Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Med Genet A ; 176(12): 2924-2929, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30302932

RESUMEN

This report summarizes and highlights the fifth International RASopathies Symposium: When Development and Cancer Intersect, held in Orlando, Florida in July 2017. The RASopathies comprise a recognizable pattern of malformation syndromes that are caused by germ line mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. Because of their common underlying pathogenetic etiology, there is significant overlap in their phenotypic features, which includes craniofacial dysmorphology, cardiac, cutaneous, musculoskeletal, gastrointestinal and ocular abnormalities, neurological and neurocognitive issues, and a predisposition to cancer. The RAS pathway is a well-known oncogenic pathway that is commonly found to be activated in somatic malignancies. As in somatic cancers, the RASopathies can be caused by various pathogenetic mechanisms that ultimately impact or alter the normal function and regulation of the MAPK pathway. As such, the RASopathies represent an excellent model of study to explore the intersection of the effects of dysregulation and its consequence in both development and oncogenesis.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas ras/genética , Animales , Regulación de la Expresión Génica , Estudios de Asociación Genética/métodos , Desarrollo Humano , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Organogénesis/genética , Transducción de Señal , Síndrome , Proteínas ras/metabolismo
2.
Oncologist ; 22(4): 379-385, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28283585

RESUMEN

BACKGROUND: Although hypomethylating agents (HMAs) are effective and approved therapies for patients with myelodysplastic syndromes (MDS), many patients do not benefit from treatment, and nearly all ultimately stop responding to HMAs. The incidence and cost burden of HMA failure are unknown yet needed to appreciate the magnitude and significance of such failure. METHODS: We analyzed a de-identified dataset of over 5 million individuals with private health insurance in the U.S. to estimate MDS incidence, prevalence, and treatments. Based on MDS provider interviews, a conceptual model of MDS patient management was constructed to create a new, claims-relevant and drug development-relevant definition of HMA treatment failure. This algorithm was used to define resource encumbrance of MDS patients in whom HMA treatment failed. RESULTS: We estimated an MDS incidence rate of ∼70 cases per 100,000 enrollees per year and a prevalence of 155 cases per 100,000 enrollees. The proportion of MDS patients receiving HMA treatment was low (∼3%), and treatment was typically initiated within 1 year of the first MDS claim. Notably, HMA-treated individuals were older and had more comorbidities than the overall MDS cohort. Total health care costs of managing MDS patients after HMA failure were high (∼$77,000 during the first 6 months) and were driven primarily by non-pharmacy costs. CONCLUSION: This study quantifies for the first time the burden of significant unmet need in caring for MDS patients following HMA treatment failure. The Oncologist 2017;22:379-385Implications for Practice: U.S.-based treatment patterns among MDS patients demonstrate the significant clinical, financial, and health care burden associated with HMA failure and call for active therapies for this patient population.


Asunto(s)
Antimetabolitos Antineoplásicos/economía , Seguro de Salud/economía , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/economía , Metilación de ADN/genética , Femenino , Recursos en Salud/economía , Trasplante de Células Madre Hematopoyéticas/economía , Humanos , Masculino , Síndromes Mielodisplásicos/patología , Insuficiencia del Tratamiento
3.
Leuk Res ; 94: 106369, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32442785

RESUMEN

Phase 1 results from a Phase 1/2 study comprise 18 patients with myelodysplastic syndromes (MDS; n = 9), acute myeloid leukemia (AML; n = 8), and chronic myelomonocytic leukemia (CMML; n = 1) who were either hypomethylating agent naïve (n = 10) or relapsed/refractory following prior hypomethylating agent therapy (n = 8) (NCT01926587). Patients received oral rigosertib, an inhibitor of Ras-effector pathways, in 3 successive cohorts (140 mg twice daily, 280 mg twice daily, or 840 mg/day [560 mg morning/280 mg evening]) for 3 weeks of a 4-week cycle. Patients received parenteral azacitidine (75 mg/m2/day × 7 days) during the second week; the cycle repeated every 4 weeks. The combination was well tolerated for a median of 4 (range 1-41) cycles, with 72% of patients experiencing ≥1 serious adverse events. No dose-limiting toxicities were observed. Thus, no maximum tolerated dose was reached. The most frequently reported adverse events were diarrhea (50%), constipation, fatigue, and nausea (each 44%), and pneumonia and back pain (each 33%). Sequential administration demonstrated an overall response rate of 56% in evaluable patients, with responses observed in 7/9 MDS/CMML patients (78%) and 2/7 AML patients (29%). Further clinical studies are warranted to investigate this doublet therapy in patients with myeloid malignancies.


Asunto(s)
Azacitidina/administración & dosificación , Glicina/análogos & derivados , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Sulfonas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/efectos adversos , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/patología , Sulfonas/efectos adversos
4.
Leuk Res ; 64: 10-16, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29144985

RESUMEN

This Phase 1/2, dose-escalating study of rigosertib enrolled 22 patients with higher-risk myelodysplastic syndromes (MDS) (n=9) and acute myeloid leukemia (AML; n=13) who had relapsed or were refractory to standard therapy and for whom no second-line therapies were approved. Patients received 3- to 7-day continuous intravenous infusions of rigosertib, an inhibitor of Ras-effector pathways that interacts with the Ras-binding domains, common to several signaling proteins including Raf and PI3 kinase. Rigosertib was administered at doses of 650-1700mg/m2/day in 14-day cycles. Initial dose escalation followed a Fibonacci scheme, followed by recommended phase 2 dose confirmation in an expanded cohort. Rigosertib was well tolerated for up to 23 cycles, with no treatment-related deaths and 18% of patients with related serious adverse events (AEs). Common AEs were fatigue, diarrhea, pyrexia, dyspnea, insomnia, and anemia. Rigosertib exhibited biologic activity, with reduction or stabilization of bone marrow blasts and improved peripheral blood counts in a subset of patients. Ten of 19 evaluable patients (53%) demonstrated bone marrow/peripheral blood responses (n=4 MDS, n=1 AML) or stable disease (n=3 MDS, n=2 AML). Median survival was 15.7 and 2.0 months for responders and non-responders, respectively. Additional studies of rigosertib are ongoing in higher-risk MDS (NCT00854646).


Asunto(s)
Antineoplásicos/administración & dosificación , Glicina/análogos & derivados , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Sulfonas/administración & dosificación , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Humanos , Infusiones Intravenosas , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/mortalidad , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Sulfonas/efectos adversos , Análisis de Supervivencia
6.
Leuk Res ; 29(5): 481-91, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15755500

RESUMEN

Appropriate treatment for nonreactive thrombocytosis resulting from a myeloproliferative disorder (MPD) is surrounded by controversy. Although few doubt the association of thrombocytosis with increased risk for life-threatening events such as thrombosis or hemorrhage, or the association between clonal myeloproliferation and the progression to acute leukemia or myelofibrosis, controversy exists regarding the timing and nature of appropriate therapeutic intervention. Studies have shown that treatment with myelosuppressive agents such as chlorambucil, busulfan, radiophosphorus (32P), and hydroxyurea reduces the platelet count. However, investigators have also identified an increased risk of drug-related leukemic transformation. An ideal cytoreductive treatment for long-term use should minimize thrombosis and avoid long-term complications, especially acute leukemia (AL). Anagrelide, an imidazoquinolin, inhibits megakaryopoiesis and more selectively reduces platelet production in humans. A retrospective analysis of an open-label, multicenter, international trial reviewing 3660 anagrelide-treated patients was performed to assess efficacy and long-term safety, specifically potential for increased leukemogenicity. The study included MPD patients with thrombocytosis diagnosed according to Polycythemia Vera Study Group (PVSG) criteria. Of all patients enrolled, 81% had previously received other myelosuppressive agents; of these, 33% were transferred from the original agent to anagrelide due to toxicity and 31% were transferred because of poor platelet control. Over 45% of patients were symptomatic due to thrombocythemia, most commonly vascular sequelae (25%). Dosage was titrated to achieve a platelet count < 600 x 10(9) L(-1) and ideally between 130 and 450 x 10(9) L(-1). The safety cohort of 3660 patients, including 2251 with essential thrombocythemia (ET), 462 with polycythemia vera (PV), and 947 with chronic myeloid leukemia (CML) and other MPDs, was analyzed to establish the incidence of leukemic transformation in patients with ET and PV. From the Safety Population, 12.8% (467/3660) of patients were treated with anagrelide as the sole cytoreductive agent (naive patients). Acute leukemia/myelodysplasia developed in 2.1% of ET patients (47/2251) with a maximum follow-up of 7.1 years. Of the PV patients, 2.8% developed acute leukemia/myelodysplastic syndrome (13/462), with a maximum follow-up of 7.0 years. ET and PV patients who transformed to AL had all been previously exposed to other cytotoxics; there were no ET or PV patients in the study who transformed to AL exposed solely to anagrelide. With maximum follow-up over 7 years, anagrelide achieved platelet control in over 75% of MPD patients and did not increase the conversion to acute leukemia during the treatment duration analyzed. Longer follow-up is required to confirm these important observations regarding the long-term safety of anagrelide.


Asunto(s)
Trastornos Mieloproliferativos/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Quinazolinas/uso terapéutico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/patología , Estudios Retrospectivos , Seguridad , Factores de Tiempo
7.
Exp Hematol ; 31(2): 118-21, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12591276

RESUMEN

OBJECTIVE: In polycythemia vera (PV) there is no specific clonal marker because the molecular lesion responsible for PV is unknown. The recent demonstration that the PRV-1 gene is overexpressed in granulocytes from patients with PV provided the rationale for the current study to investigate whether PRV-1 is structurally rearranged, thus explaining its aberrant expression. MATERIALS AND METHODS: Fluorescence in situ hybridization was used to determine chromosomal localization of PRV-1 and to study whether the PRV1 gene is rearranged in 26 patients with PV. RESULTS: PRV-1 was localized to chromosome 19, band region q13.12-2. Structural rearrangements of PRV-1 were evaluated in bone marrow cells from 26 patients with PV: 14 with a normal karyotype and 12 with an abnormal karyotype. None of 150 metaphase cells or more than 10,000 interphase cells demonstrated PRV-1 gene deletion, amplification, or separation of the probe signal, which would indicate a PRV-1 rearrangement. CONCLUSION: These findings are consistent with a lack of structural rearrangement of PRV-1 in patients with PV. Thus, overexpression of PRV-1 in granulocytes from patients with PV is related to mechanisms that do not involve structural genetic changes.


Asunto(s)
Policitemia Vera/genética , Receptores de Superficie Celular/genética , Biomarcadores/análisis , Médula Ósea/patología , Aberraciones Cromosómicas , Mapeo Cromosómico , Cromosomas Humanos Par 19 , Femenino , Proteínas Ligadas a GPI , Expresión Génica , Reordenamiento Génico , Granulocitos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Isoantígenos , Masculino , Glicoproteínas de Membrana , Policitemia Vera/diagnóstico , Policitemia Vera/etiología , Receptores de Superficie Celular/análisis
8.
Exp Hematol ; 32(9): 877-80, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15345290

RESUMEN

Assessment of donor chimerism is becoming increasingly important in patients undergoing reduced-intensity conditioning (RIC) allogeneic bone marrow transplants, due to the possibility of mixed chimeras. This regimen has been used successfully for patients with leukemia and genetic disorders with donor chimerism occurring in the myeloid, lymphoid, and/or erythroid lineages. Less toxic RIC expands the potential application of stem cell transplants to patients with nonmalignant disorders of hematopoiesis, such as the severe form of Glanzmann thrombasthenia, who previously were not considered suitable candidates based on risk-benefit analysis. To assess megakaryocyte/platelet chimerism after stem cell transplantation conducted with RIC, we used restriction fragment length polymorphism (RFLP) and sequence analyses of the HPA-3 polymorphism in the megakaryocyte/platelet-specific glycoprotein alphaIIb. In this study we show that at 23 weeks post-RIC, a leukemia patient acquired the HPA-3 donor phenotype at the DNA and platelet RNA levels.


Asunto(s)
Antígenos de Plaqueta Humana/genética , Trasplante de Células Madre Hematopoyéticas , Megacariocitos/trasplante , Quimera por Trasplante , Acondicionamiento Pretrasplante , Linaje de la Célula , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Longitud del Fragmento de Restricción , Trasplante Homólogo
9.
Semin Hematol ; 40(1 Suppl 1): 30-3, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12682880

RESUMEN

Myelofibrosis with myeloid metaplasia, also known as idiopathic myelofibrosis (IF) or agnogenic myeloid metaplasia, is one of the characteristic manifestations of polycythemia vera (PV) in the spent phase, and has a particularly adverse prognosis. IF may also present de novo. To date, treatment strategies for both spent-phase PV and IF have frustrated both clinicians and patients, with little clear progress made over the past 50 years. Treatment modalities with some benefit in chronic myeloid leukemia (CML), such as interferon (IFN), have been used to shrink the massive organomegaly seen in these patients and to improve their marrow function, but are not curative, and not all patients respond or can tolerate the agent. A curative approach is allogeneic peripheral hematopoietic stem cell transplantation. The preparative regimens used in fully ablative techniques rule out older patients for consideration, and many younger patients with good prognostic criteria may do sufficiently well on medical treatment or observation to avoid transplantation. Older patients may have the option to undergo a human leukocyte antigen (HLA)-identical sibling transplant using a reduced intensity preparative regimen in order to minimize peritransplant mortality. Thus a prerequisite to the broad use of transplantation is objective determination of candidacy. Several evaluation methods agree that anemia, age, and cytogenetic abnormalities all predict poor survival in IF, suggesting that patients with anemia and an abnormal karyotype are the prime candidates for allogeneic transplantation. Experimental peripheral blood models that may reflect the degree of marrow fibrosis, such as the serum procollagen 3 peptide assay, have been used to determine if they are more informative of patient status than a single, random bone marrow sampling. Marrow fibrosis may be patchy, and thus a marrow biopsy alone without other data about marrow function may be misleading. Considerable long-term success in eradicating fibrosis and restoring normal cytogenetics, normal bone marrow morphology, and normal complete blood cell counts through transplantation has been reported. Many questions remain to be answered, however, before the appropriate role of hematopoietic stem cell transplantation in the setting of both spent-phase PV and IF can be determined.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trastornos Mieloproliferativos/diagnóstico , Trastornos Mieloproliferativos/terapia , Humanos , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Policitemia Vera/complicaciones , Policitemia Vera/mortalidad , Policitemia Vera/terapia , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/terapia , Factores de Riesgo , Tasa de Supervivencia , Trombocitemia Esencial/complicaciones , Trombocitemia Esencial/mortalidad
10.
Semin Hematol ; 41(2 Suppl 3): 18-22, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15190519

RESUMEN

Myeloproliferative disorders (MPDs) are chronic conditions that require long-term treatment. MPDs have been considered neoplastic disorders that have a propensity to transform to acute leukemia or myelodysplastic syndrome. One of the MPDs, chronic myeloid leukemia (CML), has the distinct cytogenetic abnormality of the Philadelphia chromosome. In CML, transformation to acute leukemia is a common occurrence, with the timeline measured in years rather than decades. With the other three disorders from this group, polycythemia vera (PV), essential thrombocythemia (ET), and idiopathic myelofibrosis (IMF), transformation to acute leukemia is less frequently seen and, although variable, can take a longer period of time. The choice of therapy used for MPDs may contribute to leukemic transformation; chlorambucil, busulfan, and radiophosphorus are all examples of therapy that have been shown to be leukemogenic. Several studies have suggested the potential leukemogenicity of hydroxyurea (HU), but this remains controversial because no randomized studies have been conducted. The two newest agents used for MPDs, interferon-alpha and anagrelide, have both been studied for efficacy, but their influence on the potential for leukemic transformation has not been well studied to date.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Busulfano/uso terapéutico , Clorambucilo/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/inducido químicamente , Síndromes Mielodisplásicos/inducido químicamente , Trastornos Mieloproliferativos/tratamiento farmacológico , Radiofármacos/uso terapéutico , Antineoplásicos Alquilantes/efectos adversos , Busulfano/efectos adversos , Clorambucilo/efectos adversos , Hidroxiurea/uso terapéutico , Interferón-alfa/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Síndromes Mielodisplásicos/etiología , Trastornos Mieloproliferativos/complicaciones , Cromosoma Filadelfia , Inhibidores de Agregación Plaquetaria/uso terapéutico , Quinazolinas/uso terapéutico , Radiofármacos/efectos adversos
11.
Mt Sinai J Med ; 70(3): 166-70, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12764534

RESUMEN

Historically, a variety of hematological disorders of the bone marrow, such as acute leukemia, chronic myelocytic leukemia and severe aplastic anemia, were invariably fatal. In the past two decades, basic and clinical scientists have been able to change this grim reality into a more hopeful outcome by replacing the diseased bone marrow with stem cells from either a family donor or an unrelated volunteer donor with identical human leukocyte antigens (HLA). Because of progress in this field, the initial experience of mainly using sibling donors has now been expanded to the use of either unrelated adult donors or cord blood cells as a source of hematopoietic cells. These approaches have created ethical concerns for donors and families, concerns that need to be discussed and understood by patients, volunteer donors and health care professionals.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/ética , Anemia Aplásica/terapia , Antígenos HLA/inmunología , Humanos , Leucemia/terapia , Síndromes Mielodisplásicos/terapia
12.
Leuk Res ; 36(2): 156-62, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21983176

RESUMEN

We treated 37 patients with polycythemia vera with imatinib mesylate (IM). The overall response rate was 49%. Thirty percent had a complete response, and 19%, a partial response. Thirty-one patients were treated for >120 days. Frequent side effects included nausea, diarrhea, edema, and skin rash. Whereas IM was effective in controlling erythropoiesis and reducing spleen size it was ineffective in controlling thrombocytosis. Normocellular marrow developed in 4 patients who had a complete response. Progression to overt myelofibrosis occurred in 3. Nevertheless, 6 patients have had a sustained complete response while on IM for >6 years. These patients were young, had high phlebotomy requirements, and only slightly elevated platelet counts. Therefore, we believe there may be a role for IM in patients with these characteristics whose disease cannot be controlled by, or as an alternative to, other myelosuppressive treatments.


Asunto(s)
Antineoplásicos/uso terapéutico , Piperazinas/uso terapéutico , Policitemia Vera/tratamiento farmacológico , Pirimidinas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Benzamidas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Policitemia/tratamiento farmacológico , Pronóstico , Trombocitosis/tratamiento farmacológico
14.
Clin Cancer Res ; 16(17): 4339-52, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20601445

RESUMEN

PURPOSE: To understand the changes in gene expression in polycythemia vera (PV) progenitor cells and their relationship to JAK2V617F. EXPERIMENTAL DESIGN: Messenger RNA isolated from CD34(+) cells from nine PV patients and normal controls was profiled using Affymetrix arrays. Gene expression change mediated by JAK2V617F was determined by profiling CD34(+) cells transduced with the kinase and by analysis of leukemia cell lines harboring JAK2V617F, treated with an inhibitor. RESULTS: A PV expression signature was enriched for genes involved in hematopoietic development, inflammatory responses, and cell proliferation. By quantitative reverse transcription-PCR, 23 genes were consistently deregulated in all patient samples. Several of these genes such as WT1 and KLF4 were regulated by JAK2, whereas others such as NFIB and EVI1 seemed to be deregulated in PV by a JAK2-independent mechanism. Using cell line models and comparing gene expression profiles of cell lines and PV CD34(+) PV specimens, we have identified panels of 14 JAK2-dependent genes and 12 JAK2-independent genes. These two 14- and 12-gene sets could separate not only PV from normal CD34(+) specimens, but also other MPN such as essential thrombocytosis and primary myelofibrosis from their normal counterparts. CONCLUSIONS: A subset of the aberrant gene expression in PV progenitor cells can be attributed to the action of the mutant kinase, but there remain a significant number of genes characteristic of the disease but deregulated by as yet unknown mechanisms. Genes deregulated in PV as a result of the action of JAK2V617F or independent of the kinase may represent other targets for therapy.


Asunto(s)
Perfilación de la Expresión Génica , Janus Quinasa 2/genética , Mutación , Policitemia Vera/genética , Sustitución de Aminoácidos , Antígenos CD34/genética , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyetina/farmacología , Humanos , Janus Quinasa 2/metabolismo , Factor 4 Similar a Kruppel , Análisis de Secuencia por Matrices de Oligonucleótidos , Policitemia Vera/sangre , Policitemia Vera/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
16.
Blood ; 105(11): 4508-15, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15705794

RESUMEN

Idiopathic myelofibrosis (IM) is characterized by increased numbers of CD34(+) cells in the peripheral blood (PB). We explored the possible mechanisms underlying this abnormal trafficking of CD34(+) cells. Plasma levels of neutrophil elastase (NE), total and active matrix metalloproteinase 9 (MMP-9), and soluble vascular cell adhesion molecule-1 (sVCAM-1) were dramatically increased in IM. The absolute number of CD34(+) cells in the PB was correlated with the levels of sVCAM-1. Marked elevations of the levels of NE but not total and active MMP-9 as well as MMP-2 were detected in media conditioned by IM mononuclear cells (MNCs) as compared with that of healthy volunteers. IM MNC-conditioned media, however, was shown by zymographic analysis to contain increased gelatinolytic activity corresponding to the molecular weight of MMP-9. IM MNC-conditioned media also exhibited a greater ability to cleave VCAM-1 and c-kit in vitro, consistent with the biologic actions of NE. In addition, the increased ability of IM PB CD34(+) cells to migrate through a reconstituted basement membrane was diminished by several inhibitors of MMP-9 activity, indicating that these cells express increased levels of this MMP. These data indicate that a proteolytic environment exists in IM which might result in the sustained mobilization of CD34(+) cells.


Asunto(s)
Movimiento Celular , Células Madre Hematopoyéticas/patología , Péptido Hidrolasas/metabolismo , Mielofibrosis Primaria/enzimología , Antígenos CD34 , Células Sanguíneas/patología , Medios de Cultivo Condicionados/química , Humanos , Elastasa de Leucocito/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
Blood ; 105(4): 1699-705, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15471948

RESUMEN

Idiopathic myelofibrosis (IM) is characterized by the constitutive mobilization of CD34(+) cells. IM peripheral blood (PB) CD34(+) cells had a reduced cloning efficiency and a lower frequency of cobblestone areas compared with normal granulocyte colony-stimulating factor (G-CSF)-mobilized PB CD34(+) cells. IM CD34(+) cells engrafted nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, demonstrating that they contain bone marrow (BM)-repopulating cells. G-CSF-mobilized CD34(+) cells produced multiple hematopoietic lineages within the NOD/SCID mice with a predominance of CD19(+) cells. By contrast, IM CD34(+) cells produced predominantly CD33(+) cells, increased numbers of CD41(+) cells, but fewer CD19(+) cells. Transcriptional clonality assays of the engrafted human IM cells demonstrated their clonal origin. CD34(+) cells from one patient isolated prior to leukemic transformation were capable of generating acute leukemia in NOD/SCID mice. The engrafted human cells exhibited the same abnormal karyotype as primary cells in a portion of the population. These findings demonstrate that BM-repopulating cells and more differentiated progenitor cells are constitutively mobilized into the PB in IM, and that their differentiation program is abnormal. In addition, the NOD/SCID model may be useful in gaining an understanding of the events occurring during the transition of IM to acute leukemia.


Asunto(s)
Células de la Médula Ósea/patología , Movimiento Celular , Hematopoyesis , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/patología , Enfermedad Aguda , Animales , Antígenos CD34/biosíntesis , Células de la Médula Ósea/inmunología , Células Clonales , Femenino , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Humanos , Inmunofenotipificación , Leucemia/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mielofibrosis Primaria/genética
18.
Blood ; 105(10): 4115-9, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15671439

RESUMEN

A total of 21 patients with myelofibrosis with myeloid metaplasia (MMM), with a median age of 54 years (range, 27-68 years), were prepared with a reduced-intensity conditioning (RIC) regimen. The patients received an allogeneic marrow (n = 3) or peripheral blood stem-cell (n = 18) transplant from HLA-matched related (n = 18) or unrelated (n = 2), or 1 Ag-mismatched related (n = 1), donors. RIC regimens included fludarabine/total body irradiation 200 cGy (n = 5) or 450 cGy (n = 1), fludarabine/melphalan (n = 7), thiotepa/cyclophosphamide (n = 7), and thiotepa/fludarabine (n = 1). At the time of transplantation, all of the patients were at intermediate (n = 13) or high (n = 8) risk, according to the Dupriez classification. Of the patients, 19 had grade III or IV marrow fibrosis. All of the patients achieved full engraftment but one. Posttransplantation chimerism analysis showed more than 95% donor cells in 18 patients, while 2 patients achieved complete donor chimerism after donor leukocyte infusion (DLI). Acute graft-versus-host disease (GVHD) grades II to IV was observed in 7 patients, grades III to IV in 2, and extensive chronic GVHD in 8 of 18 evaluable patients. There were 3 patients who died from acute GVHD, infection, and relapse. There are 18 patients alive 12 to 122 months (median, 31 months) after transplantation, and 17 are in remission (1 after a second transplantation). The use of RIC regimens in allogeneic stem cell transplantation results in prolonged survival in intermediate/high-risk MMM patients.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/terapia , Acondicionamiento Pretrasplante , Adulto , Anciano , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/cirugía , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia , Factores de Tiempo , Trasplante Homólogo
19.
Br J Haematol ; 119(2): 558-66, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12406101

RESUMEN

Between 1986 and 2001, 220 patients with polycythaemia vera (PV) were studied using conventional cytogenetics. Of 204 evaluable patients, 52 (25.4%) had clonal abnormalities. The recurrent chromosomal rearrangements were those of chromosome 9 (21.1%), del(20q) (19.2%), trisomy 8 (19.2%), rearrangements of 13q (13.4%), abnormalities of 1q (11.5%), and of chromosomes 5 and 7 (9.6%). Subsequent analysis of 32 patients, performed at follow-up of up to 14.8 years, revealed new clonal abnormalities in five patients and the disappearance of an abnormal clone in four. Eleven patients remained normal up to 11.5 years and seven patients maintained an abnormality for over 10 years. Fifty-three patients were studied retrospectively using interphase fluorescence in situ hybridization (I-FISH), utilizing probes for centromere enumeration of chromosomes 8 and 9, and for 13q14 and 20q12 loci. Conventional cytogenetics demonstrated clonal chromosome abnormalities in 23% of these 53 patients. The addition of I-FISH increased the detection of abnormalities to 29% and permitted clarification of chromosome 9 rearrangements in an additional 5.6% of patients. FISH uncovered rearrangements of chromosome 9 in 53% of patients with an abnormal FISH pattern, which represented the most frequent genomic alteration in this series.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 9 , Policitemia Vera/genética , Mapeo Cromosómico , Femenino , Humanos , Hibridación Fluorescente in Situ , Incidencia , Masculino , Estudios Prospectivos , Estudios Retrospectivos , Factores de Tiempo
20.
Biol Blood Marrow Transplant ; 9(7): 453-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12869959

RESUMEN

Nonmyeloablative allogeneic peripheral blood progenitor cell transplantation with low-dose total body irradiation (TBI; 200 cGy) plus fludarabine followed by cyclosporine and mycophenolate mofetil results in modest graft rejection rates. Acute and chronic graft-versus-host diseases (GVHD) are also seen and may not differ substantially from those that occur after fully ablative transplantation. Adding antithymocyte globulin (ATG) to pretransplant conditioning produces substantial immunosuppression. Because of its persistence in the circulation, ATG can achieve in vivo T-cell depletion. Twenty-five patients who were not eligible for conventional fully ablative allogeneic stem cell transplantation by virtue of age or comorbidities underwent nonmyeloablative allogeneic transplantation with ATG 15 mg/kg/d days -4 to -1, TBI 200 cGy on a single fraction on day -5, and fludarabine 30 mg/m(2)/d on days -4 to -2. Oral mycophenolate mofetil 15 mg/kg every 12 hours and cyclosporine 6 mg/kg every 12 hours were started on day -5. Grafts were unmanipulated peripheral blood progenitor cells mobilized with filgrastim 10 microg/kg/d and collected on day 5. The median age of the recipients was 57 years (range, 30-67 years); diagnoses were non-Hodgkin lymphoma (n = 11), acute myeloid leukemia (n = 6), multiple myeloma (n = 3), acute lymphoblastic leukemia (n = 2), severe aplastic anemia (n = 1), paroxysmal nocturnal hemoglobinuria (n = 1), and myelodysplastic syndrome (n = 1). The median CD34(+) and CD3(+) contents of the grafts were 7.6 x 10(6)/kg and 1.6 x 10(8)/kg, respectively. Five patients received voluntary unrelated donor grafts. Three patients, 2 with voluntary unrelated donor grafts and 1 with a sib donor, received a 1 antigen-mismatched graft. The rest were fully matched. Twenty-two of 25 patients were evaluable for chimerism. Sixteen had >/=95% donor chimerism. Four patients displayed 80% to 90% donor chimerism, 1 displayed 78%, and 1 displayed 64%. Eleven patients relapsed with their original disease. One patient rejected the graft at 180 days. The median hospital stay was 27 days. Complications included GVHD in 6 patients (3 patients had grade I or II GVHD of skin and liver, and 3 patients had grade III or IV GVHD of liver and gut). Two of the patients with GVHD had mismatched grafts. Transplant-related toxicity was seen in 4 patients and infection in 5 patients. The median length of follow-up was 162 days (range, 17-854 days). Complete remissions were seen in 10 patients. Four patients remained in complete response (CR) at 280 to 595 days. One patient relapsed with non-Hodgkin lymphoma after a CR of 728 days. Of the 25 patients, 16 died (6 of relapsed disease, 4 of GVHD, 3 of infection, and 3 of transplant-related toxicity) and 9 are alive (6 with CR-2 of them after donor leukocyte infusion-and 3 with relapsed disease). The addition of ATG to low-dose TBI and fludarabine nonmyeloablative conditioning was well tolerated and resulted in >80% donor engraftment in this small cohort. As in other series of truly nonmyeloablative transplantation, a high rate of relapse was observed. Donor engraftment may be facilitated by the addition of ATG to low-dose TBI and fludarabine conditioning.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Enfermedades de la Médula Ósea/radioterapia , Trasplante de Médula Ósea , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico , Irradiación Corporal Total , Adulto , Anciano , Animales , Enfermedades de la Médula Ósea/tratamiento farmacológico , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Rechazo de Injerto/tratamiento farmacológico , Humanos , Inmunosupresores/efectos adversos , Leucemia/radioterapia , Linfoma/radioterapia , Masculino , Persona de Mediana Edad , Dosificación Radioterapéutica , Quimera por Trasplante , Trasplante Homólogo , Irradiación Corporal Total/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA