Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Blood ; 117(9): 2691-9, 2011 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-21097671

RESUMO

Clinical observations and laboratory evidence link bone marrow failure in myelodysplastic syndrome (MDS) to a T cell-mediated immune process that is responsive to immunosuppressive treatment (IST) in some patients. Previously, we showed that trisomy 8 MDS patients had clonally expanded CD8(+) T-cell populations that recognized aneuploid hematopoietic progenitor cells (HPC). Furthermore, microarray analyses showed that Wilms tumor 1 (WT1) gene was overexpressed by trisomy 8 hematopoietic progenitor (CD34(+)) cells compared with CD34(+) cells from healthy donors. Here, we show that WT1 mRNA expression is up-regulated in the bone marrow mononuclear cells of MDS patients with trisomy 8 relative to healthy controls and non-trisomy 8 MDS; WT1 protein levels were also significantly elevated. In addition, using a combination of physical and functional assays to detect the presence and reactivity of specific T cells, respectively, we demonstrate that IST-responsive MDS patients exhibit significant CD4(+) and CD8(+) T-cell responses directed against WT1. Finally, WT1-specific CD8(+) T cells were present within expanded T-cell receptor Vß subfamilies and inhibited hematopoiesis when added to autologous patient bone marrow cells in culture. Thus, our results suggest that WT1 is one of the antigens that triggers T cell-mediated myelosuppression in MDS.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Terapia de Imunossupressão , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/terapia , Proteínas WT1/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Estudos de Casos e Controles , Cromossomos Humanos Par 8/genética , Cromossomos Humanos Par 8/imunologia , Regulação da Expressão Gênica , Antígenos HLA-A/química , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Epitopos Imunodominantes/imunologia , Estrutura Quaternária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Trissomia/genética , Trissomia/imunologia , Proteínas WT1/genética , Proteínas WT1/metabolismo
2.
Transfusion ; 52(3): 537-41, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21883270

RESUMO

BACKGROUND: Reports of Monosomy 7 in patients receiving granulocyte-colony-stimulating factor (G-CSF) have raised concerns that this cytokine may promote genomic instability. However, there are no studies addressing whether repeated administration of G-CSF produces Monosomy 7 aneuploidy in healthy donors. STUDY DESIGN AND METHODS: We examined Chromosomes 7 and 8 by fluorescent in situ hybridization (FISH) in CD34+ cells from 35 healthy hematopoietic stem cell transplant (HSCT) donors after G-CSF administration for 5 days and by spectral karyotyping analysis (SKY) in four individuals to assess chromosomal integrity. We also studied 38 granulocyte donors who received up to 42 doses of G-CSF and dexamethasone (Dex) using FISH for Chromosomes 7 and 8. RESULTS: We found no abnormalities in Chromosomes 7 and 8 in G-CSF-mobilized CD34+ cells when assessed by FISH or SKY, nor did we detect aneuploidy in G-CSF- and Dex-treated donors. CONCLUSION: G-CSF does not promote clinically detectable Monosomy 7 or Trisomy 8 aneuploidy in HSCT or granulocyte donors. These findings should be reassuring to healthy HSCT and granulocyte donors.


Assuntos
Instabilidade Genômica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Granulócitos/efeitos dos fármacos , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Aneuploidia , Deleção Cromossômica , Cromossomos Humanos Par 7/efeitos dos fármacos , Dexametasona/administração & dosagem , Glucocorticoides/administração & dosagem , Granulócitos/fisiologia , Granulócitos/transplante , Mobilização de Células-Tronco Hematopoéticas/efeitos adversos , Mobilização de Células-Tronco Hematopoéticas/normas , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Doadores de Tecidos
3.
Blood ; 113(4): 875-82, 2009 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-18922853

RESUMO

Primitive quiescent CD34(+) chronic myeloid leukemia (CML) cells are more biologically resistant to tyrosine kinase inhibitors than their cycling counterparts; however, graft-versus-leukemia (GVL) effects after allogeneic stem cell transplantation (SCT) probably eliminate even these quiescent cells in long-term surviving CML transplant recipients. We studied the progeny of CD34(+) cells from CML patients before SCT, which were cultured 4 days in serum-free media with hematopoietic growth factors. BCR-ABL expression was similar in both cycling and quiescent noncycling CD34(+) populations. Quiescent CD34(+) cells from CML patients were less susceptible than their cycling CD34(+) and CD34(-) counterparts to lysis by natural killer (NK) cells from their HLA-identical sibling donors. Compared with cycling populations, quiescent CD34(+) CML cells had higher surface expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors DR4 and DR5. Bortezomib up-regulated TRAIL receptor expression on quiescent CD34(+) CML cells, and further enhanced their susceptibility to cytotoxicity by in vitro expanded donor NK cells. These results suggest that donor-derived NK cell-mediated GVL effects may be improved by sensitizing residual quiescent CML cells to NK-cell cytotoxicity after SCT. Such treatment, as an adjunct to donor lymphocyte infusions and pharmacologic therapy, may reduce the risk of relapse in CML patients who require treatment by SCT.


Assuntos
Antígenos CD34/metabolismo , Ácidos Borônicos/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Pirazinas/farmacologia , Adolescente , Adulto , Bortezomib , Separação Celular , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Predisposição Genética para Doença/genética , Genótipo , Humanos , Células Matadoras Naturais/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Masculino , Pessoa de Meia-Idade , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Receptores de Morte Celular/metabolismo , Regulação para Cima/efeitos dos fármacos
4.
JAMA ; 305(8): 814-9, 2011 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-21343581

RESUMO

Myelodysplastic syndromes (MDS) are a heterogeneous group of bone marrow disorders characterized by ineffective hematopoiesis and a tendency to develop leukemia. In some patients, laboratory and clinical evidence supports a role for the immune system in the pathogenesis of early MDS. Many younger patients who respond to immunosuppressive therapy with drugs such as antithymocyte globulin and cyclosporine have clonal expansions of cytotoxic CD8(+) T cells that suppress normal hematopoiesis, as well as expansion of CD4(+) helper T-cell subsets that promote and sustain autoimmunity. Immunosuppressive therapy can produce hematologic responses in some patients and may improve survival and halt leukemic progression. In this report, we describe a 56-year-old woman who presented with fatigue and easy bruising, eventually became pancytopenic, and was diagnosed with MDS. After treatment with a clinical protocol using alemtuzumab, an anti-CD52 antibody, her blood cell counts returned to normal and she has remained in complete remission for more than 2 years of follow-up. In this article, we review the pathobiology of immune dysregulation in MDS and summarize the role of immunosuppressive therapy in MDS.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Sistema Imunitário/fisiopatologia , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/imunologia , Alemtuzumab , Anticorpos Monoclonais Humanizados , Feminino , Humanos , Imunossupressores/uso terapêutico , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/epidemiologia , Prognóstico , Resultado do Tratamento
5.
Biol Blood Marrow Transplant ; 16(12): 1665-73, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20659573

RESUMO

Graft-versus-host disease (GVHD) is a major risk factor for secondary malignancy after hematopoietic stem cell transplantation. Squamous cell carcinoma (SCC) of the skin and mucous membranes are especially frequent in this setting where aneuploidy and tetraploidy are associated with aggressive disease. The current study is directed at the mechanism of neoplasia in this setting. Unmanipulated keratinocytes from areas of oral GVHD in 9 patients showed tetraploidy in 10% to 46% of cells when examined by florescein in situ hybridization (FISH). Keratinocytes isolated from biopsy sites of GVHD but not from normal tissue showed even greater numbers of tetraploid cells (mean = 78%, range: 15%-85%; N = 9) after culture. To mimic the inflammatory process in GVHD, allogeneic HLA-mismatched lymphocytes were mixed with normal keratinocytes. After 2 weeks, substantial numbers of aneuploid and tetraploid cells were evident in cultures with lymphocytes and with purified CD8 but not CD4 cells. Telomere length was substantially decreased in the lymphocyte-treated sample. No mutations were present in the p53 gene, although haploinsufficiency for p53 due to the loss of chromosome 17 was common in cells exposed to lymphocytes. These findings suggest that in GVHD, inflammation and repeated cell division correlate with the development of karyotypic abnormalities.


Assuntos
Aberrações Cromossômicas , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Queratinócitos/patologia , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Deleção Cromossômica , Cromossomos Humanos Par 17 , Técnicas de Cocultura , Feminino , Genes p53 , Doença Enxerto-Hospedeiro/patologia , Antígenos HLA/imunologia , Humanos , Queratinócitos/imunologia , Leucemia/genética , Leucemia/patologia , Leucemia/cirurgia , Linfócitos/citologia , Linfócitos/imunologia , Masculino , Pessoa de Meia-Idade , Ploidias , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Síndrome de Smith-Magenis , Telômero/genética , Adulto Jovem
6.
Haematologica ; 95(3): 382-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20207845

RESUMO

BACKGROUND: Pure red cell aplasia and moderate aplastic anemia are marrow failure states with an immune pathogenesis. Previously, we described short-term improvements in blood counts in two pilot studies treating moderate aplastic anemia (mAA) and pure red cell aplasia (PRCA) patients with daclizumab, a humanized monoclonal antibody to the interleukin-2 receptor; we now report our long-term experience with a larger cohort of patients. DESIGN AND METHODS: After a median follow-up period of 4.8 years, 19 of 45 (42%) evaluable mAA patients and 10 of 26 (38%) patients with PRCA responded by three months and 2 additional mAA patients responded by six months following administration of the drug. RESULTS: Seven of 28 (25%) mAA patients achieved long-term packed red blood cell PRBC transfusion independence, and all PRCA responders achieved long-term transfusion PRBC transfusion independence. CONCLUSIONS: Red cell transfusion-independence prior to treatment in mAA patients predicted response. The only significant adverse treatment-related events were transient rashes and arthralgias. Daclizumab is safe and effective, and produces lengthy remissions in patients with PRCA and mAA.


Assuntos
Anemia Aplástica/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Imunoglobulina G/uso terapêutico , Imunossupressores/uso terapêutico , Aplasia Pura de Série Vermelha/tratamento farmacológico , Anticorpos Monoclonais Humanizados , Daclizumabe , Seguimentos , Humanos , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Segurança , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento
7.
JAMA ; 304(12): 1358-64, 2010 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-20858879

RESUMO

CONTEXT: Critically short telomeres produce apoptosis, cell senescence, and chromosomal instability in tissue culture and animal models. Variations in telomere length have been reported in severe aplastic anemia but their clinical significance is unknown. OBJECTIVE: To investigate the relationship between telomere length and clinical outcomes in severe aplastic anemia. DESIGN, SETTING, AND PATIENTS: Single institution analysis of 183 patients with severe aplastic anemia who were treated in sequential prospective protocols at the National Institutes of Health from 2000 to 2008. The pretreatment leukocyte age-adjusted telomere length of patients with severe aplastic anemia consecutively enrolled in immunosuppression protocols with antithymocyte globulin plus cyclosporine for correlation with clinical outcomes were analyzed. MAIN OUTCOME MEASURES: Hematologic response, relapse, clonal evolution, and survival. RESULTS: There was no relationship between hematologic response and telomere length with response rates of 56.5% of 46 patients in the first, 54.3% of 46 in the second, 60% of 45 in the third, and 56.5% of 46 in the fourth quartiles. Multivariate analysis demonstrated that telomere length was associated with relapse, clonal evolution, and mortality. Evaluated as a continuous variable, telomere length inversely correlated with the probability of hematologic relapse (hazard ratio [HR], 0.16; 95% confidence interval [CI], 0.03-0.69; P = .01). The probability of clonal evolution was higher in patients in the first quartile (24.5%; 95% CI, 8.7%-37.5%) than in quartiles 2 through 4 (8.4%; 95% CI, 3.2%-13.3%; P = .009), and evolution to monosomy 7 or complex cytogenetics was more common in the first quartile (18.8%; 95% CI, 3.5%-31.6%) [corrected] than in quartiles 2 through 4 (4.5%; 95% CI, 0.5%-8.2%; P = .002) [corrected]. Survival between these 2 groups differed, with 66% (95% CI, 52.9%-82.5%) surviving 6 years in the first quartile compared with 83.8% (95% CI, 77.3%-90.9%) in quartiles 2 through 4 (P = .008). CONCLUSION: In a cohort of patients with severe aplastic anemia receiving immunosuppressive therapy, telomere length was unrelated to response but was associated with risk of relapse, clonal evolution, and overall survival.


Assuntos
Anemia Aplástica/genética , Transformação Celular Neoplásica , Instabilidade Cromossômica , Telômero/ultraestrutura , Adulto , Anemia Aplástica/sangue , Anemia Aplástica/tratamento farmacológico , Anemia Aplástica/patologia , Estudos de Coortes , Feminino , Humanos , Imunossupressores/uso terapêutico , Leucócitos/ultraestrutura , Masculino , Prognóstico , Recidiva , Análise de Sobrevida , Resultado do Tratamento
8.
Haematologica ; 94(3): 348-54, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19181786

RESUMO

BACKGROUND: We hypothesized that the addition of sirolimus to standard horse antithymocyte globulin (h-ATG) and cyclosporine (CsA) would improve response rates in severe aplastic anemia, due to its complementary and synergistic properties to cyclosporine A. DESIGN AND METHODS: To test this hypothesis, we conducted a prospective randomized study comparing hATG/CsA/sirolimus to standard h-ATG/CsA. A total of 77 patients were treated from June 2003 to November 2005; 35 received h-ATG/CsA/sirolimus and 42 h-ATG/CsA. The two groups were well matched demographically and in blood counts prior to therapy. The primary end-point was hematologic response rate at 3 months, defined as no longer meeting the criteria for severe aplastic anemia. The study was powered to show a superior hematologic response rate of h-ATG/CsA/sirolimus compared to standard h-ATG/CsA. RESULTS: The overall response rate at 3 months was 37% for h-ATG/CsA/sirolimus and 50% for h-ATG/CsA and at 6 months 51% for h-ATG/CsA/sirolimus and 62% for h-ATG/CsA. After a planned interim analysis of 30 evaluable patients in each arm, accrual to the h-ATG/CsA/sirolimus arm was closed, as the conditional power for rejecting the null hypothesis was less than 1%. The rate of relapse, clonal evolution, and survival (secondary outcomes) did not differ significantly between patients treated with the two different regimens. CONCLUSIONS: Despite a theoretical rationale for its use, sirolimus did not improve the response rate in patients with severe aplastic anemia when compared to standard h-ATG/CsA.


Assuntos
Anemia Aplástica/tratamento farmacológico , Soro Antilinfocitário/uso terapêutico , Ciclosporina/uso terapêutico , Sirolimo/uso terapêutico , Adolescente , Adulto , Idoso , Anemia Aplástica/patologia , Animais , Criança , Pré-Escolar , Quimioterapia Combinada , Feminino , Cavalos , Humanos , Imunossupressores/uso terapêutico , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Estudos Prospectivos , Recidiva , Resultado do Tratamento , Adulto Jovem
9.
Exp Hematol ; 36(12): 1616-24, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18954937

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired genetic disorder of the bone marrow that produces intravascular hemolysis, proclivity to venous thrombosis, and hematopoietic failure. Mutation in the PIG-A gene of a hematopoietic stem cell abrogates synthesis of glycosylphosphoinositol (GPI) anchors and expression of all GPI-anchored proteins on the surface of progeny erythrocytes, leukocytes, and platelets. Urokinase plasminogen activator receptor (uPAR), a GPI-linked protein expressed on neutrophils, mediates endogenous thrombolysis through a urokinase-dependent mechanism. Here we show that membrane GPI-anchored uPAR is decreased or absent on granulocytes and platelets of patients with PNH, while soluble uPAR (suPAR) levels are increased in patients' plasma. Serum suPAR concentrations correlated with the number of GPI-negative neutrophils and were highest in patients who later develop thrombosis. In vitro, suPAR is released from PNH hematopoietic cells and from platelets upon activation, suggesting that these cells are the probable source of plasma suPAR in the absence of GPI anchor synthesis and trafficking of uPAR to the cell membrane. In vitro, the addition of recombinant suPAR results in a dose-dependent decrease in the activity of single-chain urokinase. We hypothesized that suPAR, prevents the interaction of urokinase with membrane-anchored uPAR on residual normal cells.


Assuntos
Fibrinolisina/análise , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Trombose/sangue , Ativador de Plasminogênio Tipo Uroquinase/sangue , Células Sanguíneas/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Hematopoese/genética , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/genética , Hemólise/genética , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Transporte Proteico/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Solubilidade , Trombose/etiologia , Trombose/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
10.
Semin Hematol ; 45(1): 39-48, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18179968

RESUMO

Patients with myelodysplastic syndrome (MDS) have intrinsic, usually acquired genetic defects in their hematopoietic stem cells, but some others exhibit T-cell-mediated inhibition of hematopoiesis and good responses to immunosuppression. In these cases, MDS shares a similar pathophysiology with aplastic anemia (AA). Here, we review the evidence supporting a role of the immune system in the pathophysiology of MDS and the results of clinical trials of immunosuppressive agents.


Assuntos
Sistema Imunitário/fisiopatologia , Imunossupressores/uso terapêutico , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/terapia , Soro Antilinfocitário/uso terapêutico , Ciclosporina/uso terapêutico , Humanos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Vacinas/imunologia
11.
Exp Hematol ; 35(12): 1777-81, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17697745

RESUMO

OBJECTIVE: To study if telomere length can be used as a surrogate marker for the mitotic history in normal and affected hematopoietic cells from patients with paroxysmal nocturnal hemoglobinuria (PNH). METHODS: The telomere length was measured by automated multicolor flow fluorescence in situ hybridization in glycosyl-phosphatidyl-inositol anchored protein (GPI)-negative and GPI-positive peripheral blood leukocytes. Eleven patients were studied, two with predominantly hemolytic PNH and nine with PNH associated with marrow failure. RESULTS: Telomere length in GPI-negative cells was significantly shorter than in GPI-positive cells of the same patient (p < 0.01, n = 11). The difference in telomere length (telomere length in GPI-positive minus telomere length in GPI-negative cells) correlated with the percentage of GPI-negative white blood cells. CONCLUSION: Our results support the hypothesis that telomere length is correlated to the replicative history of GPI-positive and GPI-negative cells and warrant further studies of telomere length in relation to disease progression in PNH.


Assuntos
Hemoglobinúria Paroxística/genética , Telômero , Citometria de Fluxo , Humanos
12.
Ann Intern Med ; 144(3): 181-5, 2006 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-16461962

RESUMO

BACKGROUND: Pure red-cell aplasia (PRCA) is a rare hematologic disease characterized by anemia, reticulocytopenia, and absence of bone marrow erythroid precursors. Most patients respond to some form of immunosuppressive treatment, but few prospective clinical trials have been performed. OBJECTIVE: To examine the efficacy of a humanized monoclonal antibody to the interleukin-2 receptor (daclizumab) for treating PRCA. DESIGN: Pilot study. SETTING: Federal government research hospital. PATIENTS: 15 patients with idiopathic PRCA. INTERVENTION: Daclizumab, 1 mg/kg of body weight, every 2 weeks for a total of 5 infusions. Pure red-cell aplasia was defined as transfusion-dependent anemia with a reticulocyte count of 60 x 10(9) cells/L or less and bone marrow showing absent or diminished erythroid precursors. MEASUREMENTS: Response to therapy was assessed by transfusion independence, increments in reticulocyte count, and nontransfused hemoglobin. RESULTS: Daclizumab had little toxicity. Of the 15 patients, 6 patients (40%) responded to treatment. All responders became transfusion-independent and achieved normal or near-normal hemoglobin values and normal reticulocyte counts. LIMITATIONS: The study was unblinded, and the number of patients was too small to assess drug safety reliably. CONCLUSIONS: Daclizumab seems safe. Its efficacy in this pilot protocol suggests that expanded study in PRCA and other bone marrow failure syndromes is warranted.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Imunoglobulina G/efeitos adversos , Imunoglobulina G/uso terapêutico , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Receptores de Interleucina-2/antagonistas & inibidores , Aplasia Pura de Série Vermelha/tratamento farmacológico , Adolescente , Idoso , Anticorpos Monoclonais Humanizados , Criança , Daclizumabe , Exantema/induzido quimicamente , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-2 , Masculino , Pessoa de Meia-Idade , Projetos Piloto
13.
Leuk Res ; 30(5): 643-7, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16226311

RESUMO

We examined expression profiles of hematopoietic tissue-specific microRNAs (miRNAs; miR-142, miR-155, miR-181 and miR-223) in 17 commercially available malignant hematopoietic cell lines and compared to those in highly purified normal human B, T, monocytic and granulocytic lineages. Although malignant cell lines examined showed miRNA expression patterns similar to normal human hematopoietic lineages, the levels of miRNA expression among cell lines and normal cell lineages were considerably different, indicating the significance of miRNAs in human hematopoietic diseases. Further our results showed differences in miRNA expression between mouse and human hematopoietic cells, suggesting important regulatory roles of miRNAs in human hematopoiesis and oncogenesis.


Assuntos
Linfócitos B/metabolismo , Perfilação da Expressão Gênica , Granulócitos/metabolismo , MicroRNAs/biossíntese , Monócitos/metabolismo , Linfócitos T/metabolismo , Animais , Linhagem Celular , Linhagem da Célula , Separação Celular , Citometria de Fluxo , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/isolamento & purificação , Especificidade da Espécie
14.
Exp Hematol ; 30(8): 943-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12160846

RESUMO

OBJECTIVE: Recent studies suggest that primitive bone marrow-derived cells contribute to regeneration of many tissues, including muscle, endothelium, myocardium, neural tissues, liver, and skin. Conversely, primitive cells resident in muscle and other tissues have been reported to reconstitute hematopoiesis. We investigated the contribution of cells with a primitive hematopoietic phenotype to human epidermal skin formation in recipients of allogeneic mobilized peripheral blood hematopoietic stem cell (HSC) transplantation. PATIENTS AND METHODS: Our study population included female patients who had received granulocyte colony-stimulating factor mobilized peripheral blood HSC transplants from male donors for a variety of benign and malignant hematologic disorders at least 6 months before study entry, with a history of skin graft-vs-host disease. Epidermal skin cells (keratinocytes) obtained from punch biopsies of the skin were cultured under conditions specific for growth and expansion of homogenous populations of keratinocytes from keratinocyte stem cells. After multiple passages, DNA was extracted from cultured cells and evaluated by two different polymerase chain reaction (PCR) method for detection of Y chromosome specific sequences. RESULTS: Neither sensitive PCR-based technique revealed the presence of male donor-derived keratinocyte stem cells in keratinocytes cultured from skin biopsies of female allogeneic transplantation recipients. CONCLUSIONS: We could not confirm the contribution of donor mobilized peripheral blood hematopoietic stem cells to keratinocyte stem cell populations after HSC transplantation. These results cannot explain the presence of donor-derived cells with keratinocyte phenotypic markers in tissue sections of HSC transplant recipients.


Assuntos
Células Epidérmicas , Sobrevivência de Enxerto , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Queratinócitos/citologia , Células-Tronco/citologia , Transplante Homólogo , Adulto , Amelogenina , Biópsia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Proteínas do Esmalte Dentário/genética , Feminino , Seguimentos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Hibridização in Situ Fluorescente , Antígenos Comuns de Leucócito/análise , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , Especificidade de Órgãos , Doadores de Tecidos , Quimeras de Transplante , Cromossomo X/genética , Cromossomo Y/genética
16.
Leuk Res ; 36(8): 982-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22524974

RESUMO

BACKGROUND: We previously demonstrated upregulation of c-myc, survivin, and cyclin D1 in CD34+ bone marrow mononuclear cells (BMMNCs) of patients with trisomy 8 and monosomy 7 myelodysplastic syndromes (MDS). "Knockdown" of cyclin D1 by RNA interference decreased trisomy 8 cell growth, suggesting that this might be a therapeutic target in MDS. EXPERIMENTAL DESIGN: We performed preclinical studies using BMMNCs from patients with MDS and AML to examine the effects of the styryl sulfone ON 01910.Na on cyclin D1 accumulation, aneuploidy, and CD34+ blast percentage. We next treated twelve patients with higher risk MDS and two trisomy 8 AML patients with ON 01910.Na on a phase I clinical protocol (NCT00533416). RESULTS: ON 01910.Na inhibited cyclin D1 expression, and was selectively toxic to trisomy 8 cells in vitro. Flow cytometry studies demonstrated increased mature CD15+ myeloid cells and decreased CD34+ blasts. Three patients treated with ON 01910.Na on a clinical had decreased bone marrow blasts by ≥ 50%, and three patients had hematologic improvements, one of which was sustained for 33 months. Patients with hematologic responses to ON 01910.Na had decreased cyclin D1 expression in their CD34+ cells. CONCLUSIONS: The preclinical results and responses of patients on a clinical trial warrant further investigation of ON 01910.Na as a potential novel targeted therapy for higher risk MDS patients.


Assuntos
Ciclina D1/antagonistas & inibidores , Glicina/análogos & derivados , Terapia de Alvo Molecular/métodos , Síndromes Mielodisplásicas/tratamento farmacológico , Sulfonas/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Cromossomos Humanos Par 8 , Relação Dose-Resposta a Droga , Feminino , Glicina/administração & dosagem , Glicina/efeitos adversos , Glicina/farmacologia , Glicina/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular/efeitos adversos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Sulfonas/administração & dosagem , Sulfonas/efeitos adversos , Sulfonas/farmacologia , Trissomia/patologia , Células Tumorais Cultivadas
18.
Genes Cancer ; 1(4): 331-45, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20717479

RESUMO

Here we report the discovery of ON044580, an α-benzoyl styryl benzyl sulfide that possesses potent inhibitory activity against two unrelated kinases, JAK2 and BCR-ABL, and exhibits cytotoxicity to human tumor cells derived from chronic myelogenous leukemia (CML) and myelodysplasia (MDS) patients or cells harboring a mutant JAK2 kinase. This novel spectrum of activity is explained by the non-ATP-competitive inhibition of JAK2 and BCR-ABL kinases. ON044580 inhibits mutant JAK2 kinase and the proliferation of JAK2(V617F)-positive leukemic cells and blocks the IL-3-mediated phosphorylation of JAK2 and STAT5. Interestingly, this compound also directly inhibits the kinase activity of both wild-type and imatinib-resistant (T315I) forms of the BCR-ABL kinase. Finally, ON044580 effectively induces apoptosis of imatinib-resistant CML patient cells. The apparently unrelated JAK2 and BCR-ABL kinases share a common substrate, STAT5, and such substrate competitive inhibitors represent an alternative therapeutic strategy for development of new inhibitors. The novel mechanism of kinase inhibition exhibited by ON044580 renders it effective against mutant forms of kinases such as the BCR-ABL(T315I) and JAK2(V617F). Importantly, ON044580 selectively reduces the number of aneuploid cells in primary bone marrow samples from monosomy 7 MDS patients, suggesting another regulatory cascade amenable to this agent in these aberrant cells. Data presented suggest that this compound could have multiple therapeutic applications including monosomy 7 MDS, imatinib-resistant CML, and myeloproliferative neoplasms that develop resistance to ATP-competitive agents.

19.
J Clin Oncol ; 28(35): 5166-73, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21041705

RESUMO

PURPOSE: Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis and progression to leukemia. Clinical and experimental evidence suggests an immune-mediated pathophysiology in some patients, in whom immunosuppressive therapy (IST) with horse antithymocyte globulin (h-ATG) and cyclosporine (CsA) can be effective. Because of the toxicities associated with h-ATG/CsA, we investigated an alternative regimen with alemtuzumab in MDS. PATIENTS AND METHODS: We conducted a nonrandomized, off-label, pilot, phase I/II study of alemtuzumab monotherapy in patients with MDS who were judged likely to respond to IST based on the following criteria: HLA-DR15-negative patients whose age plus the number of months of RBC transfusion dependence (RCTD) was less than 58; and HLA-DR15-positive patients whose age plus RCTD was less than 72. In total, 121 patients with MDS were screened, of whom 32 met eligibility criteria to receive alemtuzumab 10 mg/d intravenously for 10 days. Primary end points were hematologic responses at 3, 6, and 12 months after alemtuzumab. RESULTS: Seventeen (77%) of 22 evaluable intermediate-1 patients and four (57%) of seven evaluable intermediate-2 patients responded to treatment with a median time to response of 3 months. Four of seven evaluable responders with cytogenetic abnormalities before treatment had normal cytogenetics by 1 year after treatment. Five (56%) of nine responding patients evaluable at 12 months had normal blood counts, and seven (78%) of nine patients were transfusion independent. CONCLUSION: Alemtuzumab is safe and active in MDS and may be an attractive alternative to ATG in selected patients likely to respond to IST.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Síndromes Mielodisplásicas/tratamento farmacológico , Adulto , Idoso , Alemtuzumab , Anticorpos Monoclonais Humanizados , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/mortalidade , Uso Off-Label , Projetos Piloto , Resultado do Tratamento , Adulto Jovem
20.
Hematol Oncol Clin North Am ; 23(2): 347-60, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19327588

RESUMO

Myelodysplasia must be considered in the differential diagnosis of patients who have bone marrow failure, but bone marrow cellularity per se may not substantially affect either response to therapy or prognosis. It is unclear whether the primary pathophysiologic defect differs between hyper- and hypoplastic patients who have myelodysplasia. Cellularity does not seem to affect response to immunosuppressive therapy significantly and does not seem to be the major factor affecting improvements in response to lenalidomide, stem cell transplantation, or hematopoietic growth factors.


Assuntos
Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/tratamento farmacológico , Fatores Etários , Animais , Soro Antilinfocitário/uso terapêutico , Células da Medula Óssea/patologia , Ciclosporina/uso terapêutico , Humanos , Imunossupressores/uso terapêutico , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/fisiopatologia , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA