Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Biother Radiopharm ; 18(2): 133-45, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12804039

RESUMO

A study was undertaken to investigate the suitability of using a high affinity (Kd = 1.1 nM) anti-CD45 monoclonal antibody for delivering the high energy beta-particle emitting isotope (90)Y to lymphohematopoietic target cells in vivo. The antibody, AHN-12, recognized the tyrosine phosphatase CD45 expressed on the surface of normal and malignant hematopoietic cells and studies showed that it reacted with both CD45-expressing normal peripheral blood cells and leukemia cells from patients. The antibody was readily labeled with (90)Y using the highly stable chelate 1B4M-DTPA and the radioimmunoconjugate was designated (90)Y-anti-CD45. The agent selectively bound to CD45(+) B cell line Daudi, but not CD45(-) control cells and significantly (p = 0.007) more bound to Daudi tumors growing in athymic nude mice than did a control non-reactive antibody. Moreover, biodistribution data correlated well to an anti-Daudi effect observed against established tumors in nude mice. The effect was dose dependent and irreversible with the best results in mice receiving a single dose of 137 microCi (90)Y-anti-CD45. These mice displayed a significantly (p < 0.0095) better anti-tumor effect than a control (90)Y-labeled antibody and survived over 135 days with no evidence of tumor. Histology studies showed no significant injury to kidney, liver, or small intestine even at 254 microCi, the highest dose tested. Because radiolabeled anti-CD45 antibody can be used to deliver radiation selectively to lymphohematopoietic tissue, these data indicate that this agent may be used to improve treatment of hematopoietic malignancies, particularly leukemia and lymphoma, when combined with hematopoietic stem cell transplantation in a future clinical trial.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfoma de Burkitt/radioterapia , Células-Tronco Hematopoéticas/efeitos da radiação , Leucemia Mieloide/radioterapia , Antígenos Comuns de Leucócito/imunologia , Radioisótopos de Ítrio/uso terapêutico , Animais , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Radioisótopos de Índio , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Ácido Pentético , Radioimunoterapia , Distribuição Tecidual , Células Tumorais Cultivadas
2.
Blood ; 96(3): 1157-65, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910937

RESUMO

Immunotoxin (IT) therapy shows potential for selectively eliminating GVHD-causing T cells in vivo, but the field has been hampered by toxicity. Previously, we showed that a genetically engineered IT consisting of a single-chain protein, including the anti-CD3sFv spliced to a portion of diphtheria-toxin (DT(390)) has anti-GVHD effects, but pronounced organ toxicity common to this class of agent. A recombinant DT(390) anti-CD3sFv protein previously shown to have anti-GVHD activity was modified to reduce its filtration into kidney by genetically inserting a cysteine residue downstream of the sFv moiety at the c-terminus of the protein. This modification produced an intermolecular disulfide bridge, resulting in a bivalent, rather than a monovalent IT, termed SS2, that selectively inhibited T-cell proliferation in vitro. Although monomer and SS2 were similar in in vitro activity, SS2 had a superior therapeutic index in vivo with at least 8-fold more being tolerated with reduced kidney toxicity. Most importantly, in a lethal model of GVHD, 40 microg SS2 given for 1 day, protected 100% of the mice from lethal GVHD for 3 months, whereas the maximum tolerated dose (MTD) of monomer protected only 33%. To our knowledge, this is the first time disulfide bonded ITs have been created in this way and this simple molecular modification may address several problems in the IT field because it (1) markedly increased efficacy curing mice of GVHD after a single daily treatment, (2) markedly decreased organ toxicity, (3) increased the tolerated dosage, and (4) created a therapeutic window where none existed before.


Assuntos
Complexo CD3/imunologia , Toxina Diftérica/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Imunotoxinas/imunologia , Linfócitos T/imunologia , Animais , Cisteína , Toxina Diftérica/administração & dosagem , Doença Enxerto-Hospedeiro/imunologia , Fragmentos de Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/imunologia , Imunotoxinas/administração & dosagem , Camundongos
3.
J Biol Chem ; 272(38): 23905-11, 1997 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-9295340

RESUMO

Palytoxin is a novel skin tumor promoter that does not activate protein kinase C. Previous studies demonstrated that palytoxin stimulates a sodium-dependent signaling pathway that activates the c-Jun NH2-terminal kinase/stress-activated protein kinase (JNK) in Swiss 3T3 fibroblasts. In this study we show that a JNK kinase known as the stress-activated protein kinase/extracellular signal-regulated kinase-1 (SEK1) plays an important role in the regulation of JNK by palytoxin. We found that palytoxin stimulates the sustained activation of both JNK and SEK1 in COS7 and HeLa cells. Transiently expressed SEK1 isolated from palytoxin-treated cells can phosphorylate and activate JNK, which, in turn, can phosphorylate c-Jun. Furthermore, expression of a dominant negative mutant of SEK1 blocks activation of JNK by palytoxin. Sodium appears to play an important role in the regulation of JNK and SEK1 by palytoxin. Activation of JNK and SEK1 by palytoxin, but not anisomycin, requires extracellular sodium. Complementary studies showed that the sodium ionophore gramicidin can mimic palytoxin by regulating JNK and SEK1 through a sodium-dependent mechanism. Collectively, these results demonstrate that palytoxin stimulates a sodium-dependent signaling pathway that activates the SEK1/JNK/c-Jun protein kinase cascade.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno , Proteínas Quinases/metabolismo , Transdução de Sinais , Sódio/metabolismo , Acrilamidas/farmacologia , Animais , Células COS , Venenos de Cnidários , Ativação Enzimática , Feminino , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno
4.
Cancer Res ; 56(3): 637-44, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8564984

RESUMO

Previous studies have shown that structurally diverse tumor promoters can modulate protein kinases involved in signal transduction. In this study, we show that palytoxin, a potent non-12-O-tetradecanoylphorbol-13-acetate (TPA)-type skin tumor promoter, induces a signaling pathway leading to the activation of the stress-activated protein kinases/c-Jun N-terminal kinases (JNK) in Swiss 3T3 fibroblasts. Treatment of cells with doses as low as 0.1 mN palytoxin results in significant activation of JNK. In contrast to epidermal growth factor, which induces a transient activation of JNK in Swiss 3T3 cells, palytoxin causes prolonged enzyme activation. Since stimulation of ion flux appears to play an important role in the mechanism of action of palytoxin in other systems, we investigated the role of sodium and calcium in the activation of JNK: (a) our results show that incubation of Swiss 3T3 cells in a sodium-free medium dramatically reduced the magnitude of JNK activation by palytoxin; and (b) we found that the sodium ionophore gramicidin activates JNK. Together, these results suggest that sodium influx, which is a hallmark of palytoxin action, may play a key role in the activation of JNK by palytoxin. Our results indicate that calcium influx is not necessary or sufficient for palytoxin-induced activation of JNK. In contrast to palytoxin, the TPA-type tumor promoter phorbol 12,13-dibutyrate and the non-TPA-type tumor promoters thapsigargin and okadaic acid do not appear to activate JNK in this system. In contrast to phorbol 12,13-dibutyrate, palytoxin does not activate the p42/p44 mitogen-activated protein kinases. Our results demonstrate that Swiss 3T3 fibroblasts, palytoxin can activate a protein kinase signaling pathway that is distinct from that activated by the prototypical phorbol ester tumor promoters and other potent skin tumor promoters.


Assuntos
Acrilamidas/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Carcinógenos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3/efeitos dos fármacos , Células 3T3/enzimologia , Animais , Venenos de Cnidários , Ativação Enzimática/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Sódio/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...