Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Commun Biol ; 4(1): 44, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420283

RESUMO

Bispecific T cell engaging antibodies (BiTEs) address tumor associated antigens that are over-expressed on cancer but that can also be found on healthy tissues, causing substantial on-target/off-tumor toxicities. To overcome this hurdle, we recently introduced hemibodies, a pair of complementary antibody fragments that redirect T cells against cancer-defining antigen combinations. Here we show that hemibodies addressing CD38 and SLAMF7 recruit T cells for the exquisite elimination of dual antigen positive multiple myeloma cells while leaving single antigen positive bystanders unharmed. Moreover, CD38 and SLAMF7 targeting BiTEs, but not hemibodies induce massive cytokine release and T cell fratricide reactions, a major drawback of T cell recruiting strategies. Together, we provide evidence in vitro and in vivo that hemibodies can be developed for the effective and highly specific immunotherapy of multiple myeloma.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Imunoterapia/métodos , Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/terapia , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , ADP-Ribosil Ciclase 1/metabolismo , Linhagem Celular Tumoral , Humanos , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiplo/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo
3.
Nat Commun ; 10(1): 5387, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31772172

RESUMO

T cell-engaging immunotherapies are changing the landscape of current cancer care. However, suitable target antigens are scarce, restricting these strategies to very few tumor types. Here, we report on a T cell-engaging antibody derivative that comes in two complementary halves and addresses antigen combinations instead of single molecules. Each half, now coined hemibody, contains an antigen-specific single-chain variable fragment (scFv) fused to either the variable light (VL) or variable heavy (VH) chain domain of an anti-CD3 antibody. When the two hemibodies simultaneously bind their respective antigens on a single cell, they align and reconstitute the original CD3-binding site to engage T cells. Employing preclinical models for aggressive leukemia and breast cancer, we show that by the combinatorial nature of this approach, T lymphocytes exclusively eliminate dual antigen-positive cells while sparing single positive bystanders. This allows for precision targeting of cancers not amenable to current immunotherapies.


Assuntos
Anticorpos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Complexo CD3/metabolismo , Imunoterapia/métodos , Linfócitos T/imunologia , Animais , Anticorpos/genética , Antineoplásicos Imunológicos/imunologia , Sítios de Ligação , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Efeito Espectador , Linhagem Celular Tumoral , Feminino , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Medicina de Precisão/métodos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Blood ; 129(1): 100-104, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27784674

RESUMO

The CD19 antigen is a promising target for immunotherapy of acute lymphoblastic leukemia (ALL), but CD19- relapses remain a major challenge in about 10% to 20% of patients. Here, we analyzed 4 CD19- ALL relapses after treatment with the CD19/CD3 bispecific T-cell engager (BiTE) blinatumomab. Three were on-drug relapses, with the CD19- escape variant first detected after only 2 treatment courses. In 1 patient, the CD19- clone appeared as a late relapse 19 months after completion of blinatumomab treatment. All 4 cases showed a cellular phenotype identical to the primary diagnosis except for CD19 negativity. This argued strongly in favor of an isolated molecular event and against a common lymphoid CD19- progenitor cell or myeloid lineage shift driving resistance. A thorough molecular workup of 1 of the cases with early relapse confirmed this hypothesis by revealing a disrupted CD19 membrane export in the post-endoplasmic reticulum compartment as molecular basis for blinatumomab resistance.


Assuntos
Antígenos CD19/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adulto , Idoso , Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/uso terapêutico , Western Blotting , Membrana Celular/metabolismo , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Transporte Proteico
5.
Br J Haematol ; 160(4): 465-76, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23252346

RESUMO

The heat shock transcription factor 1 (HSF1) has recently been reported to promote malignant transformation and growth. Here we provide experimental evidence for a role of HSF1 in the pathogenesis of multiple myeloma (MM). Immunohistochemical analyses revealed that HSF1 was overexpressed in half of the investigated MM samples, including virtually all cases with extramedullary manifestations or anaplastic morphology. HSF1 function was inhibited either by siRNA-mediated knockdown or pharmacologically through treatment with triptolide. Both approaches caused depletion of HSF1, lowered the constitutively high expression of a multitude of protective HSPs (such as HSP90, HSP70, HSP40 and HSP27), induced apoptosis in human MM cells in vitro, and strongly reduced MM tumour growth in vivo. Furthermore, we observed that treatment-induced upregulation of HSPs after proteasome or HSP90 inhibition was critically dependent on HSF1. Importantly, the apoptotic effects of the HSP90 inhibitor NVP-AUY922 or the proteasome inhibitor bortezomib were strongly enhanced in combination with triptolide, suggesting a salvage role of HSF1-dependent HSP induction in response to drug treatment. Collectively, our data indicate that inhibition of HSF1 affects multiple protective HSPs and might therefore represent a therapeutic strategy - in particular in combination with proteasome or HSP90 inhibitors.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mieloma Múltiplo/tratamento farmacológico , Fatores de Transcrição/fisiologia , Animais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ácidos Borônicos/farmacologia , Bortezomib , Células Cultivadas , Proteínas de Ligação a DNA/antagonistas & inibidores , Diterpenos/farmacologia , Regulação para Baixo , Compostos de Epóxi/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Fatores de Transcrição de Choque Térmico , Proteínas de Choque Térmico/metabolismo , Humanos , Imuno-Histoquímica , Isoxazóis/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Fenantrenos/farmacologia , Plasmócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirazinas/farmacologia , RNA Interferente Pequeno/farmacologia , Resorcinóis/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Transplante Heterólogo
6.
Anticancer Res ; 32(2): 453-62, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22287732

RESUMO

BACKGROUND: HSP90 inhibitors effectively reduce expression and activity levels of oncogenic survival proteins. However, their clinical anti-multiple myeloma (MM) activity has been found to be rather weak, spurring the exploration of combination therapies and development of compounds with improved physicochemical properties. MATERIALS AND METHODS: Preclinical effects of the novel orally bioavailable HSP90 inhibitor NVP-HSP990 on the viability, apoptosis and client protein levels of MM cells (established cell lines and clinical specimens) were tested alone and in combination with other drugs. RESULTS: NVP-HSP990 exerted profound activity against MM cells, with a molecular mode of action conforming well with its role as HSP90 inhibitor. Enhanced activity was most obvious in combination with melphalan. Combination with a phosphatidylinositol-3-kinase (PI3-kinase)/mammalian target of rapamycin (mTOR) inhibitor, rendered the HSP90 blockade-mediated stress response ineffective and considerably increased the anti-MM toxicity. CONCLUSION: Given the current interest in both HSP90 and PI3-kinase/mTOR as potential clinical targets, these observations could broaden the therapeutic utility of either class of inhibitor in MM.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Piridonas/farmacologia , Pirimidinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Furanos/farmacologia , Proteínas de Choque Térmico HSP72/antagonistas & inibidores , Humanos , Mieloma Múltiplo/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/farmacologia , Piridonas/administração & dosagem , Pirimidinas/administração & dosagem , Regulação para Cima/efeitos dos fármacos
7.
Blood ; 118(24): 6392-8, 2011 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21860020

RESUMO

Proinflammatory cytokines such as TNFα are elevated in patients with myeloproliferative neoplasms (MPN), but their contribution to disease pathogenesis is unknown. Here we reveal a central role for TNFα in promoting clonal dominance of JAK2(V617F) expressing cells in MPN. We show that JAK2(V617F) kinase regulates TNFα expression in cell lines and primary MPN cells and TNFα expression is correlated with JAK2(V617F) allele burden. In clonogenic assays, normal controls show reduced colony formation in the presence of TNFα while colony formation by JAK2(V617F)-positive progenitor cells is resistant or stimulated by exposure to TNFα. Ectopic JAK2(V617F) expression confers TNFα resistance to normal murine progenitor cells and overcomes inherent TNFα hypersensitivity of Fanconi anemia complementation group C deficient progenitors. Lastly, absence of TNFα limits clonal expansion and attenuates disease in a murine model of JAK2(V617F)-positive MPN. Altogether our data are consistent with a model where JAK2(V617F) promotes clonal selection by conferring TNFα resistance to a preneoplastic TNFα sensitive cell, while simultaneously generating a TNFα-rich environment. Mutations that confer resistance to environmental stem cell stressors are a recognized mechanism of clonal selection and leukemogenesis in bone marrow failure syndromes and our data suggest that this mechanism is also critical to clonal selection in MPN.


Assuntos
Transformação Celular Neoplásica/metabolismo , Janus Quinase 2/metabolismo , Transtornos Mieloproliferativos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Substituição de Aminoácidos , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Leucemia Mieloide Crônica Atípica BCR-ABL Negativa/sangue , Leucemia Mieloide Crônica Atípica BCR-ABL Negativa/tratamento farmacológico , Leucemia Mieloide Crônica Atípica BCR-ABL Negativa/genética , Leucemia Mieloide Crônica Atípica BCR-ABL Negativa/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mutantes/metabolismo , Células Progenitoras Mieloides/metabolismo , Transtornos Mieloproliferativos/sangue , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/genética , Mutação Puntual , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Fator de Necrose Tumoral alfa/genética
9.
Blood ; 116(17): 3278-85, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-20519627

RESUMO

Point mutations in the kinase domain of BCR-ABL are the most common mechanism of drug resistance in chronic myeloid leukemia (CML) patients treated with ABL kinase inhibitors, including imatinib. It has also been shown in vitro that mutations outside the kinase domain in the neighboring linker, SH2, SH3, and Cap domains can confer imatinib resistance. In the context of ABL, these domains have an autoinhibitory effect on kinase activity, and mutations in this region can activate the enzyme. To determine the frequency and relevance to resistance of regulatory domain mutations in CML patients on imatinib, we screened for such mutations in a cohort of consecutive CML patients with various levels of response. Regulatory domain mutations were detected in 7 of 98 patients, whereas kinase domain mutations were detected in 29. One mutation (T212R) conferred in vitro tyrosine kinase inhibitor resistance and was associated with relapse, whereas most other mutations did not affect drug sensitivity. Mechanistic studies showed that T212R increased the activity of ABL and BCR-ABL and that T212R-induced resistance may be partially the result of stabilization of an active kinase conformation. Regulatory domain mutations are uncommon but may explain resistance in some patients without mutations in the kinase domain.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Mutação , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Domínios de Homologia de src , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular , Estudos de Coortes , Feminino , Proteínas de Fusão bcr-abl/química , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Adulto Jovem
10.
Blood ; 115(25): 5232-40, 2010 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-20385788

RESUMO

Activating alleles of Janus kinase 2 (JAK2) such as JAK2(V617F) are central to the pathogenesis of myeloproliferative neoplasms (MPN), suggesting that small molecule inhibitors targeting JAK2 may be therapeutically useful. We have identified an aminopyrimidine derivative (CYT387), which inhibits JAK1, JAK2, and tyrosine kinase 2 (TYK2) at low nanomolar concentrations, with few additional targets. Between 0.5 and 1.5muM CYT387 caused growth suppression and apoptosis in JAK2-dependent hematopoietic cell lines, while nonhematopoietic cell lines were unaffected. In a murine MPN model, CYT387 normalized white cell counts, hematocrit, spleen size, and restored physiologic levels of inflammatory cytokines. Despite the hematologic responses and reduction of the JAK2(V617F) allele burden, JAK2(V617F) cells persisted and MPN recurred upon cessation of treatment, suggesting that JAK2 inhibitors may be unable to eliminate JAK2(V617F) cells, consistent with preliminary results from clinical trials of JAK2 inhibitors in myelofibrosis. While the clinical benefit of JAK2 inhibitors may be substantial, not the least due to reduction of inflammatory cytokines and symptomatic improvement, our data add to increasing evidence that kinase inhibitor monotherapy of malignant disease is not curative, suggesting a need for drug combinations to optimally target the malignant cells.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Neoplasias Hematológicas/tratamento farmacológico , Hematopoese/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Transtornos Mieloproliferativos/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Animais , Apoptose/genética , Apoptose/imunologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Hematológicas/enzimologia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Hematopoese/genética , Hematopoese/imunologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/genética , Janus Quinase 1/imunologia , Janus Quinase 1/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/imunologia , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/enzimologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/imunologia
11.
Blood ; 112(5): 1960-70, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18559973

RESUMO

BCR-ABL is proposed to impair cell-cycle control by disabling p27, a tumor suppressor that inhibits cyclin-dependent kinases. We show that in cell lines p27 expression is inversely correlated with expression of SKP2, the F-box protein of SCF(SKP2) (SKP1/Cul1/F-box), the E3 ubiquitin ligase that promotes proteasomal degradation of p27. Inhibition of BCR-ABL kinase causes G(1) arrest, down-regulation of SKP2, and accumulation of p27. Ectopic expression of wild-type SKP2, but not a mutant unable to recognize p27, partially rescues cell-cycle progression. A similar regulation pattern is seen in cell lines transformed by FLT3-ITD, JAK2(V617F), and TEL-PDGFRbeta, suggesting that the SKP2/p27 conduit may be a universal target for leukemogenic tyrosine kinases. Mice that received transplants of BCR-ABL-infected SKP2(-/-) marrow developed a myeloproliferative syndrome but survival was significantly prolonged compared with recipients of BCR-ABL-expressing SKP2(+/+) marrow. SKP2(-/-) leukemic cells demonstrated higher levels of nuclear p27 than SKP2(+/+) counterparts, suggesting that the attenuation of leukemogenesis depends on increased p27 expression. Our data identify SKP2 as a crucial mediator of BCR-ABL-induced leukemogenesis and provide the first in vivo evidence that SKP2 promotes oncogenesis. Hence, stabilization of p27 by inhibiting its recognition by SCF(SKP2) may be therapeutically useful.


Assuntos
Genes abl , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Animais , Sequência de Bases , Transplante de Medula Óssea , Ciclo Celular , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Primers do DNA/genética , Proteínas de Fusão bcr-abl , Expressão Gênica , Leucemia Mielogênica Crônica BCR-ABL Positiva/etiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo
12.
Cancer Res ; 66(23): 11156-65, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17145859

RESUMO

The JAK2(V617F) mutation is present in almost all patients with polycythemia vera (PV), large proportions of patients with essential thrombocythemia and idiopathic myelofibrosis, and less frequently in atypical myeloproliferative disorders (MPD). We show that transplantation of JAK2(V617F)-transduced bone marrow into BALB/c mice induces MPD reminiscent of human PV, characterized by erythrocytosis, granulocytosis, extramedullary hematopoiesis, and bone marrow fibrosis, but not thrombocytosis. Fluorescence-activated cell sorting of bone marrow and spleen showed proportional expansion of common myeloid progenitors, granulocyte-monocyte and megakaryocyte-erythrocyte progenitors. Megakaryocyte and late erythroid progenitors were dramatically increased, with only modest expansion of early erythroid progenitors. Erythropoietin (Epo) receptor expression was reduced on early, but normal on late erythroblasts. Serum levels of Epo and granulocyte colony-stimulating factor, but not granulocyte macrophage colony-stimulating factor, were reduced, whereas tumor necrosis factor-alpha was increased, possibly exerting a negative effect on JAK2(V617F)-negative hematopoiesis. These data suggest that erythrocytosis and granulocytosis in JAK2(V617F) mice are the net result of a complex interplay between cell intrinsic and extrinsic factors. There were no thromboembolic events and no animals succumbed to their disease, implicating additional factors in the manifestation of human disease. The disease was not transplantable and prolonged observation showed normalization of blood counts in most JAK2(V617F) mice, suggesting that the mutation may not confer self-renewal capacity.


Assuntos
Transplante de Medula Óssea/métodos , Janus Quinase 2/genética , Mutação de Sentido Incorreto/genética , Transtornos Mieloproliferativos/patologia , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/efeitos adversos , Linhagem Celular , Células Clonais/metabolismo , Células Clonais/patologia , Eritropoetina/sangue , Fibrose , Fator Estimulador de Colônias de Granulócitos/sangue , Hematopoese Extramedular , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/genética , Policitemia/etiologia , Policitemia/metabolismo , Policitemia/patologia , Policitemia Vera/etiologia , Policitemia Vera/genética , Policitemia Vera/patologia , Receptores da Eritropoetina/metabolismo , Baço/metabolismo , Baço/patologia , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/sangue
13.
Mol Cell Biol ; 26(16): 6082-93, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16880519

RESUMO

Kinase domain (KD) mutations of Bcr-Abl interfering with imatinib binding are the major mechanism of acquired imatinib resistance in patients with Philadelphia chromosome-positive leukemia. Mutations of the ATP binding loop (p-loop) have been associated with a poor prognosis. We compared the transformation potency of five common KD mutants in various biological assays. Relative to unmutated (native) Bcr-Abl, the ATP binding loop mutants Y253F and E255K exhibited increased transformation potency, M351T and H396P were less potent, and the performance of T315I was assay dependent. The transformation potency of Y253F and M351T correlated with intrinsic Bcr-Abl kinase activity, whereas the kinase activity of E255K, H396P, and T315I did not correlate with transforming capabilities, suggesting that additional factors influence transformation potency. Analysis of the phosphotyrosine proteome by mass spectroscopy showed differential phosphorylation among the mutants, a finding consistent with altered substrate specificity and pathway activation. Mutations in the KD of Bcr-Abl influence kinase activity and signaling in a complex fashion, leading to gain- or loss-of-function variants. The drug resistance and transformation potency of mutants may determine the outcome of patients on therapy with Abl kinase inhibitors.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Proteínas de Fusão bcr-abl/metabolismo , Mutação/genética , Fosfotransferases/metabolismo , Piperazinas/farmacologia , Pirimidinas/farmacologia , Sequência de Aminoácidos , Animais , Benzamidas , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Modelos Animais de Doenças , Feminino , Proteínas de Fusão bcr-abl/química , Proteínas de Fusão bcr-abl/genética , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Células Progenitoras Mieloides/citologia , Fosfotirosina/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais , Especificidade por Substrato
14.
Haematologica ; 89(1): 49-57, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14754606

RESUMO

BACKGROUND AND OBJECTIVES: Imatinib is the new standard drug treatment for patients with chronic myelogenous leukemia (CML). Quantitative reverse transcription-polymerase chain reaction (qPCR) for detection of BCR-ABL transcripts is frequently used for monitoring patients in addition to or instead of conventional cytogenetics, although its place in the overall diagnostic framework is not yet clear. In this study, we compared qPCR and conventional cytogenetics for monitoring of patients during the early phases of imatinib therapy. DESIGN AND METHODS: One hundred and seventeen patients treated with imatinib for CML in chronic or accelerated phase were prospectively followed with qPCR and karyotyping. Comparisons were made between both methods and between qPCR results from bone marrow and peripheral blood. To determine the prognostic impact of qPCR and cytogenetics during the early phase of imatinib treatment on subsequent cytogenetic response and progression-free survival (PFS), a multivariate model was generated that included established prognostic baseline variables. RESULTS: We found a significant correlation between the proportion of Philadelphia (Ph) chromosome-positive metaphases and qPCR in the bone marrow and peripheral blood. Low qPCR values after 3 months of therapy were correlated with major cytogenetic response (MCyR) at 6 months and PFS at 2 years. However, in multivariate analysis, the cytogenetic response at 3 months emerged as the only independent parameter predictive of MCyR at 6 months and PFS at 2 years. INTERPRETATION AND CONCLUSIONS: Our data suggest that conventional karyotyping should remain the standard method for following patients on imatinib during the early phases of therapy.


Assuntos
Análise Citogenética/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Adulto , Idoso , Benzamidas , Medula Óssea/química , Medula Óssea/metabolismo , Medula Óssea/patologia , Análise Citogenética/estatística & dados numéricos , Intervalo Livre de Doença , Feminino , Proteínas de Fusão bcr-abl/sangue , Proteínas de Fusão bcr-abl/genética , Gliceraldeído-3-Fosfato Desidrogenases/sangue , Gliceraldeído-3-Fosfato Desidrogenases/genética , Humanos , Mesilato de Imatinib , Hibridização in Situ Fluorescente/métodos , Hibridização in Situ Fluorescente/estatística & dados numéricos , Leucemia Mielogênica Crônica BCR-ABL Positiva/sangue , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Cromossomo Filadélfia , Prognóstico , Indução de Remissão/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/estatística & dados numéricos
15.
Blood ; 101(5): 1941-9, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12411298

RESUMO

Chronic myelogenous leukemia (CML) is characterized by the presence of a Bcr-Abl fusion protein with deregulated tyrosine kinase activity that is required for maintaining the malignant phenotype. Imatinib, a selective inhibitor of Bcr-Abl, induces major cytogenetic remission (MCR) or complete cytogenetic remission (CCR) in the majority of patients with CML in first chronic phase. However, thorough re-evaluation of cytogenetics in a cohort of patients in MCR or CCR demonstrated clonal karyotypic abnormalities in more than 10% of cases, some of which were clinically associated with a myelodysplastic syndrome (MDS). Further analysis identified previous exposure to cytarabine and idarubicin as significant risk factors for the subsequent occurrence of abnormalities in Philadelphia chromosome-negative (Ph-) cells. To investigate if cytogenetically normal but clonal hematopoiesis might be present in other patients in cytogenetic remission, we studied X-chromosome inactivation as a marker of clonality by polymerase chain reaction analysis of the human androgen receptor (HUMARA). We find that imatinib restores a polyclonal pattern in most patients in CCR and MCR. Nonetheless, our results are consistent with the notion that targeted therapy of CML with imatinib favors the manifestation of Ph- clonal disorders in some patients. They indicate that patients on imatinib should be followed with conventional cytogenetics, even after induction of CCR.


Assuntos
Antineoplásicos/uso terapêutico , Linfócitos B/patologia , Células Clonais/patologia , Inibidores Enzimáticos/uso terapêutico , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Hematopoese , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Adulto , Idoso , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzamidas , Células Sanguíneas/patologia , Células da Medula Óssea/patologia , Estudos de Coortes , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Mecanismo Genético de Compensação de Dose , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Idarubicina/administração & dosagem , Idarubicina/efeitos adversos , Mesilato de Imatinib , Cariotipagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/patologia , Cromossomo Filadélfia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Receptores Androgênicos/análise , Indução de Remissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...