Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 107(5): 1026-1033, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34348455

RESUMEN

Novel treatment strategies are needed to improve cure for all children with acute lymphoblastic leukemia (ALL). To this end, we investigated the therapeutic potential of clofarabine in primary ALL in trial CoALL 08-09 (clinicaltrials gov. identifier: NCT01228331). The primary study objective was the minimal residual disease (MRD)- based comparative assessment of cytotoxic efficacies of clofarabine 5x40 mg/m2 versus high-dose cytarabine (HIDAC) 4x3g/m2, both in combination with PEG-ASP 2,500 IU/m2 as randomized intervention in early consolidation. The secondary objective was an outcome analysis focused on treatment arm dependence and MRD after randomized intervention. In B-cell precursor (BCP)-ALL, eradication of MRD was more profound after clofarabine compared to cytarabine, with 93 versus 79 of 143 randomized patients per arm reaching MRD-negativity (c2 test P=0.03, leftsided P [Fisher's exact test]=0.04). MRD status of BCP-ALL after randomized intervention maintained its prognostic relevance, with a significant impact on event-free survival (EFS) and relapse rate. However, no difference in outcome regarding EFS and overall survival (OS) between randomized courses was observed (5-year EFS: clofarabine 85.7, SE=4.1 vs. HIDAC 84.8, SE=4.7 [P=0.96]; OS: 95.7, SE=1.9 vs. 92.2, SE=3.2 [P=0.59]), independent of covariates or overall risk strata. Severe toxicities between randomized and subsequent treatment elements were also without significant difference. In conclusion, clofarabine/PEG-ASP is effective and safe, but greater cytotoxic efficacy of clofarabine compared to HIDAC did not translate into improved outcomes indicating a lack of surrogacy of post-intervention MRD at the trial level as opposed to the patient level, which hampers a broader implementation of this regimen in the frontline treatment of ALL.


Asunto(s)
Antineoplásicos , Linfoma no Hodgkin , Leucemia-Linfoma Linfoblástico de Células Precursoras , Enfermedad Aguda , Antineoplásicos/uso terapéutico , Niño , Clofarabina , Citarabina/uso terapéutico , Supervivencia sin Enfermedad , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Resultado del Tratamiento
2.
Pediatr Blood Cancer ; 66(8): e27780, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31034759

RESUMEN

BACKGROUND: Assessment of minimal residual disease (MRD) is an integral component for response monitoring and treatment stratification in acute lymphoblastic leukemia (ALL). We aimed to evaluate the genomic ETV6-RUNX1 fusion sites as a single marker for MRD quantification. PROCEDURE: In a representative, uniformly treated cohort of pediatric relapsed ALL patients (n = 52), ETV6-RUNX1 fusion sites were compared to the current gold standard, immunoglobulin/T-cell receptor (Ig/TCR) gene rearrangements. RESULTS: Primer/probe sets designed to ETV6-RUNX1 fusions achieved significantly more frequent a sensitivity and a quantitative range of at least 10-4 compared to the gold standard with 100% and 73% versus 76% and 47%, respectively. The breakpoint sequence was identical at diagnosis and relapse in all tested cases. There was a high degree of concordance between quantitative MRD results assessed using ETV6-RUNX1 and the highest Ig/TCR marker (Spearman's 0.899, P < .01) with differences >½ log-step in only 6% of patients. A high proportion of ETV6-RUNX1-positive ALL relapses (40%) in our cohort showed a poor response to induction treatment at relapse, and therefore had an indication for hematopoietic stem cell transplantation, demonstrating the need of accurate identification of this subgroup. CONCLUSIONS: ETV6-RUNX1 fusion sites are highly sensitive and reliable MRD markers. Our data confirm that they are unaffected by clonal evolution and selection during front-line and second-line chemotherapy in contrast to Ig/TCR rearrangements, which require several markers per patient to compensate for the observed loss of target clones. In future studies, the genomic ETV6-RUNX1 fusion can be used as single MRD marker.


Asunto(s)
Biomarcadores de Tumor/genética , Evolución Clonal , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Genómica/métodos , Trasplante de Células Madre Hematopoyéticas , Neoplasia Residual/patología , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Estudios de Seguimiento , Humanos , Neoplasia Residual/genética , Neoplasia Residual/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Pronóstico , Estudios Prospectivos , Curva ROC
3.
Eur J Immunol ; 47(2): 364-373, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27925643

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening inflammatory syndrome characterized by hyperactivation of lymphocytes and histiocytes. T cells play a key role in HLH pathogenesis, but their differentiation pattern is not well characterized in patients with active HLH. We compared T-cell activation patterns between patients with familial HLH (1°HLH), 2°HLH without apparent infectious trigger (2°HLH) and 2°HLH induced by a viral infection (2°V-HLH). Polyclonal CD8+ T cells are highly activated in 1°HLH and 2°V-HLH, but less in 2°HLH as assessed by HLA-DR expression and marker combination with CD45RA, CCR7, CD127, PD-1 and CD57. Absence of increased HLA-DR expression on T cells excluded active 1° HLH with high sensitivity and specificity. A high proportion of polyclonal CD127- CD4+ T cells expressing HLA-DR, CD57, and perforin is a signature of infants with 1°HLH, much less prominent in virus-associated 2°HLH. The similar pattern and extent of CD8+ T-cell activation compared to 2° V-HLH is compatible with a viral trigger of 1°HLH. However, in most 1°HLH patients no triggering infection was documented and the unique activation of cytotoxic CD4+ T cells indicates that the overall T-cell response in 1°HLH is different. This may reflect different pathways of pathogenesis of these two HLH variants.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Virosis/inmunología , Antígenos CD57/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Lactante , Recién Nacido , Activación de Linfocitos , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/etiología , Masculino , Perforina/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Virosis/complicaciones , Virosis/diagnóstico
4.
Blood ; 127(8): 997-1006, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26744459

RESUMEN

Genetic disorders affecting biogenesis and transport of lysosome-related organelles are heterogeneous diseases frequently associated with albinism. We studied a patient with albinism, neutropenia, immunodeficiency, neurodevelopmental delay, generalized seizures, and impaired hearing but with no mutation in genes so far associated with albinism and immunodeficiency. Whole exome sequencing identified a homozygous mutation in AP3D1 that leads to destabilization of the adaptor protein 3 (AP3) complex. AP3 complex formation and the degranulation defect in patient T cells were restored by retroviral reconstitution. A previously described hypopigmented mouse mutant with an Ap3d1 null mutation (mocha strain) shares the neurologic phenotype with our patient and shows a platelet storage pool deficiency characteristic of Hermansky-Pudlak syndrome (HPS) that was not studied in our patient because of a lack of bleeding. HPS2 caused by mutations in AP3B1A leads to a highly overlapping phenotype without the neurologic symptoms. The AP3 complex exists in a ubiquitous and a neuronal form. AP3D1 codes for the AP3δ subunit of the complex, which is essential for both forms. In contrast, the AP3ß3A subunit, affected in HPS2 patients, is substituted by AP3ß3B in the neuron-specific heterotetramer. AP3δ deficiency thus causes a severe neurologic disorder with immunodeficiency and albinism that we propose to classify as HPS10.


Asunto(s)
Complejo 3 de Proteína Adaptadora/genética , Subunidades delta de Complexo de Proteína Adaptadora/genética , Síndrome de Hermanski-Pudlak/clasificación , Síndrome de Hermanski-Pudlak/genética , Síndromes de Inmunodeficiencia/genética , Convulsiones/genética , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Mutación , Transfección
5.
Pediatr Blood Cancer ; 65(11): e27344, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30070073

RESUMEN

In this report, we evaluate the hypothesis that hemophagocytic lymphohistiocytosis in patients with defects of lymphocyte cytotoxicity is usually triggered by infections. We show that in the majority of patients, extensive virus PCR panels performed in addition to routine microbiological investigations remain negative and summarize 25 patients with onset of hemophagocytic lymphohistiocytosis in utero or within the first 10 days of life, in none of which an associated bacterial or viral infection was reported. These observations, even though preliminary, invite to consider a key role of lymphocyte cytotoxicity in controlling T-cell homeostasis also in the absence of apparent infectious stimuli.


Asunto(s)
Linfohistiocitosis Hemofagocítica/etiología , Infecciones Bacterianas/epidemiología , Niño , Femenino , Feto , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/etiología , Masculino , Embarazo , Virosis/epidemiología , Adulto Joven
6.
J Clin Immunol ; 37(8): 770-780, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28936583

RESUMEN

We report our experience in using flow cytometry-based immunological screening prospectively as a decision tool for the use of genetic studies in the diagnostic approach to patients with hemophagocytic lymphohistiocytosis (HLH). We restricted genetic analysis largely to patients with abnormal immunological screening, but included whole exome sequencing (WES) for those with normal findings upon Sanger sequencing. Among 290 children with suspected HLH analyzed between 2010 and 2014 (including 17 affected, but asymptomatic siblings), 87/162 patients with "full" HLH and 79/111 patients with "incomplete/atypical" HLH had normal immunological screening results. In 10 patients, degranulation could not be tested. Among the 166 patients with normal screening, genetic analysis was not performed in 107 (all with uneventful follow-up), while 154 single gene tests by Sanger sequencing in the remaining 59 patients only identified a single atypical CHS patient. Flow cytometry correctly predicted all 29 patients with FHL-2, XLP1 or 2. Among 85 patients with defective NK degranulation (including 13 asymptomatic siblings), 70 were Sanger sequenced resulting in a genetic diagnosis in 55 (79%). Eight patients underwent WES, revealing mutations in two known and one unknown cytotoxicity genes and one metabolic disease. FHL3 was the most frequent genetic diagnosis. Immunological screening provided an excellent decision tool for the need and depth of genetic analysis of HLH patients and provided functionally relevant information for rapid patient classification, contributing to a significant reduction in the time from diagnosis to transplantation in recent years.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Células Asesinas Naturales/inmunología , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM/genética , Linfohistiocitosis Hemofagocítica/diagnóstico , Proteínas Musculares/genética , Factores de Transcripción/genética , Enfermedades Asintomáticas , Degranulación de la Célula , Niño , Citometría de Flujo , Pruebas Genéticas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Proteínas Musculares/metabolismo , Mutación/genética , Trasplante de Órganos , Guías de Práctica Clínica como Asunto , Pronóstico , Estudios Prospectivos , Hermanos , Factores de Transcripción/metabolismo , Secuenciación del Exoma
7.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28453885

RESUMEN

Blinatumomab is a bispecific T-cell engaging αCD19 antibody used in refractory or relapsed B-cell precursor acute lymphoblastic leukemia (ALL). Recently, lineage switch to a myeloid phenotype has been described following CD19 targeting treatment in three pediatric patients with mixed lineage leukemia (MLL) rearranged ALL. We report the case of a female who received blinatumomab for a first relapse of ALL without MLL alterations. She suffered from a second relapse early after hematopoietic stem cell transplantation and was treated with blinatumomab again. During this treatment, the leukemia lost CD19 expression as well as nearly all other B-cell markers, while still harboring the initial minimal residual disease marker, and switched to a myeloid phenotype.


Asunto(s)
Anticuerpos Biespecíficos/efectos adversos , Antígenos CD19/metabolismo , Linaje de la Célula , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Leucemia Mieloide Aguda/inducido químicamente , Recurrencia Local de Neoplasia/tratamiento farmacológico , Antineoplásicos/efectos adversos , Niño , Femenino , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteína de la Leucemia Mieloide-Linfoide/genética , Recurrencia Local de Neoplasia/epidemiología , Neoplasia Residual/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Pronóstico
8.
Br J Haematol ; 170(4): 539-49, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25940575

RESUMEN

Haemophagocytic lymphohistiocytosis (HLH) in the context of malignancy is mainly considered a challenge of adult haematology. While this association is also observed in children, little is known regarding inciting factors, appropriate treatment and prognosis. We retrospectively analysed 29 paediatric and adolescent patients for presenting features, type of neoplasm or preceding chemotherapy, treatment and outcome. Haemophagocytic lymphohistiocytosis was considered triggered by the malignancy (M-HLH) in 21 patients, most of whom had T- (n = 12) or B-cell neoplasms (n = 7), with Epstein-Barr virus as a co-trigger in five patients. In eight patients, HLH occurred during chemotherapy (Ch-HLH) for malignancy, mainly acute leukaemias (n = 7); an infectious trigger was found in seven. In M- and Ch-HLH, median overall survival was 1·2 and 0·9 years, and the 6 month survival rates were 67% and 63%, respectively. Seven of 11 deceased M-HLH patients exhibited active malignancy and HLH at the time of death, while only two out of five deceased Ch-HLH patients had evidence of active HLH. To overcome HLH, malignancy- and HLH-directed treatments were administered in the M-HLH cohort; however, it was not possible to determine superiority of one approach over the other. For Ch-HLH, treatment ranged from postponement of chemotherapy to the use of etoposide-containing regimens.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Leucemia , Linfohistiocitosis Hemofagocítica , Enfermedad Aguda , Adolescente , Adulto , Niño , Preescolar , Supervivencia sin Enfermedad , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/mortalidad , Femenino , Humanos , Lactante , Recién Nacido , Leucemia/tratamiento farmacológico , Leucemia/mortalidad , Linfohistiocitosis Hemofagocítica/etiología , Linfohistiocitosis Hemofagocítica/mortalidad , Linfohistiocitosis Hemofagocítica/terapia , Masculino , Estudios Retrospectivos , Tasa de Supervivencia
9.
J Clin Immunol ; 35(1): 22-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25491289

RESUMEN

Familial hemophagocytic lymphohistiocytosis (FHL) is a genetically heterogeneous hyperinflammatory syndrome, caused by an uncontrolled and ineffective proliferation and activation of T-lymphocytes, NK-cells, and macrophages that infiltrate multiple organs. Herein, a patient is presented who suffered from hepatitis and atypical brain lesions. Genetic studies revealed a homozygous mutation in the STXP2 gene; and thus, the diagnosis of FHL5 was confirmed.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Proteínas Munc18/genética , Agammaglobulinemia/genética , Agammaglobulinemia/inmunología , Edad de Inicio , Encéfalo/patología , Niño , Resultado Fatal , Hepatitis Autoinmune/genética , Hepatitis Autoinmune/inmunología , Homocigoto , Humanos , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/inmunología , Imagen por Resonancia Magnética , Masculino , Mutación , Linfocitos T/inmunología
10.
Blood ; 121(4): 604-13, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23190531

RESUMEN

Syntaxin-11 (Stx11), an atypical member of the SNARE protein family, is part of the cytolytic machinery of T and NK cells and involved in the fusion of lytic granules with the plasmamembrane. Functional loss of syntaxin-11 in humans causes defective degranulation and impaired cytolytic activity of T and NK cells. Furthermore, patients with STX11 deficiency develop familial hemophagocytic lymphohistiocytosis type 4 (FHL4), a life-threatening disease of severe hyperinflammation. We established Stx11-deficient mice as an animal model for FHL4. Stx11-deficient mice exhibited severely reduced degranulation and cytolytic activity of CTL and NK cells and developed all clinical symptoms of hemophagocytic lymphohistiocytosis (HLH) after infection with lymphocytic choriomeningitis virus (LCMV). The HLH phenotype was further characterized by hyperactive CD8 T cells and continuous IFN-γ production. However, in contrast to perforin-deficient mice, which represent a model for FHL2, progression of HLH was not fatal. Survival of Stx11-deficient mice was determined by exhaustion of antigen-specific T cells, characterized by expression of inhibitory receptors, sequential loss of effector functions, and finally T-cell deletion. Blockade of inhibitory receptors on T cells in Stx11-deficient mice converted nonfatal disease course into fatal HLH, identifying T-cell exhaustion as an important factor for determination of disease severity in HLH.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Proteínas Qa-SNARE/genética , Animales , Antígenos CD/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Degranulación de la Célula/genética , Degranulación de la Célula/inmunología , Citotoxicidad Inmunológica/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Virus de la Coriomeningitis Linfocítica , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/virología , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Proteína del Gen 3 de Activación de Linfocitos
11.
Blood ; 121(15): 2943-51, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23403622

RESUMEN

Genetic disorders of lymphocyte cytotoxicity predispose patients to hemophagocytic lymphohistiocytosis (HLH). Reduced lymphocyte cytotoxicity has been demonstrated in Hermansky-Pudlak syndrome type 2 (HPS2), but only a single patient was reported who developed HLH. Because that patient also carried a potentially contributing heterozygous RAB27A mutation, the risk for HLH in HPS2 remains unclear. We analyzed susceptibility to HLH in the pearl mouse model of HPS2. After infection with lymphocytic choriomeningitis virus, pearl mice developed all key features of HLH, linked to impaired virus control caused by a moderate defect in CTL cytotoxicity in vivo. However, in contrast to perforin-deficient mice, the disease was transient, and all mice fully recovered and controlled the infection. An additional heterozygous Rab27a mutation did not aggravate the cytotoxicity defect or disease parameters. In the largest survey of 22 HPS2 patients covering 234 patient years, we identified only 1 additional patient with HLH and 2 with incomplete transient HLH-like episodes, although cytotoxicity or degranulation was impaired in all 16 patients tested. HPS2 confers a risk for HLH that is lower than in Griscelli or Chediak-Higashi syndrome, probably because of a milder defect in cytotoxicity. Preemptive hematopoietic stem cell transplantation does not appear justified in HPS2.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Síndrome de Hermanski-Pudlak/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Complejo 3 de Proteína Adaptadora/deficiencia , Complejo 3 de Proteína Adaptadora/genética , Complejo 3 de Proteína Adaptadora/inmunología , Subunidades beta de Complejo de Proteína Adaptadora/deficiencia , Subunidades beta de Complejo de Proteína Adaptadora/genética , Subunidades beta de Complejo de Proteína Adaptadora/inmunología , Adolescente , Adulto , Animales , Niño , Preescolar , Citotoxicidad Inmunológica/genética , Citometría de Flujo , Síndrome de Hermanski-Pudlak/complicaciones , Síndrome de Hermanski-Pudlak/genética , Humanos , Linfohistiocitosis Hemofagocítica/etiología , Linfohistiocitosis Hemofagocítica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Factores de Riesgo , Linfocitos T Citotóxicos/metabolismo , Adulto Joven , Proteínas de Unión al GTP rab/deficiencia , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/inmunología , Proteínas rab27 de Unión a GTP
13.
Eur J Immunol ; 43(1): 194-208, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23042080

RESUMEN

Syntaxin 11 (STX11) controls vesicular trafficking and is a key player in exocytosis. Since Stx11 mutations are causally associated with a familial hemophagocytic lymphohistio-cytosis, we wanted to clarify whether STX11 is functionally important for key immune cell populations. This was studied in primary cells obtained from newly generated Stx11(-/-) mice. Our data revealed that STX11 is not only widely expressed in different immune cells, but also induced upon LPS or IFN-γ treatment. However, Stx11 deficiency does not affect macrophage phagocytic function and cytokine secretion, mast cell activation, or antigen presentation by DCs. Instead, STX11 selectively controls lymphocyte cytotoxicity in NK and activated CD8(+) T cells and degranulation in neutrophils. Stx11(-/-) NK cells and CTLs show impaired degranulation, despite a comparable activation, maturation and expression of the complex-forming partners MUNC18-2 and VTI1B. In addition, Stx11(-/-) CTLs and NK cells produce abnormal levels of IFN-γ. Since functional reconstitution rescues the defective phenotype of Stx11(-/-) CTLs, we suggest a direct, specific and key role of STX11 in controlling lymphocyte cytotoxicity, cytokine production and secretion. Finally, we show that these mice are a very useful tool for dissecting the role of STX11 in vesicular trafficking and secretion.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Neutrófilos/inmunología , Proteínas Qa-SNARE/inmunología , Animales , Degranulación de la Célula/genética , Línea Celular , Citotoxicidad Inmunológica/genética , Humanos , Interferón gamma/inmunología , Lipopolisacáridos/inmunología , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Munc18/inmunología , Mutación/genética , Proteínas Qa-SNARE/genética , Proteínas Qb-SNARE/inmunología
14.
Blood ; 119(12): 2754-63, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22294731

RESUMEN

Familial hemophagocytic lymphohistiocytosis (FHL) is a life-threatening disorder of immune regulation caused by defects in lymphocyte cytotoxicity. Rapid differentiation of primary, genetic forms from secondary forms of hemophagocytic lymphohistiocytosis (HLH) is crucial for treatment decisions. We prospectively evaluated the performance of degranulation assays based on surface up-regulation of CD107a on natural killer (NK) cells and cytotoxic T lymphocytes in a cohort of 494 patients referred for evaluation for suspected HLH. Seventy-five of 77 patients (97%) with FHL3-5 and 11 of 13 patients (85%) with Griscelli syndrome type 2 or Chediak-Higashi syndrome had abnormal resting NK-cell degranulation. In contrast, NK-cell degranulation was normal in 14 of 16 patients (88%) with X-linked lymphoproliferative disease and in 8 of 14 patients (57%) with FHL2, who were identified by diminished intracellular SLAM-associated protein (SAP), X-linked inhibitor of apoptosis protein (XIAP), and perforin expression, respectively. Among 66 patients with a clinical diagnosis of secondary HLH, 13 of 59 (22%) had abnormal resting NK-cell degranulation, whereas 0 of 43 had abnormal degranulation using IL-2-activated NK cells. Active disease or immunosuppressive therapy did not impair the assay performance. Overall, resting NK-cell degranulation below 5% provided a 96% sensitivity for a genetic degranulation disorder and a specificity of 88%. Therefore, degranulation assays allow a rapid and reliable classification of patients, benefiting treatment decisions.


Asunto(s)
Degranulación de la Célula/fisiología , Pruebas Inmunológicas/métodos , Células Asesinas Naturales/fisiología , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfocitos T Citotóxicos/fisiología , Humanos , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas , Estudios Prospectivos , Sensibilidad y Especificidad , Factores de Tiempo
15.
Blood ; 119(25): 6016-24, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22451424

RESUMEN

Familial hemophagocytic lymphohistiocytosis (FHL) is a genetically determined hyperinflammatory syndrome caused by uncontrolled immune response mediated by T-lymphocytes, natural killer (NK) cells, and macrophages. STXBP2 mutations have recently been associated with FHL5. To better characterize the genetic and clinical spectrum of FHL5, we analyzed a cohort of 185 patients with suspected FHL for mutations in STXBP2. We detected biallelic mutations in 37 patients from 28 families of various ethnic origins. Missense mutations and mutations affecting 1 of the exon 15 splice sites were the predominant changes detectable in this cohort. Patients with exon 15 splice-site mutations (n = 13) developed clinical manifestations significantly later than patients with other mutations (median age, 4.1 year vs 2 months) and showed less severe impairment of degranulation and cytotoxic function of NK cells and CTLs. Patients with FHL5 showed several atypical features, including sensorineural hearing deficit, abnormal bleeding, and, most frequently, severe diarrhea that was only present in early-onset disease. In conclusion, we report the largest cohort of patients with FHL5 so far, describe an extended disease spectrum, and demonstrate for the first time a clear genotype-phenotype correlation.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Proteínas Munc18/genética , Mutación , Adolescente , Adulto , Prueba de Desgranulación de los Basófilos , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Epistasis Genética , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Recién Nacido , Linfohistiocitosis Hemofagocítica/clasificación , Linfohistiocitosis Hemofagocítica/etnología , Masculino , Modelos Biológicos , Proteínas Munc18/fisiología , Mutación/fisiología , Proteínas Qa-SNARE/genética , Adulto Joven
16.
Br J Haematol ; 163(2): 240-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23937310

RESUMEN

Clofarabine was the latest new drug to be approved, in 2004, for relapsed or refractory acute lymphoblastic leukaemia (ALL). To investigate its value in the frontline treatment of ALL we applied clofarabine 5 × 40 mg/m(2) in combination with pegylated asparaginase (PEG-ASP) 1 × 2500 iu/m(2) in high risk ALL patients as a novel post-induction element in the German Co-operative Study Group for treatment of ALL (CoALL) trial 08-09. Newly diagnosed ALL patients, defined by a significant minimal residual disease (MRD) load at the end of induction (B-progenitor ALL at day 29 ≥ 10(-4) and T-ALL at day 43 ≥ 10(-3) ) were eligible for this phase II trial. All other patients received the standard treatment consisting of high-dose cytarabine (HIDAC) 4 × 3 g/m² in combination with Peg-ASP 2500 iu/m². Forty-two patients (39 B-progenitor; 3 T-ALL) fulfilled the criteria, were stratified and received the clofarabine/PEG-ASP treatment resulting in 24/39 (61%) MRD-negative B-progenitor patients compared to 18/39 (46%) after HIDAC/PEG-ASP in CoALL 07-03. Overall, the toxicity profile of clofarabine/PEG-ASP was similar to HIDAC/PEG-ASP without unexpected severe side effects. Clofarabine combined with PEG-ASP is safe and effective in the frontline treatment of ALL. A prospective, randomized trial is warranted to evaluate the antileukaemic efficacy of clofarabine versus HIDAC combined with PEG-ASP.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Nucleótidos de Adenina/administración & dosificación , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Arabinonucleósidos/administración & dosificación , Asparaginasa/administración & dosificación , Niño , Preescolar , Clofarabina , Esquema de Medicación , Femenino , Humanos , Masculino , Neoplasia Residual/diagnóstico , Polietilenglicoles/administración & dosificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Resultado del Tratamiento
17.
Blood ; 118(17): 4620-9, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21878672

RESUMEN

Perforin-mediated cytotoxicity is important for controlling viral infections, but also for limiting immune reactions. Failure of this cytotoxic pathway leads to hemophagocytic lymphohistiocytosis (HLH), a life-threatening disorder of uncontrolled T-cell and macrophage activation. We studied susceptibility to HLH in 2 mouse strains (souris and beige(J)) and a cohort of patients with partial defects in perforin secretion resulting from different mutations in the LYST gene. Although both strains lacked NK-cell cytotoxicity, only souris mice developed all clinical and histopathologic signs of HLH after infection with lymphocytic choriomeningitis virus. The 2 strains showed subtle differences in CTL cytotoxicity in vitro that had a large impact on virus control in vivo. Whereas beige(J) CTLs eliminated lymphocytic choriomeningitis virus infection, souris CTLs failed to control the virus, which was associated with the development of HLH. In LYST-mutant patients with Chediak-Higashi syndrome, CTL cytotoxicity was reduced in patients with early-onset HLH, whereas it was retained in patients who later or never developed HLH. Thus, the risk of HLH development is set by a threshold that is determined by subtle differences in CTL cytotoxicity. Differences in the cytotoxic capacity of CTLs may be predictive for the risk of Chediak-Higashi syndrome patients to develop HLH.


Asunto(s)
Síndrome de Chediak-Higashi/etiología , Síndrome de Chediak-Higashi/inmunología , Linfohistiocitosis Hemofagocítica/etiología , Linfohistiocitosis Hemofagocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Secuencia de Bases , Células Cultivadas , Síndrome de Chediak-Higashi/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Humanos , Individualidad , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Linfohistiocitosis Hemofagocítica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Perforina/genética , Linfocitos T Citotóxicos/fisiología , Proteínas de Transporte Vesicular/genética
18.
Pediatr Blood Cancer ; 60(7): 1215-22, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23382066

RESUMEN

BACKGROUND: Familial hemophagocytic lymphohistiocytosis (FHL) is a rare primary immune disorder defined by mutations in the syntaxin binding protein 2 (STXBP2) alias MUNC18-2. Despite defective immunity and a hyper-inflammatory state, clinical findings such as neurological, gastrointestinal, and bleeding disorders are present in a significant number of patients and suggest an impaired expression and function of STXBP2 in cells other than cytotoxic lymphocytes. PROCEDURE: We investigated four patients with FHL5 suffering from severe enteropathy and one of whom also had renal tubular dysfunction despite successful hematopoietic stem cell transplantation (HSCT). Gastrointestinal and renal biopsy specimens were analyzed by immunohistochemistry and electron microscopy. RESULTS: Histopathology revealed an intracytoplasmatic accumulation of PAS-positive granules and an enlarged intracytoplasmatic CD10-positive band along the apical pole of enterocytes. Electron microscopy revealed short microvilli and granules filled with electro lucent material. In addition, we described mildly dilated renal tubules and electron micrographs displayed a higher number of cytoplasmic inclusions, electrodense lysosomal and electrolucent endosomal vesicles. CONCLUSION: Mutations in STXBP2 do not only affect cytotoxic T lymphocytes but also cause changes in the intestinal and renal epithelium resulting in severe, osmotic diarrhea and renal proximal tubular dysfunction. These defects persist after successful treatment of hemophagocytic lymphohistocytosis by HSCT. Clinical manifestations in FHL5 patients despite successful HSCT may therefore be related to defective membrane trafficking in the gut and kidney.


Asunto(s)
Células Epiteliales/metabolismo , Linfohistiocitosis Hemofagocítica/metabolismo , Linfohistiocitosis Hemofagocítica/patología , Proteínas Munc18/genética , Membrana Celular/metabolismo , Membrana Celular/patología , Células Epiteliales/patología , Femenino , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Linfohistiocitosis Hemofagocítica/genética , Masculino , Microscopía Electrónica de Transmisión , Mutación , Linaje , Transporte de Proteínas
20.
Am J Hum Genet ; 85(4): 482-92, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19804848

RESUMEN

Rapid intracellular transport and secretion of cytotoxic granules through the immunological synapse requires a balanced interaction of several proteins. Disturbance of this highly regulated process underlies familial hemophagocytic lymphohistiocytosis (FHL), a genetically heterogeneous autosomal-recessive disorder characterized by a severe hyperinflammatory phenotype. Here, we have assigned FHL-5 to a 1 Mb region on chromosome 19p by using high-resolution SNP genotyping in eight unrelated FHL patients from consanguineous families. Subsequently, we found nine different mutations, either truncating or missense, in STXBP2 in twelve patients from Turkey, Saudi Arabia, and Central Europe. STXBP2 encodes syntaxin binding protein 2 (Munc18-2), involved in the regulation of vesicle transport to the plasma membrane. We have identified syntaxin 11, a SNARE protein mutated in FHL-4, as an interaction partner of STXBP2. This interaction is eliminated by the missense mutations found in our FHL-5 patients, which leads to a decreased stability of both proteins, as shown in patient lymphocytes. Activity of natural killer and cytotoxic T cells was markedly reduced or absent, as determined by CD107 degranulation. Our findings thus identify a key role for STXBP2 in lytic granule exocytosis.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Proteínas Munc18/genética , Proteínas Qa-SNARE/genética , Preescolar , Mapeo Cromosómico , Cromosomas Humanos Par 19 , Exocitosis , Femenino , Genotipo , Humanos , Lactante , Linfohistiocitosis Hemofagocítica/patología , Masculino , Proteínas Munc18/metabolismo , Mutación , Polimorfismo de Nucleótido Simple , Proteínas Qa-SNARE/metabolismo , Proteínas SNARE/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA