Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Inflamm Res ; 52(12): 508-11, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14991079

RESUMO

OBJECTIVE: Inflammatory bowel diseases such as ulcerative colitis and Crohn's disease are characterized by chronic relapsing inflammation. The transcription of many of the proteins which mediate the pathogenesis in inflammatory bowel disease (e.g., TNFalpha, ICAM-1, VCAM-1) is NF-kappaB-dependent. IkappaB kinase is critical in transducing the signal-inducible activation of NF-kappaB and, therefore, represents a potentially promising target for the development of novel agents to treat inflammatory bowel disease and other inflammatory diseases. RESULTS: Here we show that BMS-345541, a highly selective inhibitor of IkappaB kinase, inhibited the TNFalpha-induced expression of both ICAM-1 and VCAM-1 in human umbilical vein endothelial cells at the same concentration range as cytokine expression is inhibited in monocytic cells (IC(50) congruent with 5 microM). Against dextran sulfate sodium-induced colitis in mice, BMS-345541 administered orally at doses of 30 and 100 mg/kg was effective in blocking both clinical and histological endpoints of inflammation and injury. CONCLUSION: This represents the first example of an inhibitor of IkappaB kinase with anti-inflammatory activity in vivo and indicates that inhibitors of IkB kinase show the promise of being highly efficacious in inflammatory disorders such as inflammatory bowel disease.


Assuntos
Moléculas de Adesão Celular/metabolismo , Colite/patologia , Sulfato de Dextrana/farmacologia , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinoxalinas/farmacologia , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Humanos , Quinase I-kappa B , Imidazóis/química , Imidazóis/uso terapêutico , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Quinoxalinas/química , Quinoxalinas/uso terapêutico , Sulfassalazina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
2.
J Immunol ; 162(9): 5547-55, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10228036

RESUMO

Oncostatin M (OM) is a pleiotropic cytokine produced late in the activation cycle of T cells and macrophages. In vitro it shares properties with related proteins of the IL-6 family of cytokines; however, its in vivo properties and physiological function are as yet ill defined. We show that administration of OM inhibited bacterial LPS-induced production of TNF-alpha and lethality in a dose-dependent manner. Consistent with these findings, OM potently suppressed inflammation and tissue destruction in murine models of rheumatoid arthritis and multiple sclerosis. T cell function and Ab production were not impaired by OM treatment. Taken together these data indicate the activities of this cytokine in vivo are antiinflammatory without concordant immunosuppression.


Assuntos
Citocinas/fisiologia , Mediadores da Inflamação/fisiologia , Peptídeos/fisiologia , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Artrite Reumatoide/prevenção & controle , Citocinas/administração & dosagem , Citocinas/biossíntese , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/prevenção & controle , Feminino , Membro Posterior , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Lipopolissacarídeos/administração & dosagem , Ativação Linfocitária/imunologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oncostatina M , Peptídeos/administração & dosagem , Peptídeos/metabolismo
3.
Toxicol Pathol ; 27(2): 151-5, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10207978

RESUMO

Oncostatin M (OM) is a member of the interleukin-6 (IL-6) cytokine subfamily. The binding of OM to its receptor initiates signal transduction through JAK-signal transducers and activators of transcription (STAT) pathways and activates transcription activators through mitogen-activated protein (MAP) kinases. Results of in vitro assays documented that OM modulates cytokine expression and alters the production of proteases that down-regulate inflammation. Administration of OM to lipopolysaccharide (LPS)-challenged mice lowered serum tumor necrosis factor-alpha (TNF-alpha) levels and decreased the lethal effects of LPS administration. OM also reduced inflammation in animal models of human disease, including inflammatory bowel disease, antibody-induced arthritis, and experimental autoimmune encephalomyelitis. Preclinical safety studies have been conducted in the mouse and monkey. Mice were administered OM (subcutaneously) at 72, 360, or 1,560 micrograms/kg/day in a 2-wk toxicity study. Decreased body weights occurred at 1,560 micrograms/kg. Drug-related changes at 360 and 1,560 micrograms/kg consisted of dermal irritation at the injection site, leukopenia, and thymic lymphoid depletion; all changes were reversible following a 2-wk recovery period. In a 2-wk subcutaneous study in monkeys, OM was administered at 1, 5, 15, 45, or 150 micrograms/kg/day. At all doses there was reversible, transient inappetence and dermal irritation at the injection site. Drug-related changes at 5, 15, 45, and 150 micrograms/kg consisted of reversible elevations in both serum amyloid A and IL-6, and reversible thymic lymphoid depletion. Transient increases in body temperature occurred at 15, 45, and 150 micrograms/kg. The observed spectrum of immunomodulatory effects suggests that OM may have therapeutic utility in treating chronic inflammatory diseases.


Assuntos
Antineoplásicos/farmacologia , Peptídeos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/uso terapêutico , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Oncostatina M , Peptídeos/uso terapêutico
4.
Blood ; 86(4): 1310-5, 1995 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-7632937

RESUMO

Oncostatin M (OM) is a 28-kD glycoprotein that exhibits a panoply of biologic effects. Based on histologic observations of increased splenic megakaryocytes in nude mice implanted with an OM-secreting cell line, the thrombocytopoietic properties of OM in mice were investigated in culture and in vivo. Alone, OM did not induce megakaryocytic colony formation, but in combination with murine interleukin-3 (IL-3), OM markedly enhanced colony formation. The effects of OM on colony formation were similar to those of IL-6. OM alone augmented acetylcholinesterase in short-term marrow cultures. In normal mice, the administration of OM augmented platelet counts without increasing other circulating blood cell counts. The increment in counts exceeded that observed with IL-6. The kinetics of the OM response suggested that maximal increases in platelets occurred 3 days after the cessation of OM administration, irrespective of the duration of administration. In irradiated mice, OM administration accelerated platelet recovery and prevented the decrease in red blood cells observed in irradiated control animals. The data show that OM behaves as a megakaryocytic maturation factor in vitro and augments platelet production in vivo. Based on these animal data, OM may have potential clinical utility as a thrombocytopoietic agent.


Assuntos
Plaquetas/citologia , Hematopoese/efeitos dos fármacos , Peptídeos/administração & dosagem , Anemia/tratamento farmacológico , Animais , Sinergismo Farmacológico , Feminino , Interleucina-3/administração & dosagem , Interleucina-6/administração & dosagem , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos C3H , Oncostatina M , Contagem de Plaquetas/efeitos dos fármacos , Fatores de Tempo
6.
Cancer Res ; 55(11): 2357-65, 1995 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-7757987

RESUMO

A cephalosporin derivative of doxorubicin (C-Dox) was evaluated as a prodrug for activation by mAb conjugates of the beta-lactamase from Enterobacter cloacae P99 (beta L; EC 3.5.2.6). The conjugates consisted of beta L and the F(ab') fragments of either of the mAbs L6, P1.17, or 96.5. L6 binds to antigens on a variety of carcinomas, including the two lung adenocarcinoma cell lines H2981 and H2987 used in this study. 96.5 binds to the melanoma-associated antigen p97, and P1.17 was used for the control conjugate. C-Dox was found to be less cytotoxic to three different tumor cell lines in vitro compared to the parent drug doxorubicin (Dox). Immunospecific activation took place when the cells were pretreated with beta L conjugates that could bind to antigens on the tumor cells. In vivo toxicity and pharmacokinetics studies in athymic female nu/nu mice revealed that C-Dox was at least 7-fold less toxic than Dox (on a molar basis), despite the fact that a > or = 320-fold greater area-under-the-curve (blood concentration versus time) of C-Dox compared to Dox was obtained 0-2 h after administration of the two agents. Pharmacokinetic studies at maximum tolerated doses in mice bearing xenografts of either H2981 or H2987 revealed that the intratumoral levels of Dox after treatment with L6-beta L in combination with C-Dox were higher than were obtained by either systemic treatment with Dox or a combination of P1.17-beta L and C-Dox. This finding suggested that the conversion of C-Dox to Dox was tumor specific and dependent on the presence of the targeted antigen. Furthermore, the best antitumor activity against both H2981 and H2987 tumors was obtained by treatment with L6-beta L and C-Dox compared to P1.17-beta L and C-Dox or Dox alone. Thus, higher levels of Dox corresponded to greater therapeutic effects in both of the tumor models studied.


Assuntos
Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Imunotoxinas/farmacocinética , Imunotoxinas/toxicidade , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , beta-Lactamases/administração & dosagem , beta-Lactamases/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Biotransformação , Doxorrubicina/toxicidade , Feminino , Humanos , Hidrólise , Fragmentos de Imunoglobulinas/metabolismo , Fragmentos de Imunoglobulinas/toxicidade , Cinética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Pró-Fármacos/toxicidade , Sensibilidade e Especificidade , Distribuição Tecidual , Transplante Heterólogo , beta-Lactamases/farmacocinética
7.
Transplantation ; 58(5): 602-10, 1994 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8091487

RESUMO

Graft-versus-host disease (GVHD), a pathological condition associated with BMT, results from activation of donor T lymphocytes by host tissues. CD28 and CTLA-4 are structurally related T cell receptors for members of the B7 (CD80) gene family, which transmit important costimulatory signals for T cell activation in vitro and in vivo. Here we have investigated the effects of CTLA4Ig, a soluble form of CTLA-4, on lethal GVHD in a murine model. Lethal GVHD was induced by transfer of parent C57BL/6 bone marrow and spleen cells into lethally irradiated (C57BL/6 x DBA/2)F1 recipients. Short courses of treatment with CTLA4Ig did not block engraftment, but prolonged survival of BMT recipients even when administration was delayed for 6 days after transplantation. CTLA4Ig-treated survivors of GVHD maintained body weight and did not exhibit visible signs of GVHD. However, treatment regimens that maximally prolonged survival did not detectably prevent T cell-mediated hematological abnormalities associated with GVHD, including pancytopenia and abnormal cellular composition of the spleen. Our data thus show that the lethality of acute GVHD in this model system is more dependent upon CD28/CTLA-4 costimulation than are other GVHD-associated abnormalities, and can be blocked for an extended period by brief treatment with CTLA4Ig.


Assuntos
Antígenos de Diferenciação/uso terapêutico , Transplante de Medula Óssea/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Imunoconjugados , Complexo Principal de Histocompatibilidade/imunologia , Abatacepte , Animais , Antígenos CD , Linfócitos B/metabolismo , Antígenos CD28/fisiologia , Células CHO , Antígeno CTLA-4 , Cricetinae , Modelos Animais de Doenças , Feminino , Doença Enxerto-Hospedeiro/sangue , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
8.
Cancer Res ; 54(10): 2719-23, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168103

RESUMO

Cytosine deaminase (CD) is a microbial enzyme that can convert the antifungal agent 5-fluorocytosine (5-FC) into the antitumor agent, 5-fluorouracil (5-FU). The enzyme was chemically conjugated to the L6 monoclonal antibody, forming a conjugate that bound to antigens on the H2981 lung adenocarcinoma. Detailed studies were undertaken to determine the extent to which L6-CD generated 5-FU in tumor-bearing mice. Very high tumor:blood ratios of L6-CD (42:1) in vivo were obtained by injecting the conjugate followed 24 h later by an antiidiotypic antibody that could bind to circulating L6-CD but not to L6-CD that was bound to H2981 cells. As a result, significantly more 5-FC could be administered (> 800 mg/kg) than 5-FU (90 mg/kg). L6-CD converted 5-FC into 5-FU such that the L6-CD/antiidiotypic monoclonal antibody/5-FC combination resulted in 17 times more intratumoral 5-FU compared to systemic 5-FU administration. The conversion was antigen dependent since much lower intratumoral 5-FU levels were obtained in H3719 tumors that failed to localize L6-CD. The conversion of 5-FC into 5-FU was low in blood, kidneys, and liver. This demonstrates that a major increase in intratumoral drug concentrations can be attained with an monoclonal antibody-enzyme conjugate in combination with an anticancer prodrug compared to systemic drug therapy.


Assuntos
Anticorpos Monoclonais , Flucitosina/metabolismo , Fluoruracila/metabolismo , Nucleosídeo Desaminases/farmacologia , Pró-Fármacos/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Citosina Desaminase , Feminino , Flucitosina/farmacocinética , Fluoruracila/farmacocinética , Camundongos , Camundongos Nus , Nucleosídeo Desaminases/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA