 1086-99, 1988 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-3258350

RESUMO

In agreement with the results of previous studies (1), it was shown that intravenous injection of endotoxin into mice bearing 9-d SA1 sarcoma resulted in a tumor hemorrhagic reaction that rapidly caused necrosis of most of the center of the tumor, and then the complete regression of the rim of living tumor tissue that survived the hemorrhagic reaction. The tumor hemorrhagic reaction was confined to the vascular bed of the tumor, and its rate and extent of development were measured in terms of the intratumor extravasation of 51Cr-labeled syngeneic red cells. The development of the hemorrhagic reaction was associated with the presence in the tumor over a 6-h period of endogenous TNF that was measured in terms of its capacity to kill L929B cells in vitro and identified by its susceptibility to neutralization with a monospecific, polyvalent anti-rTNF antibody. The same antibody was capable in vivo of inhibiting the endotoxin-induced tumor hemorrhagic reaction by only approximately 50%, even when present in the tumor in excess. However, it was capable when given in the same quantity of inhibiting the ability of endotoxin to cause complete tumor regression. The fact that TNF was generated in the tumor during the tumor hemorrhagic reaction, and that infusion of a sufficient quantity of anti-rTNF antibody severely interfered with hemorrhagic necrosis and prevented tumor regression represents convincing evidence that TNF is an essential participant in endotoxin-induced regression of an established SA1 sarcoma. Moreover, because tumor regression, as opposed to hemorrhagic necrosis, failed to occur if the tumor was growing in immunoincompetent mice, but did so if the mice were infused with tumor-sensitized T cells, it can be concluded that an adequate level of T cell-mediated immunity is also an essential requirement for endotoxin-induced tumor regression. The participation of other endotoxin-induced mediators in tumor regression cannot be ruled out.


Assuntos
Endotoxinas/uso terapêutico , Sarcoma Experimental/terapia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos/imunologia , Hemorragia/induzido quimicamente , Imunização Passiva , Imunocompetência , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Necrose , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/imunologia , Linfócitos T/transplante , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
11.
J Exp Med ; 156(2): 385-97, 1982 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-6980254

RESUMO

We have studied the ability of immunized lymphoid cells expanded in IL-2 to mediate the cure of mice with localized and disseminated syngeneic lymphoma. Mice received 500 rad total-body irradiation before injection of tumor into the footpad. Mice were treated 5 d later when a palpable local tumor and disseminated metastases were present. Intravenous injection of in vivo immune lymphocytes cured 93% of all mice, significantly better than any control group (P less than 0.0005). Immune cells, secondarily sensitized to the FBL-3 tumor in vitro, also conferred significant survival benefit (P less than 0.005) when injected intravenously, curing 79% of the animals treated. When these in vitro sensitized cells were expanded in IL-2, 8-10-fold over 7 d, 93% of the animals thus treated were cured, (P less than 0.005). When these cells were grown for multiple generations in IL-2 they retained their ability to cure mice (56% cured, P less than 0.01). This is the first demonstration that intravenous injection of sensitized cells grown in long term culture in IL-2 is capable of curing mice of established local and disseminated syngeneic tumor.


Assuntos
Fibrossarcoma/terapia , Imunoterapia , Interleucina-2/imunologia , Leucemia Experimental/terapia , Transfusão de Linfócitos , Linfocinas/imunologia , Sarcoma Experimental/terapia , Animais , Sobrevivência Celular , Citotoxicidade Imunológica , Feminino , Fibrossarcoma/imunologia , Leucemia Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Sarcoma Experimental/imunologia , Baço/imunologia , Transplante Isogênico
12.
J Exp Med ; 166(6): 1716-33, 1987 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-3500265

RESUMO

We have shown that two weakly immunogenic MCA sarcomas developed in our laboratory that are sensitive to high-dose IL-2 immunotherapy express class I MHC in vivo and in vitro. Two nonimmunogenic MCA sarcomas are relatively insensitive to IL-2 therapy and express minimal or no class I MHC molecules in vivo and in vitro. To study the role of MHC in the therapy of tumors with IL-2, a class I-deficient murine melanoma, B16BL6, was transfected with the Kb class I gene. Expression of class I MHC rendered B16BL6 advanced pulmonary macrometastases sensitive to IL-2 immunotherapy. 3-d micrometastases of CL8-2, a class I transfected clone of B16BL6, were significantly more sensitive to IL-2 therapy than a control nontransfected line. Expression of Iak, a class II MHC molecule, had no effect on IL-2 therapy of transfectant pulmonary micrometastases in F1 mice. By using lymphocyte subset depletion with mAbs directed against Lyt-2, therapy of class I transfectant macrometastases with high-dose IL-2 was shown to involve an Lyt-2 cell. In contrast, regression of micrometastases treated with low-dose IL-2 involved Lyt-2+ cells, but regression mediated by high doses of IL-2 did not. We hypothesize that both LAK and Lyt-2+ T cells effect IL-2-mediated elimination of micrometastases, but only Lyt-2+ T cells are involved in macrometastatic regression. Low doses of IL-2 stimulate Lyt-2+ cells to eliminate class I-expressing micrometastases, but high doses of IL-2 can recruit LAK cells to mediate regression of micrometastases independent of class I expression. Only high-dose IL-2, mediating its effect predominantly via Lyt-2+ cells, is capable of impacting on MHC class I-expressing macrometastases. Macrometastases devoid of class I MHC antigens appear to be resistant to IL-2 therapy.


Assuntos
Antígenos H-2/imunologia , Interleucina-2/uso terapêutico , Complexo Principal de Histocompatibilidade , Melanoma Experimental/terapia , Linfócitos T/imunologia , Animais , Antígenos Ly/análise , Relação Dose-Resposta a Droga , Antígenos H-2/genética , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Celular , Imunoterapia , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , Camundongos , Metástase Neoplásica , RNA Mensageiro/genética , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Transfecção
13.
J Exp Med ; 167(3): 1067-85, 1988 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-3351434

RESUMO

The ability of murine recombinant tumor necrosis factor (rTNF) and natural TNF in tumor-necrotizing serum (TNS) to cause regression of the SA1 sarcoma was investigated. We found that to cause regression of a 9-d SA1 sarcoma, near lethal quantities of rTNF and TNS had to be given to the host. However, even at these highly toxic doses, rTNF was not reliable at causing complete tumor regression. On the other hand, both types of TNF were reliable at causing a tumor hemorrhagic reaction that resulted in the destruction of greater than 75% of the tumor's center in 24 h. The TNF-induced hemorrhagic reaction involved the development of numerous petechial hemorrhages in the tumor's vascular bed, which apparently resulted from destruction of the tumor's blood vessels. It was possible to follow the development of the hemorrhagic reaction against time after giving rTNF or TNS by measuring the intratumor extravasation of 51Cr-labeled syngeneic red cells. According to this method, TNF-induced intratumor hemorrhaging was in progress within 1 h of giving TNF and continued for about a 6-h period. However, the hemorrhagic reaction was greatly reduced and complete regression of the rim of the living tumor tissue that survived hemorrhagic necrosis failed to occur, if SA1 sarcoma was growing in T cell-deficient (TXB) mice. This indicates that the TNF-induced hemorrhagic reaction is partly dependent, and the tumor regression that follows is completely dependent on host immunocompetence. This suggests in turn, that rTNF does not directly destroy SA1 tumor cells in vivo, even though it was shown that it can destroy SA1 tumor cells in vitro. This interpretation is supported by the additional findings that rTNF was no more therapeutic against a 3-d (3-mm) SA1 than against a 9-d (8-mm) SA1, and was no more therapeutic when injected directly into the tumor than when injected intravenously. Lastly it was possible to completely inhibit the ability of rTNF and TNS to cause tumor hemorrhagic necrosis and regression by infusing the host with a monospecific, polyvalent anti-rTNF antibody that neutralized the cytotoxic action of rTNF in vitro.


Assuntos
Sarcoma Experimental/terapia , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Hemorragia/induzido quimicamente , Imunocompetência , Infusões Intravenosas , Injeções , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Necrose , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/toxicidade
14.
J Exp Med ; 161(5): 1169-88, 1985 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-3886826

RESUMO

Incubation of resting lymphoid cells with recombinant interleukin 2 (IL-2) in vitro leads to the generation of lymphokine activated killer (LAK) cells capable of lysing fresh tumor cell suspensions in short-term chromium-release assays. Our previous studies (7) have demonstrated that the injection of LAK cells plus low doses of recombinant IL-2 were capable of inhibiting the growth of pulmonary metastases. We have now explored the ability of high doses of recombinant IL-2, administered systemically, to generate LAK cells in vivo, and to mediate antitumor effects directly. Administration of increasing doses of recombinant IL-2 intraperitoneally resulted in the generation of LAK cells in the spleens of recipient mice. Doses of 100,000 U recombinant IL-2 administered intraperitoneally approximately every 8 h for 5 d were capable of dramatically inhibiting established 3-d pulmonary metastases from the MCA-105 and MCA-106 syngeneic sarcomas and the syngeneic B16 melanoma in C57BL/6 mice. Grossly visible metastases present at 10 d after tumor injection also underwent regression following IL-2 therapy. Surprisingly, established 10 d pulmonary metastases were more susceptible to the effects of IL-2 than were the smaller 3 d pulmonary metastases. All antitumor effects of the systemic administration of recombinant IL-2 were eliminated if mice received prior treatment with 500 rad total body irradiation. The administration of high doses of recombinant IL-2 was also capable of inhibiting the growth of 3-d established subcutaneous tumors from the MCA-105 sarcoma, and of mediating the inhibition of growth and regression of established palpable subcutaneous MCA-105 sarcomas. Lymphocytes, which appeared morphologically to be activated, were present at the site of regressing tumor, and it appears that the mechanism of the antitumor effect of recombinant IL-2 administered systemically is via the generation of LAK cells in vivo, although this hypothesis remains to be proven. The ready availability of high doses of recombinant human IL-2, and the demonstration of antitumor effects seen in animal models have led us to the initiation of the clinical trials of recombinant IL-2 in humans.


Assuntos
Interleucina-2/administração & dosagem , Neoplasias Pulmonares/terapia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Animais , Esquema de Medicação , Feminino , Interleucina-2/efeitos da radiação , Células Matadoras Naturais/imunologia , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Ativação Linfocitária , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Sarcoma Experimental/terapia , Neoplasias Cutâneas/imunologia , Fatores de Tempo , Irradiação Corporal Total
15.
J Exp Med ; 175(6): 1423-31, 1992 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-1588273

RESUMO

To be recognized by CD8+ T lymphocytes, target cells must process and present peptide antigens in the context of major histocompatibility complex (MHC) class I molecules. The nonimmunogenic, low class I-expressing, methylcholanthrene (MCA)-induced murine sarcoma cell line, MCA 101, is a poor presenter of endogenously generated viral antigens to specific CD8+ T lymphocytes and cannot be used to generate tumor infiltrating lymphocytes (TIL). Since interferon gamma (IFN-gamma) has been shown to upregulate three sets of molecules important for antigen processing and presentation, we retrovirally transduced wild-type MCA 101 (101.WT) tumor with the mIFN-gamma cDNA to create the 101.NAT cell line. Unlike 101.WT, some clones of retrovirally transduced 101.NAT tumor expressed high levels of class I, and could be used to generate CD8+ TIL. More importantly, these TIL were therapeutic in vivo against established pulmonary metastases from the wild-type tumor. Although not uniformly cytotoxic amongst several separate cultures, these TIL did specifically release cytokines (IFN-gamma and tumor necrosis factor-alpha) in response to 101.WT targets. 101.WT's antigen presentation deficit was also reversed by gene modification with mIFN-gamma cDNA. 101.NAT had a greatly improved capacity to present viral antigens to CD8+ cytotoxic T lymphocytes. These findings show that a nonimmunogenic tumor, incapable of generating a CD8+ T cell immune response, could be gene-modified to generate a therapeutically useful immune response against the wild-type tumor. This strategy may be useful in developing treatments for tumor histologies not thought to be susceptible to T cell-based immunotherapy.


Assuntos
Antígenos CD8/imunologia , Interferon gama/genética , Interferon gama/imunologia , Sarcoma Experimental/imunologia , Subpopulações de Linfócitos T/imunologia , Transfecção , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos CD8/análise , Feminino , Citometria de Fluxo , Terapia Genética , Interferon gama/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Leucemia Murina de Moloney/genética , Regiões Promotoras Genéticas , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/terapia , Timidina Quinase/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
J Exp Med ; 183(4): 1357-65, 1996 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8666894

RESUMO

The BALB/c Meth A sarcoma carries a p53 missense mutation at codon 234, which occurs in a peptide, termed 234CM, capable of being presented to cytotoxic T lymphocytes (CTL) by H-2Kd molecules (Noguchi, Y., E.C. Richards, Y.-T. Chen, and L.J. Old. 1994. Proc. Natl. Acad. Sci. USA. 91:3171-3175). Immunization of BALB/c mice with bone marrow-derived dendritic cells (DC), generated in the presence of granulocyte macrophage colony-stimulating factor and interleukin 4, and prepulsed with the Meth A p53 mutant peptide, induced CTL that specifically recognized peptide-pulsed P815 cells, as well as Meth A cells naturally expressing this epitope. Immunization with this vaccine also protected naive mice from a subsequent tumor challenge, and it inhibited tumor growth in mice bearing day 7 subcutaneous Meth A tumors. We additionally determined that immunization of BALB/c mice with DC pulsed with the p53 peptide containing the wild-type residue at position 234, 234CW, induced peptide-specific CTL that reacted against several methylcholanthrene-induced BALB/c sarcomas, including CMS4 sarcoma, and rejection of CMS4 sarcoma in vaccination and therapy (day 7) protocols. These results support the efficacy of DC-based, p53-derived peptide vaccines for the immunotherapy of cancer. The translational potential of this strategy is enhanced by previous reports showing that DC can readily be generated from human peripheral blood lymphocytes.


Assuntos
Fragmentos de Peptídeos/uso terapêutico , Sarcoma Experimental/terapia , Proteína Supressora de Tumor p53/uso terapêutico , Vacinação , Sequência de Aminoácidos , Animais , Sequência de Bases , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
17.
J Exp Med ; 173(3): 647-58, 1991 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-1900079

RESUMO

We have investigated the mechanisms whereby adoptively transferred murine CD8+ lymphocytes mediate tumor regressions. Noncytolytic, CD8+ tumor-infiltrating lymphocytes (TIL) eradicated established lung tumors in irradiated mice. Many cytolytic and noncytolytic CD8+ TIL cultures specifically secreted interferon gamma (IFN-gamma) and tumor necrosis factor when stimulated with tumor cells in vitro. The effectiveness of TIL when adoptively transferred to mice bearing micrometastases correlated better with their ability to specifically secrete lymphokines than with their cytotoxicity in vitro. In 14 of 15 tests, therapeutically effective TIL specifically secreted IFN-gamma in vitro, whereas only 1 of 11 ineffective TIL specifically secreted IFN-gamma. In contrast, only 8 of 15 therapeutically effective TIL were cytolytic. Antibodies to TNF inhibited the effectiveness of two adoptively transferred TIL cultures. In five experiments, antibodies to IFN-gamma abrogated the ability of four different CD8+ TIL cultures to mediate tumor regressions, indicating that secretion of IFN-gamma is an essential part of the mechanism of action of TIL.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Fibrossarcoma/terapia , Imunoterapia Adotiva , Interferon gama/imunologia , Neoplasias Pulmonares/secundário , Sarcoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos Monoclonais , Antígenos CD8 , Células Cultivadas , Citotoxicidade Imunológica , Feminino , Fibrossarcoma/patologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos C57BL , Sarcoma Experimental/patologia , Linfócitos T Citotóxicos/transplante
18.
J Exp Med ; 181(2): 619-29, 1995 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-7836917

RESUMO

Mice carrying large established major histocompatibility complex (MHC) class 1+ sarcoma tumors can be successfully treated by immunization with genetically engineered sarcoma cells transfected with syngeneic MHC class II plus B7-1 genes. This approach is significantly more effective than previously described strategies using cytokine- or B7-transduced tumor cells which are only effective against smaller tumor loads, and which cannot mediate regression of longer-term established tumors. The most efficient tumor rejection occurs if both the class II and B7-1 molecules are coexpressed on the same tumor cell. Immunity induced by immunization with class II+B7-1(+)-transfected sarcoma cells involves CD4+ and CD8+ T cells, suggesting that the increased effectiveness of the transfectants is due to their ability to activate both of these T cell populations.


Assuntos
Antígenos de Histocompatibilidade Classe II/uso terapêutico , Imunoterapia , Sarcoma Experimental/terapia , Animais , Células Cultivadas , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Masculino , Camundongos , Camundongos Nus , Sarcoma Experimental/imunologia , Transfecção , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
19.
J Exp Med ; 183(2): 639-44, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8627175

RESUMO

Genetic modification of many types of mouse tumors to express the B7-1 or B7-2 molecules, natural ligands for the T cell-costimulatory molecule CD28, increases their immunogenicity. However, even after transfection with the B7-1 and/or B7-2 genes, poorly immunogenic tumors fail to elicit and efficient immune response. We report here that two such tumors, the Ag104A sarcoma and the K1735-M2 melanoma, become immunogenic after transfection of the genes encoding murine B7-1 together with CD48, which is the natural ligand for CD2. Tumor-specific CD8+ cytotoxic T lymphocytes were readily generated and were effective for adoptive immunotherapy of metastasis induced by wild-type Ag104A sarcoma cells. A similar approach may be useful for developing therapy for other poorly immunogenic tumors, including those in humans.


Assuntos
Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Imunização , Melanoma Experimental/terapia , Sarcoma Experimental/terapia , Animais , Apresentação de Antígeno , Antígenos CD/genética , Antígeno B7-1/genética , Sequência de Bases , Antígeno CD48 , Clonagem Molecular , Feminino , Citometria de Fluxo , Imunoterapia Adotiva , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C3H , Modelos Imunológicos , Dados de Sequência Molecular , Metástase Neoplásica , Receptores de Antígenos de Linfócitos T , Proteínas Recombinantes/metabolismo , Sarcoma Experimental/imunologia , Transdução de Sinais , Transfecção
20.
Cancer Immunol Immunother ; 59(8): 1235-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20376439

RESUMO

Natural killer (NK) cells represent a promising cell type to utilize for effective adoptive immunotherapy. However, little is known about the important cytolytic molecules and signaling pathways used by NK cells in the adoptive transfer setting. To address this issue, we developed a novel mouse model to investigate the trafficking and mechanism of action of these cells. We demonstrate that methylcholanthrene-induced RKIK sarcoma cells were susceptible to NK cell-mediated lysis in vitro and in vivo following adoptive transfer of NK cells in C57BL/6 RAG-2(-/-)gammac(-/-) mice. Cytotoxic molecules perforin, granzymes B and M as well as the death ligand TRAIL and pro-inflammatory cytokine IFN-gamma were found to be important in the anti-tumor effect mediated by adoptively transferred NK cells. Importantly, we demonstrate that adoptively transferred NK cells could traffic to the tumor site and persisted in vivo which correlated with the anti-tumor effect observed. Overall, the results of this study have important implications for enhancing NK cell-based immunotherapies.


Assuntos
Citotoxicidade Imunológica , Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Interleucina-2/imunologia , Interleucina-2/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Sarcoma Experimental/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA