Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38778148

RESUMEN

Several studies reported that patients with acute myeloid leukemia (AML) who remain in long-term remission after allogeneic or autologous transplant have a shorter life expectancy, compared to the general population. However, little is known about the life expectancy of adult long-term survivors of AML who were treated with chemotherapy alone without a transplant and there have been no comparisons with survival among the general population. The current study indicates that the life expectancy of AML patients who achieved and maintained CR for at least 3 years is shorter than expected for age in the US population. This was observed also in patients who did not undergo a transplant including those who have not relapsed during the entire long follow-up period. Thus, late relapse does not explain why patients without transplants have a shortened life expectancy. Taken together, these data strongly suggest that prior chemotherapy for the underlying AML is at least a major contributing factor for the known shortened life expectancy post-transplant.

2.
Bone Marrow Transplant ; 52(12): 1592-1598, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28581459

RESUMEN

The introduction of the tyrosine kinase inhibitors (TKI) into the treatment of patients with Ph or BCR-ABL1-positive acute lymphoblastic leukemia has revolutionized the treatment of this poor prognosis acute leukemia. The combination of TKI with chemotherapy has improved response rates and allowed more patients to proceed to allogeneic hematopoietic cell transplant (alloHCT). Older patients have excellent responses to TKI and corticosteroids or in combination with minimal chemotherapy. This raises the question as to whether patients require full-intensity chemotherapy with TKI to achieve molecular remissions. The pediatricians have proposed that cure is achievable without alloHCT in children. These results have suggested that many patients may not require traditional chemotherapy in addition to TKI to achieve remission, and that patients who achieve a negative minimal residual disease state may not require alloHCT. The data in support of these questions is presented here and a suggested future clinical trial design based on these data is proposed.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adulto , Antineoplásicos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Neoplasia Residual , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto Joven
5.
Bone Marrow Transplant ; 47(10): 1332-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22307018

RESUMEN

Idiopathic Pneumonia Syndrome (IPS) is a common complication after allo-SCT and results in high mortality rates. Conventional treatment for IPS typically includes supportive care and high-dose corticosteroids (CS). Data suggests that TNF-α is important in the pathogenesis of IPS and that the TNF-α inhibitor etanercept may be useful for IPS treatment. We performed a retrospective comparison of consecutive patients treated at our center for IPS with CS only from 1999 to 2003 (group 1, n=22) or CS plus etanercept from 2004 to 2007 (group 2, n=17). In all, 18% of patients in group 1 vs 53% in group 2 were successfully taken off respiratory support and discharged from the hospital (P=0.039). OS was significantly better for recipients of CS plus etanercept (P=0.003). The estimated survival at 28 days and 2 years after IPS was 36.4% (95% CI 17-56%) and 9.1% (95% CI 2-25%) for group 1 and 88.2% (95% CI 61-97%) and 18% (95% CI 4-38%) for group 2, respectively. Our retrospective comparison suggests that the addition of etanercept to CS for IPS improves response rates and OS. However, outcomes remain limited in both groups, highlighting the need for more effective interventions to treat early and late complications of IPS.


Asunto(s)
Corticoesteroides/administración & dosificación , Antiinflamatorios no Esteroideos/administración & dosificación , Inmunoglobulina G/administración & dosificación , Neumonía/tratamiento farmacológico , Neumonía/mortalidad , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Trasplante de Células Madre , Adulto , Supervivencia sin Enfermedad , Etanercept , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/terapia , Humanos , Masculino , Persona de Mediana Edad , Neumonía/sangre , Neumonía/etiología , Estudios Retrospectivos , Tasa de Supervivencia , Síndrome , Factores de Tiempo , Trasplante Homólogo
6.
Bone Marrow Transplant ; 47(2): 203-11, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21441963

RESUMEN

Although reduced-intensity conditioning (RIC) and non-myeloablative (NMA)-conditioning regimens have been used for over a decade, their relative efficacy vs myeloablative (MA) approaches to allogeneic hematopoietic cell transplantation in patients with AML and myelodysplasia (MDS) is unknown. We compared disease status, donor, graft and recipient characteristics with outcomes of 3731 MA with 1448 RIC/NMA procedures performed at 217 centers between 1997 and 2004. The 5-year univariate probabilities and multivariate relative risk outcomes of relapse, TRM, disease-free survival (DFS) and OS are reported. Adjusted OS at 5 years was 34, 33 and 26% for MA, RIC and NMA transplants, respectively. NMA conditioning resulted in inferior DFS and OS, but there was no difference in DFS and OS between RIC and MA regimens. Late TRM negates early decreases in toxicity with RIC and NMA regimens. Our data suggest that higher regimen intensity may contribute to optimal survival in patients with AML/MDS, suggesting roles for both regimen intensity and graft vs leukemia in these diseases. Prospective studies comparing regimens are needed to confirm this finding and determine the optimal approach to patients who are eligible for either MA or RIC/NMA conditioning.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Mieloide Aguda/cirugía , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/cirugía , Trasplante Homólogo , Resultado del Tratamiento , Adulto Joven
7.
Bone Marrow Transplant ; 46(3): 430-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20498647

RESUMEN

Treatment options for steroid-refractory GVHD (SR-GVHD) are unsatisfactory and prognosis is poor. Inflammatory cytokines IL-2 and TNF-α are important mediators of GVHD and may be critical targets for therapy. We retrospectively reviewed our experience using combination anti-cytokine therapy of daclizumab and infliximab. Seventeen evaluable patients had a median age of 47 years (range 35-63). The conditioning regimen was myeloablative in 13 and non-myeloablative in 4 cases. GVHD occurred at a median of 49 days after transplant in 12 patients (range 21-231 days) and at a median of 46 days (range 25-119 days) after donor lymphocyte infusion in 5 patients. All patients had persistent or progressive GVHD despite 1-2 mg/kg/day of corticosteroids for a median of 7 days (range 2-26 days). They received a combination of daclizumab and infliximab for acute GVHD IBMTR severity index B (3), C (10) or D (4). Of the 17 patients analyzed, 47% responded to treatment, 24% had complete resolution of symptoms and 24% had partial responses. Survival was limited and all the patients died a median of 6.7 months (range 1.6-26) from transplant and 35 days from initiation of daclizumab/infliximab. This retrospective analysis suggests that combination anti-cytokine therapy with daclizumab/infliximab has significant activity in SR-GVHD, but outcomes remain poor. New methods to prevent and treat GVHD are urgently needed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Enfermedad Aguda , Adulto , Anticuerpos Monoclonales Humanizados , Daclizumab , Resistencia a Medicamentos , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inmunosupresores/uso terapéutico , Infliximab , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Esteroides/farmacología , Análisis de Supervivencia , Resultado del Tratamiento
8.
Bone Marrow Transplant ; 46(1): 59-63, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20421869

RESUMEN

Lenalidomide is an active treatment for multiple myeloma (MM) and is increasingly used as part of the initial treatment of this disease. Recent reports have suggested decreases in the number of CD34+ cells collected and increases in the failure rate among patients whose initial therapy contained lenalidomide when mobilized with G-CSF alone. A retrospective data analysis of 364 patients with MM who underwent stem cell mobilization and attempted harvest at the Hospital of the University of Pennsylvania between January 2002 and December 2007 was performed. Forty-three of the patients received lenalidomide in their induction regimen, and were mobilized with either CY and G-CSF or G-CSF alone. The number of apheresis cycles and the failure rate were lower, whereas the mean number of collected stem cells was higher in patients who were mobilized with CY and G-CSF in comparison with G-CSF alone. This suggests that lenalidomide does not prevent the harvest of adequate numbers of CD34 cells for autologous stem cell transplant, but mobilization with G-CSF and CY may be required to obtain adequate numbers of stem cells. Finally, in our study, the number of lenalidomide cycles did not correlate with stem cell yield.


Asunto(s)
Antineoplásicos/uso terapéutico , Ciclofosfamida/uso terapéutico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Movilización de Célula Madre Hematopoyética/métodos , Mieloma Múltiple/terapia , Trasplante de Células Madre de Sangre Periférica , Talidomida/análogos & derivados , Antígenos CD34/sangre , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Eliminación de Componentes Sanguíneos/estadística & datos numéricos , Quimioterapia Combinada , Humanos , Lenalidomida , Registros Médicos , Mieloma Múltiple/sangre , Inducción de Remisión/métodos , Estudios Retrospectivos , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talidomida/uso terapéutico , Acondicionamiento Pretrasplante/efectos adversos , Acondicionamiento Pretrasplante/métodos , Trasplante Autólogo , Resultado del Tratamiento
9.
Bone Marrow Transplant ; 43(5): 417-22, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18850013

RESUMEN

Therapeutic options for patients with multiple myeloma whose disease has relapsed after a prior auto-SCT include novel biologic therapies, traditional chemotherapy or a second transplant, with no clear standard of care. Few published studies address the safety and efficacy of a second auto-SCT for relapsed disease. We reviewed the Abramson Cancer Center experience with salvage auto-SCT for relapsed multiple myeloma. Forty-one patients had received a salvage auto-SCT at our institution; the median time between transplants was 37 months (range 3-91). The overall response rate in assessable patients was 55%, and treatment-related mortality was 7%. With a median follow-up time of 15 months, the median PFS was 8.5 months and the median overall survival (OS) was 20.7 months. In a multivariate analysis of OS, independent prognostic factors were >or=5 prior lines of therapy and time to progression after initial auto-SCT of

Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple/terapia , Terapia Recuperativa/métodos , Adulto , Anciano , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Pronóstico , Recurrencia , Trasplante Autólogo
10.
Bone Marrow Transplant ; 38(3): 211-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16770314

RESUMEN

We conducted a retrospective analysis of 50 lymphoma patients (Hodgkin's disease and non-Hodgkin's lymphoma) who had an 18F-fluoro-deoxyglucose positron emission tomography (FDG-PET) scan after at least two cycles of salvage chemotherapy and before autologous stem cell transplantation (ASCT) at our institution. The patients were categorized into FDG-PET negative (N = 32) and positive (N = 18) groups. The median follow-up after ASCT was 19 months (range: 3-59). In the FDG-PET-negative group, the median progression-free survival (PFS) was 19 months (range: 2-59) with 15 (54%) patients without progression at 12 months after ASCT. The median overall survival (OS) for this group was not reached. In the FDG-PET-positive group, the median PFS was 5 months (range: 1-19) with only one (7%) patient without progression at 12 months after ASCT. The median OS was 19 months (range: 1-34). In the FDG-PET-negative group, chemotherapy-resistant patients by CT-based criteria had a comparable outcome to those with chemotherapy-sensitive disease. A positive FDG-PET scan after salvage chemotherapy and prior ASCT indicates an extremely poor chance of durable response after ASCT.


Asunto(s)
Fluorodesoxiglucosa F18 , Enfermedad de Hodgkin/diagnóstico por imagen , Linfoma no Hodgkin/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Trasplante de Células Madre/métodos , Adulto , Anciano , Terapia Combinada/métodos , Femenino , Enfermedad de Hodgkin/terapia , Humanos , Linfoma no Hodgkin/terapia , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Terapia Recuperativa/métodos , Análisis de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
11.
Bone Marrow Transplant ; 37(11): 989-95, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16633362

RESUMEN

Long-term disease-free survival (DFS) has been reported after autologous stem cell transplantation for acute lymphoblastic leukemia. Phase II studies have evaluated its role in first and subsequent complete remission (CR) with DFS rates of up to 50%. It has been under-utilized in 1st CR in part, due to a concern that patients who relapse after autologous stem cell transplantation (ASCT) have fewer options for salvage treatment of relapsed disease. Unfortunately, survival rates of <5% are reported in patients who relapse, regardless of initial therapy. Few prospective, randomized trials have analyzed large enough numbers of patients to allow us to determine the appropriate patient population for autologous transplantation. Although variability in the available studies makes it difficult to draw a definite conclusion, and many issues remain unresolved, available data suggests that there may be a group of patients for whom ASCT in first remission is a reasonable and perhaps superior treatment choice. Factors such as risk features at diagnosis, and minimal residual disease following induction therapy greatly affect outcome following ASCT. The available data as well as the questions that remain to be answered will be discussed and reviewed.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Trasplante de Células Madre , Purgación de la Médula Ósea , Ensayos Clínicos como Asunto , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Neoplasia Residual , Selección de Paciente , Recurrencia , Inducción de Remisión , Trasplante Autólogo , Resultado del Tratamiento
12.
Bone Marrow Transplant ; 36(11): 955-61, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16205727

RESUMEN

Although follicular lymphoma (FL) is generally responsive to conventional-dose chemotherapy, improved survival in patients with this disease has been difficult to demonstrate. High-dose chemo/radiotherapy followed by autologous stem-cell transplantation (ASCT) can improve response rates, although its effects on survival remain controversial. Between 1990 and 2003, we transplanted 49 patients with low-grade FL at our institution. Twenty-two patients (45%) had undergone histologic transformation at the time of ASCT. In all, 44 patients (90%) had relapsed disease and five patients (10%) were resistant to chemotherapy at the time of transplantation. After ASCT, 30 patients (61%) were in complete remission (CR). The median overall survival (OS) has not been reached, while the median event-free survival (EFS) is 2.4 years. At a median follow-up of 5.5 years (longest 12.4 years), a plateau has been reached with 56% of patients remaining alive, and 35% event-free. ASCT was well tolerated except for two (4%) treatment-related deaths. In multivariable analysis, CR after ASCT and age less than 60 years are the best predictors of EFS and OS. ASCT is thus a safe therapeutic approach in FL, resulting in long-term EFS and OS for some patients, even with transformed disease.


Asunto(s)
Antineoplásicos/administración & dosificación , Trasplante de Células Madre Hematopoyéticas/métodos , Linfoma Folicular/terapia , Linfoma no Hodgkin/terapia , Sobrevivientes , Adulto , Factores de Edad , Anciano , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Linfoma Folicular/mortalidad , Linfoma Folicular/patología , Linfoma no Hodgkin/mortalidad , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad , Pronóstico , Inducción de Remisión , Estudios Retrospectivos , Análisis de Supervivencia , Trasplante Autólogo
13.
Bone Marrow Transplant ; 35(9): 921-6, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15765118

RESUMEN

Nonmyeloablative stem cell transplantation (NST) harnesses the graft-versus-tumor effect while minimizing regimen-related toxicity, and can result in donor chimerism and remission. Acute graft-versus-host disease (GVHD) and infections are major complications after sibling NST. Toxicity of unrelated-donor (UD) NST and the most appropriate GVHD prophylaxis in this setting remain poorly defined. We describe 25 patients who received UD-NST conditioned with fludarabine and cyclophosphamide. The first six patients received cyclosporine (Cs) and mycophenolate mofetil (MMF) (n=5) or methotrexate (MTX) (n=1) as GVHD prophylaxis (group 1) and all developed grade III-IV acute GVHD. The next 19 patients received the same conditioning regimen with the addition of alemtuzumab, and all received Cs/MTX post-transplant. Engraftment and donor chimerism were achieved in all but one evaluable patient. In all, 15 patients died: five of six deaths in group 1 were attributable to acute GVHD, while deaths in group 2 were due to infection or progressive disease (P=0.05). The combination of Cs/MMF is inadequate GVHD prophylaxis for UD-NST. The use of Cs, MTX, and alemtuzumab eliminated severe acute GVHD; its impact on response merits further study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Trastornos Linfoproliferativos/terapia , Ácido Micofenólico/análogos & derivados , Acondicionamiento Pretrasplante , Vidarabina/análogos & derivados , Adulto , Alemtuzumab , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Anticuerpos Antineoplásicos/administración & dosificación , Ciclofosfamida/administración & dosificación , Femenino , Enfermedad Injerto contra Huésped/complicaciones , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Trastornos Linfoproliferativos/complicaciones , Trastornos Linfoproliferativos/mortalidad , Masculino , Persona de Mediana Edad , Ácido Micofenólico/administración & dosificación , Quimera por Trasplante , Acondicionamiento Pretrasplante/métodos , Vidarabina/administración & dosificación
14.
Ann Hematol ; 81(7): 410-3, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12185517

RESUMEN

Nonmyeloablative allogeneic stem cell transplantation (NMASCT) can be used to exploit the graft-versus-tumor (GVT) potential of allogeneic donor cells in the setting of reduced conditioning regimen toxicity. This approach is particularly attractive for patients who have received extensive prior therapy and are poor candidates for traditional allogeneic stem cell transplantation. However, toxicity in heavily pretreated patients remains uncertain. Additional immunosuppression in already immunocompromised patients may result in unexpected toxicity. We report a case of probable progressive multifocal leukoencephalopathy (PML) responsive to interleukin-2 (IL-2) following a NMASCT in a 29-year-old woman with relapsed Hodgkin's lymphoma. The patient developed severe neurological symptoms approximately 6 weeks following NMASCT associated with low CD4+ cell counts and magnetic resonance imaging (MRI) was consistent with PML. IL-2 therapy resulted in increasing CD4+ counts and progressive resolution of neurological symptoms. Disruption of IL-2 therapy led to neurological deterioration, which responded to reinstitution of IL-2 therapy. The patient's lymphoma initially progressed following NMASCT, but has responded to donor leukocyte infusions (DLI). This case reiterates the potent GVT potential of NMASCT in patients with Hodgkin's disease. However, it demonstrates the potential for severe complications related to immunosuppression, especially in heavily pretreated patients. The toxicity after NMASCT should not be understated and will need to be explored further.


Asunto(s)
Enfermedad de Hodgkin/complicaciones , Enfermedad de Hodgkin/cirugía , Interleucina-2/uso terapéutico , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Leucoencefalopatía Multifocal Progresiva/etiología , Trasplante de Células Madre/efectos adversos , Adulto , Femenino , Enfermedad de Hodgkin/terapia , Humanos , Terapia de Inmunosupresión/efectos adversos , Transfusión de Leucocitos , Leucoencefalopatía Multifocal Progresiva/diagnóstico , Hepatopatías/diagnóstico por imagen , Hepatopatías/etiología , Imagen por Resonancia Magnética , Recurrencia Local de Neoplasia , Tomografía Computarizada por Rayos X , Trasplante Homólogo
15.
Bone Marrow Transplant ; 30(1): 23-8, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12105773

RESUMEN

Gemtuzumab ozogamicin (GO) (Mylotarg, CMA-676) is a novel chemotherapeutic agent consisting of an anti-CD33 monoclonal antibody linked to calicheamicin, and is associated with a 30% response rate in patients with CD33-positive acute myeloid leukemia (AML) in first relapse. GO therapy has a 20% incidence of grade 3 or 4 hepatotoxicity, and has recently been associated with hepatic veno-occlusive disease (VOD). The efficacy and toxicity of GO in patients with AML who have relapsed after hematopoietic stem cell transplant (HSCT) is unknown, as this population was largely excluded from phase II studies. We reviewed the outcomes of eight consecutive patients with AML who received GO following relapse after HSCT. Two (25%) had responses to GO. One patient, who had had two previous HSCT and prior hyperbilirubinemia, developed severe VOD and died 14 days after GO therapy. The other seven patients did not meet diagnostic criteria for VOD. We conclude that GO can be safe and effective in patients who relapse following HSCT, but that caution is warranted in patients with multiple risk factors for VOD.


Asunto(s)
Aminoglicósidos , Antibacterianos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide/tratamiento farmacológico , Enfermedad Aguda , Adolescente , Adulto , Antibacterianos/toxicidad , Anticuerpos Monoclonales/toxicidad , Anticuerpos Monoclonales Humanizados , Causas de Muerte , Femenino , Gemtuzumab , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Enfermedad Veno-Oclusiva Hepática/etiología , Humanos , Inmunotoxinas/administración & dosificación , Inmunotoxinas/toxicidad , Incidencia , Leucemia Mieloide/complicaciones , Leucemia Mieloide/terapia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Terapia Recuperativa , Equivalencia Terapéutica , Resultado del Tratamiento
16.
Blood ; 98(8): 2301-7, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11588023

RESUMEN

Human acute myelogenous leukemia (AML) is thought to arise from a rare population of malignant stem cells. Cells of this nature, herein referred to as leukemic stem cells (LSCs), have been documented for nearly all AML subtypes and appear to fulfill the criteria for stem cells in that they are self-renewing and give rise to the cells found in many leukemic populations. Because these cells are likely to be critical for the genesis and perpetuation of leukemic disease, the present studies sought to characterize unique molecular properties of the LSC population, with particular emphasis on the transcription factor, nuclear factor-kappaB (NF-kappaB). Previous experiments have shown that unstimulated human CD34(+) progenitor cells do not express NF-kappaB. In contrast, primary AML CD34(+) cells display readily detectable NF-kappaB activity as assessed by electrophoretic mobility shift assay and gene expression studies. Furthermore, detailed analyses of enriched AML stem cells (CD34(+)/CD38(-)/CD123(+)) indicate that NF-kappaB is also active in the LSC population. Given the expression of NF-kappaB in leukemic, but not normal primitive cells, the hypothesis that inhibition of NF-kappaB might induce leukemia-specific apoptosis was tested by treating primary cells with the proteasome inhibitor MG-132, a well-known inhibitor of NF-kappaB. Leukemic CD34(+)/CD38(-) cells displayed a rapid induction of cell death in response to MG-132, whereas normal CD34(+)/CD38(-) cells showed little if any effect. Taken together, these data indicate that primitive AML cells aberrantly express NF-kappaB and that the presence of this factor may provide unique opportunities to preferentially ablate LSCs.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide Aguda/sangre , FN-kappa B/sangre , Células Madre/fisiología , Actinas/genética , Antígenos CD/análisis , Antígenos CD34/análisis , Células de la Médula Ósea/citología , Ciclo Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Leupeptinas/farmacología , FN-kappa B/antagonistas & inhibidores , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
17.
Blood ; 98(2): 266-71, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11435292

RESUMEN

Arsenic trioxide has been shown to be effective in treating acute promyelocytic leukemia (APL), with minimal overall toxicity reported to date. A phase I/II study was initiated in June 1998 using arsenic trioxide for relapsed APL to determine the maximum tolerated or minimal effective dose and to determine the efficacy of treatment at that dose. Ten patients received 1 to 4 monthly cycles of treatment with 0.1 mg/kg per day intravenous arsenic trioxide. Six of 7 patients evaluable for response achieved cytogenetic or molecular complete remission. However, 3 patients died suddenly during the first cycle of treatment. Autopsies obtained on 2 of these failed to identify a cause of sudden death, despite evidence of pulmonary hemorrhage in one. A third patient, for whom an autopsy was not performed, became asystolic and died while on continuous cardiac telemetry. These observations suggest that arsenic trioxide may be significantly or even fatally toxic at doses currently used and that caution is warranted in its use.


Asunto(s)
Antineoplásicos/efectos adversos , Arsenicales/efectos adversos , Muerte Súbita , Leucemia Promielocítica Aguda/tratamiento farmacológico , Óxidos/efectos adversos , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Trióxido de Arsénico , Arsenicales/administración & dosificación , Arsenicales/farmacocinética , Niño , Femenino , Humanos , Cinética , Masculino , Persona de Mediana Edad , Óxidos/administración & dosificación , Óxidos/farmacocinética , Recurrencia
18.
Biol Blood Marrow Transplant ; 7(4): 230-8, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11349810

RESUMEN

Allogeneic donor leukocytes can be used after nonmyeloablative conditioning to exploit their graft-versus-tumor (GVT) activity in the setting of reduced conditioning-regimen toxicity. This approach may be particularly useful for patients who relapse after autologous stem cell transplantation (SCT). However, GVT activity, toxicity, and ability to establish mixed chimerism may differ in patients who were heavily pretreated prior to SCT compared with patients treated earlier in the course of their disease. We have performed a series of studies of nonmyeloablative allogeneic transplantation and present data on the subset of 14 patients treated for relapse after autologous SCT: 4 patients received no conditioning and unstimulated donor leukocyte infusions (DLI), 10 patients received conditioning with fludarabine and cyclophosphamide followed by unstimulated or granulocyte-colony-stimulating factor (G-CSF)-stimulated allogeneic peripheral blood stem cells (PBSCs), 4 patients received no graft-versus-host disease (GVHD) prophylaxis, and 10 patients received cyclosporine GVHD prophylaxis. All but 1 patient had sustained donor chimerism at least 30 days after allogeneic cell therapy (ACT), and 8 patients had more than 80% donor chimerism after ACT. Acute GVHD developed in 11 patients (grade III-IV, n = 6). Aplasia was more frequent in the patients receiving unstimulated PBSCs, despite the development of mixed chimerism. There were 6 complete responses and 4 partial responses; response was independent of conditioning and growth-factor stimulation of the donor graft. Five patients died of treatment-related causes and 4 patients died from progressive disease. Four patients remained alive 27 to 194 weeks (median, 66 weeks) after ACT. Prior autologous SCT may define a subset of patients at particularly high risk for GVHD and other toxicity after ACT. However, these data show that ACT with either DLI or G-CSF-stimulated blood cells results in direct GVT activity in some patients with Hodgkin's disease, myeloma, and non-Hodgkin's lymphoma, even after relapse from autologous SCT. Most patients developed donor chimerism with minimal conditioning. Alternative prophylactic regimens that control GVHD while maintaining GVT are needed to improve outcomes in these heavily pretreated patients.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Terapia Recuperativa , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Quimera , Terapia Combinada , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Tumor , Factor Estimulante de Colonias de Granulocitos/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/terapia , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/uso terapéutico , Infecciones/etiología , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Acondicionamiento Pretrasplante/mortalidad , Trasplante Autólogo , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/mortalidad , Vidarabina/efectos adversos , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico
19.
Blood ; 97(7): 2177-9, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11264190

RESUMEN

Previous studies indicate that human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells. Cells of this nature can initiate and maintain leukemic cell growth in both long-term cultures and nonobese diabetic/severe combined immune-deficient mice. To characterize the biology of primitive AML cells, gene expression screens were performed with 7 primary AML and 3 normal specimens. For each sample, stem cell populations (CD34(+)/CD38(-)) were isolated and used to synthesize radiolabeled complementary DNA (cDNA). AML vs normal probes were then hybridized to cDNA arrays containing genes related to cancer and apoptosis. Of approximately 1400 genes analyzed, 2 tumor-suppressor genes were identified that were overexpressed in all 7 of the AML CD34(+)/CD38(-) cell populations: death-associated protein kinase and interferon regulatory factor 1. Expression of each gene was confirmed by reverse-transcription polymerase chain reaction and immunoblot analysis. It is proposed that tumor-suppressor proteins play a role in the biology of primitive AML cells. (Blood. 2001;97:2177-2179)


Asunto(s)
Antígenos CD , Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Regulación Leucémica de la Expresión Génica , Genes Supresores de Tumor , Leucemia Mieloide/enzimología , Proteínas de Neoplasias/biosíntesis , Fosfoproteínas/biosíntesis , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Enfermedad Aguda , Antígenos CD34/análisis , Antígenos de Diferenciación/análisis , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas de Unión al ADN/genética , Proteínas Quinasas Asociadas a Muerte Celular , Inducción Enzimática , Células Madre Hematopoyéticas/enzimología , Humanos , Inmunofenotipificación , Factor 1 Regulador del Interferón , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Glicoproteínas de Membrana , NAD+ Nucleosidasa/análisis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/enzimología , Fosfoproteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Leukemia ; 14(10): 1777-84, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11021753

RESUMEN

Recent studies suggest that the population of malignant cells found in human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells (LSCs). LSCs have been documented for nearly all AML subtypes and have been phenotypically described as CD34+/CD38- or CD34+/HLA-DR-. Given the potentially critical role of these primitive cells in perpetuating leukemic disease, we sought to further investigate their molecular and cellular characteristics. Flow cytometric studies using primary AML tissue showed that the interleukin-3 receptor alpha chain (IL-3Ralpha or CD123) was strongly expressed in CD34+/CD38- cells (98 +/- 2% positive) from 16 of 18 primary specimens. Conversely, normal bone marrow derived CD34+/CD38- cells showed virtually no detectable expression of the CD123 antigen. To assess the functional role of IL-3Ralpha positive cells, purified CD34+/CD123+ leukemia cells were transplanted into immune deficient NOD/SCID mice. These experiments showed that CD123+ cells were competent to establish and maintain leukemic populations in vivo. To begin to elucidate a biological role for CD123 in leukemia, primary AML samples were analyzed with respect to signal transduction activity in the MAPK, Akt, and Stat5 pathways. Phosphorylation was not detected in response to IL-3 stimulation, thereby suggesting CD123 is not active in conventional IL-3-mediated signaling. Collectively, these data indicate that CD123 represents a unique marker for primitive leukemic stem cells. Given the strong expression of this receptor on LSCs, we propose that targeting of CD123 may be a promising strategy for the preferential ablation of AML cells.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Receptores de Interleucina-3/metabolismo , Células Madre/metabolismo , Animales , Humanos , Inmunofenotipificación , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de Interleucina-3/química , Células Madre/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...