Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Transpl Infect Dis ; 22(5): e13331, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32424944

ABSTRACT

Human herpesvirus-6 (HHV-6) is a common pathogen affecting the human population. Primary HHV-6 infection generally occurs during infancy and causes exanthema subitum. Moreover, HHV-6 may exhibit inherited chromosomally integrated HHV-6 (iciHHV-6) in certain individuals. Although iciHHV-6 is generally known to be nonpathogenic, it may cause reactivation in patients with primary immunodeficiency disease (PID). XIAP deficiency is a rare PID characterized by recurrent hemophagocytic lymphohistiocytosis (HLH). It has been reported that the Epstein-Barr virus primarily causes HLH; however, the other pathogens, including HHV-6, can also cause this complication. We encountered a case of XIAP deficiency accompanied by iciHHV-6. He suffered from recurrent HLH, for which allogeneic bone marrow transplantation (BMT) was performed as a curative therapy. During the course of BMT, the patient experienced HLH three times, but there was no reactivation of endogenous HHV-6 from iciHHV-6. Finally, the patient achieved complete donor chimerism and a decline in HHV-6 DNA copy number in whole blood. This case report demonstrates no evidence of reactivation of iciHHV-6 during BMT in a patient with XIAP deficiency.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 6, Human , Genetic Diseases, X-Linked , Herpesvirus 4, Human , Humans , Lymphoproliferative Disorders , Male , Virus Integration , X-Linked Inhibitor of Apoptosis Protein
2.
Haematologica ; 104(1): 128-137, 2019 01.
Article in English | MEDLINE | ID: mdl-30171027

ABSTRACT

Fusion genes involving MEF2D have recently been identified in precursor B-cell acute lymphoblastic leukemia, mutually exclusive of the common risk stratifying genetic abnormalities, although their true incidence and associated clinical characteristics remain unknown. We identified 16 cases of acute lymphoblastic leukemia and 1 of lymphoma harboring MEF2D fusions, including MEF2D-BCL9 (n=10), MEF2D-HNRNPUL1 (n=6), and one novel MEF2D-HNRNPH1 fusion. The incidence of MEF2D fusions overall was 2.4% among consecutive precursor B-cell acute lymphoblastic leukemia patients enrolled onto a single clinical trial. They frequently showed a cytoplasmic µ chain-positive pre-B immunophenotype, and often expressed an aberrant CD5 antigen. Besides up- and down-regulation of HDAC9 and MEF2C, elevated GATA3 expression was also a characteristic feature of MEF2D fusion-positive patients. Mutations of PHF6, recurrent in T-cell acute lymphoblastic leukemia, also showed an unexpectedly high frequency (50%) in these patients. MEF2D fusion-positive patients were older (median age 9 years) with elevated WBC counts (median: 27,300/ml) at presentation and, as a result, were mostly classified as NCI high risk. Although they responded well to steroid treatment, MEF2D fusion-positive patients showed a significantly worse outcome, with 53.3% relapse and subsequent death. Stem cell transplantation was ineffective as salvage therapy. Interestingly, relapse was frequently associated with the presence of CDKN2A/CDKN2B gene deletions. Our observations indicate that MEF2D fusions comprise a distinct subgroup of precursor B-cell acute lymphoblastic leukemia with a characteristic immunophenotype and gene expression signature, associated with distinct clinical features.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins , Oncogene Proteins, Fusion , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Translocation, Genetic , Adolescent , Child , Disease-Free Survival , Female , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Survival Rate
3.
Genes Chromosomes Cancer ; 56(11): 800-809, 2017 11.
Article in English | MEDLINE | ID: mdl-28710806

ABSTRACT

High PRDM16 (also known as MEL1) expression is a representative marker of acute myeloid leukemia (AML) with NUP98-NSD1 and is a significant predictive marker for poor prognosis in pediatric AML. However, the clinical features of adult AML with PRDM16 expression remain unclear. PRDM16 is highly homologous to MDS1/EVI1, which is an alternatively spliced transcript of MECOM (also known as EVI1). We investigated PRDM16 expression in 151 AML patients, with 47 (31%) exhibiting high PRDM16 expression (PRDM16/ABL1 ratio ≥ 0.010). High PRDM16 expression significantly correlated with DNMT3A (43% vs. 15%, P < 0.001) and NPM1 (43% vs. 21%, P = 0.010) mutations and partial tandem duplication of KMT2A (22% vs. 1%, P < 0.001). Remarkably, high-PRDM16-expression patients were frequent in the noncomplete remission group (48% vs. 21%, P = 0.002). Overall survival (OS) was significantly worse in high-PRDM16-expression patients than in low-PRDM16-expression patients (5-year OS, 18% vs. 34%; P = 0.002). This trend was observed more clearly among patients aged <65 years (5-year OS, 21% vs. 50%; P = 0.001), particularly in FLT3-ITD-negative patients in the intermediate cytogenetic risk group (5-year OS, 25% vs. 59%; P = 0.009). These results suggest that high PRDM16 expression is a significant predictive marker for poor prognosis in adult AML patients, similar to pediatric AML patients.


Subject(s)
Biomarkers, Tumor/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Transcription Factors/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/metabolism , Female , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Transcription Factors/metabolism , fms-Like Tyrosine Kinase 3/genetics
4.
Genes Chromosomes Cancer ; 56(5): 394-404, 2017 05.
Article in English | MEDLINE | ID: mdl-28063190

ABSTRACT

Pediatric acute megakaryoblastic leukemia in non-Down syndrome (AMKL) is a unique subtype of acute myeloid leukemia (AML). Novel CBFA2T3-GLIS2 and NUP98-KDM5A fusions recurrently found in AMKL were recently reported as poor prognostic factors. However, their detailed clinical and molecular characteristics in patients treated with recent improved therapies remain uncertain. We analyzed molecular features of 44 AMKL patients treated on two recent Japanese AML protocols, the AML99 and AML-05 trials. We identified CBFA2T3-GLIS2, NUP98-KDM5A, RBM15-MKL1, and KMT2A rearrangements in 12 (27%), 4 (9%), 2 (5%), and 3 (7%) patients, respectively. Among 459 other AML patients, NUP98-KDM5A was identified in 3 patients, whereas CBFA2T3-GLIS2 and RBM15-MKL1 were only present in AMKL. GATA1 mutations were found in 5 patients (11%). Four-year overall survival (OS) and event-free survival (EFS) rates of CBFA2T3-GLIS2-positive patients in AMKL were 41.7% and 16.7%, respectively. Three-year cumulative incidence of relapse in CBFA2T3-GLIS2-positive patients was significantly higher than that of CBFA2T3-GLIS2-negative patients (75.0% vs. 35.7%, P = 0.024). In multivariate analyses, CBFA2T3-GLIS2 was an independent poor prognostic factor for OS (HR, 4.34; 95% CI, 1.31-14.38) and EFS (HR, 2.95; 95% CI, 1.20-7.23). Furthermore, seven (54%) of 13 infant AMKL patients were CBFA2T3-GLIS2-positive. Notably, out of 7 CBFA2T3-GLIS2-positive infants, six (86%) relapsed and five (71%) died. Moreover, all of CBFA2T3-GLIS2-positive patients who experienced induction failure (n = 3) were infants, indicating worse prognosis of CBFA2T3-GLIS2-positive infants. These findings indicated the significance of CBFA2T3-GLIS2 as a poor prognostic factor in AMKL patients, particularly in infants.


Subject(s)
Biomarkers, Tumor/genetics , Down Syndrome/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Mutation/genetics , Oncogene Proteins, Fusion/genetics , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Leukemia, Megakaryoblastic, Acute/pathology , Male , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate
5.
Genes Chromosomes Cancer ; 56(5): 382-393, 2017 05.
Article in English | MEDLINE | ID: mdl-28063196

ABSTRACT

ASXL2 is an epigenetic regulator involved in polycomb repressive complex regulation or recruitment. Clinical features of pediatric acute myeloid leukemia (AML) patients with ASXL2 mutations remain unclear. Thus, we investigated frequencies of ASXL1 and ASXL2 mutations, clinical features of patients with these mutations, correlations of these mutations with other genetic alterations including BCOR/BCORL1 and cohesin complex component genes, and prognostic impact of these mutations in 369 pediatric patients with de novo AML (0-17 years). We identified 9 (2.4%) ASXL1 and 17 (4.6%) ASXL2 mutations in 25 patients. These mutations were more common in patients with t(8;21)(q22;q22)/RUNX1-RUNX1T1 (ASXL1, 6/9, 67%, P = 0.02; ASXL2, 10/17, 59%, P = 0.01). Among these 25 patients, 4 (27%) of 15 patients with t(8;21) and 6 (60%) of 10 patients without t(8;21) relapsed. However, most patients with relapse were rescued using stem cell transplantation irrespective of t(8;21). The overall survival (OS) and event-free survival (EFS) rates showed no differences among pediatric AML patients with t(8;21) and ASXL1 or ASXL2 mutations and ASXL wild-type (5-year OS, 75% vs. 100% vs. 91% and 5-year EFS, 67% vs. 80% vs. 67%). In 106 patients with t(8;21) AML, the coexistence of mutations in tyrosine kinase pathways and chromatin modifiers and/or cohesin complex component genes had no effect on prognosis. These results suggest that ASXL1 and ASXL2 mutations play key roles as cooperating mutations that induce leukemogenesis, particularly in pediatric AML patients with t(8;21), and these mutations might be associated with a better prognosis than that reported previously.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Repressor Proteins/genetics , Translocation, Genetic/genetics , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/pathology , Male , Neoplasm Staging , Prognosis , RUNX1 Translocation Partner 1 Protein , Survival Rate
6.
Br J Haematol ; 172(4): 581-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26684393

ABSTRACT

Recent reports described the NUP98-NSD1 fusion as an adverse prognostic marker for acute myeloid leukaemia (AML) and PRDM16 (also known as MEL1) as the representative overexpressed gene in patients harbouring NUP98-NSD1 fusion. PRDM16 gene expression levels were measured via real-time polymerase chain reaction in 369 paediatric patients with de novo AML, of whom 84 (23%) exhibited PRDM16 overexpression (PRDM16/ABL1 ratio ≥0·010). The frequencies of patients with high or low PRDM16 expression differed widely with respect to each genetic alteration, as follows: t(8;21), 4% vs. 96%, P < 0·001; inv(16), 0% vs. 100%, P < 0·001; KMT2A (also termed MLL)- partial tandem duplication, 100% vs. 0%, P < 0·001; NUP98-NSD1, 100% vs. 0%, P < 0·001. The overall survival (OS) and event-free survival (EFS) among PRDM16-overexpressing patients were significantly worse than in patients with low PRDM16 expression (3-year OS: 51% vs. 81%, P < 0·001, 3-year EFS: 32% vs. 64%, P < 0·001) irrespective of other cytogenetic alterations except for NPM1. PRDM16 gene expression was particularly useful for stratifying FLT3-internal tandem duplication-positive AML patients (3-year OS: high = 30% vs. low = 70%, P < 0·001). PRDM16 overexpression was highly recurrent in de novo paediatric AML patients with high/intermediate-risk cytogenetic profiles and was independently associated with an adverse outcome.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Transcription Factors/genetics , Adolescent , Child , Child, Preschool , Female , Gene Expression/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Infant , Infant, Newborn , Japan/epidemiology , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Male , Mutation/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Pore Complex Proteins/genetics , Nucleophosmin , Prognosis , fms-Like Tyrosine Kinase 3/genetics
7.
Br J Haematol ; 175(3): 476-489, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27470916

ABSTRACT

Acute myeloid leukaemia (AML) is a molecularly and clinically heterogeneous disease. Targeted sequencing efforts have identified several mutations with diagnostic and prognostic values in KIT, NPM1, CEBPA and FLT3 in both adult and paediatric AML. In addition, massively parallel sequencing enabled the discovery of recurrent mutations (i.e. IDH1/2 and DNMT3A) in adult AML. In this study, whole-exome sequencing (WES) of 22 paediatric AML patients revealed mutations in components of the cohesin complex (RAD21 and SMC3), BCORL1 and ASXL2 in addition to previously known gene mutations. We also revealed intratumoural heterogeneities in many patients, implicating multiple clonal evolution events in the development of AML. Furthermore, targeted deep sequencing in 182 paediatric AML patients identified three major categories of recurrently mutated genes: cohesion complex genes [STAG2, RAD21 and SMC3 in 17 patients (8·3%)], epigenetic regulators [ASXL1/ASXL2 in 17 patients (8·3%), BCOR/BCORL1 in 7 patients (3·4%)] and signalling molecules. We also performed WES in four patients with relapsed AML. Relapsed AML evolved from one of the subclones at the initial phase and was accompanied by many additional mutations, including common driver mutations that were absent or existed only with lower allele frequency in the diagnostic samples, indicating a multistep process causing leukaemia recurrence.


Subject(s)
Clonal Evolution/genetics , Exome , High-Throughput Nucleotide Sequencing , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Mutation , Child , DNA Mutational Analysis , Disease Progression , Epigenesis, Genetic , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Leukemia, Myeloid, Acute/mortality , Male , Nucleophosmin , Prognosis , Recurrence
8.
Br J Haematol ; 170(3): 391-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25858548

ABSTRACT

Mutations in the colony-stimulating factor 3 receptor (CSF3R) and calreticulin (CALR) genes have been reported in a proportion of adults with myeloproliferative disease. However, little is known about CSF3R or CALR mutations in paediatric myeloid disorders. We analysed CSF3R exons 14 and 17, and CALR exon 9, using direct sequencing in samples of paediatric acute myeloid leukaemia (AML; n = 521), juvenile myelomonocytic leukaemia (JMML; n = 40), myelodysplastic syndrome (MDS; n = 20) and essential thrombocythaemia (ET; n = 21). CSF3R mutations were found in 10 (1.2%) of 521 patients with AML; two in exon 14 (both missense mutations resulting in p.T618I) and eight in exon 17 (three frameshift mutations: p.S715X, p.Q774R, and p.S783Q; and five novel missense mutations: p.Q754K, p.R769H, p.L777F, p.T781I, and S795R). All of the patients with mutations in CSF3R exon 17 had chromosomal translocations, including four with t(8;21). At the time of reporting, seven of these ten patients are alive; three have died, due to side effects of chemotherapy. No CSF3R mutations were found in cases of MDS, JMML or ET. The only mutation found in the CALR gene was a frameshift (p.L367 fs) in one ET patient. We discuss the potential impact of these findings for the leukaemogenesis and clinical features of paediatric myeloid disorders.


Subject(s)
Calreticulin/genetics , Frameshift Mutation , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Receptors, Colony-Stimulating Factor/genetics , Thrombocythemia, Essential/genetics , Translocation, Genetic , Adolescent , Adult , Child , Child, Preschool , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Exons , Female , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/mortality , Male , Myelodysplastic Syndromes/mortality , Thrombocythemia, Essential/mortality
10.
Blood ; 119(11): 2608-11, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22294728

ABSTRACT

Trib1 has been identified as a myeloid oncogene in a murine leukemia model. Here we identified a TRIB1 somatic mutation in a human case of Down syndrome-related acute megakaryocytic leukemia. The mutation was observed at well-conserved arginine 107 residue in the pseudokinase domain. This R107L mutation remained in leukocytes of the remission stage in which GATA1 mutation disappeared, suggesting the TRIB1 mutation is an earlier genetic event in leukemogenesis. The bone marrow transfer experiment showed that acute myeloid leukemia development was accelerated by transducing murine bone marrow cells with the R107L mutant in which enhancement of ERK phosphorylation and C/EBPα degradation by Trib1 expression was even greater than in those expressing wild-type. These results suggest that TRIB1 may be a novel important oncogene for Down syndrome-related acute megakaryocytic leukemia.


Subject(s)
Down Syndrome/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Megakaryoblastic, Acute/etiology , Mutation/genetics , Oncogenes/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Down Syndrome/complications , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunoblotting , Leukemia, Megakaryoblastic, Acute/pathology , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
11.
Blood ; 119(11): 2612-4, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22138511

ABSTRACT

Familial platelet disorder with a propensity to develop acute myeloid leukemia (FPD/AML) is a rare autosomal dominant disease characterized by thrombocytopenia, abnormal platelet function, and a propensity to develop myelodysplastic syndrome (MDS) and AML. So far, > 20 affected families have been reported. Recently, a second RUNX1 alteration has been reported; however, no additional molecular abnormalities have been found so far. We identified an acquired CBL mutation and 11q-acquired uniparental disomy (11q-aUPD) in a patient with chronic myelomonocytic leukemia (CMML) secondary to FPD with RUNX1 mutation but not in the same patient during refractory cytopenia. This finding suggests that alterations of the CBL gene and RUNX1 gene may cooperate in the pathogenesis of CMML in patients with FPD/AML. The presence of CBL mutations and 11q-aUPD was an important "second hit" that could be an indicator of leukemic transformation of MDS or AML in patients with FPD/AML.


Subject(s)
Blood Platelet Disorders/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Genetic Predisposition to Disease , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Chronic/etiology , Mutation/genetics , Proto-Oncogene Proteins c-cbl/genetics , Adult , Blood Platelet Disorders/complications , Child , Chromosomes, Human, Pair 11/genetics , Female , Genes, Dominant , Haplotypes/genetics , Humans , Immunoblotting , Leukemia, Myeloid, Acute/complications , Male , Middle Aged , Pedigree , Polymerase Chain Reaction
12.
J Immunol ; 189(3): 1521-6, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22730530

ABSTRACT

Chronic mucocutaneous candidiasis (CMC) is a heterogeneous group of primary immunodeficiency diseases characterized by chronic and recurrent Candida infections of the skin, nails, and oropharynx. Gain-of-function mutations in STAT1 were very recently shown to be responsible for autosomal-dominant or sporadic cases of CMC. The reported mutations have been exclusively localized in the coiled-coil domain, resulting in impaired dephosphorylation of STAT1. However, recent crystallographic analysis and direct mutagenesis experiments indicate that mutations affecting the DNA-binding domain of STAT1 could also lead to persistent phosphorylation of STAT1. To our knowledge, this study shows for the first time that a DNA-binding domain mutation of c.1153C>T in exon 14 (p.T385M) is the genetic cause of sporadic CMC in two unrelated Japanese patients. The underlying mechanisms involve a gain of STAT1 function due to impaired dephosphorylation as observed in the coiled-coil domain mutations.


Subject(s)
Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/immunology , DNA-Binding Proteins/genetics , Mutation/immunology , STAT1 Transcription Factor/genetics , Adolescent , Amino Acid Sequence , Asian People , Candidiasis, Chronic Mucocutaneous/metabolism , Cell Line, Transformed , Child , Female , Humans , Male , Molecular Sequence Data , Mutation/genetics , Phosphorylation/genetics , Phosphorylation/immunology , Protein Structure, Tertiary/genetics
13.
Genes Chromosomes Cancer ; 52(7): 683-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23630019

ABSTRACT

The cryptic t(5;11)(q35;p15.5) creates a fusion gene between the NUP98 and NSD1 genes. To ascertain the significance of this gene fusion, we explored its frequency, clinical impact, and gene expression pattern using DNA microarray in pediatric acute myeloid leukemia (AML) patients. NUP98-NSD1 fusion transcripts were detected in 6 (4.8%) of 124 pediatric AML patients. Supervised hierarchical clustering analyses using probe sets that were differentially expressed in these patients detected a characteristic gene expression pattern, including 18 NUP98-NSD1-negative patients (NUP98-NSD1-like patients). In total, a NUP98-NSD1-related gene expression signature (NUP98-NSD1 signature) was found in 19% (24/124) and in 58% (15/26) of cytogenetically normal cases. Their 4-year overall survival (OS) and event-free survival (EFS) were poor (33.3% in NUP98-NSD1-positive and 38.9% in NUP98-NSD1-like patients) compared with 100 NUP98-NSD1 signature-negative patients (4-year OS: 86.0%, 4-year EFS: 72.0%). Interestingly, t(7;11)(p15;p15)/NUP98-HOXA13, t(6;11)(q27;q23)/MLL-MLLT4 and t(6;9)(p22;q34)/DEK-NUP214, which are known as poor prognostic markers, were found in NUP98-NSD1-like patients. Furthermore, another type of NUP98-NSD1 fusion transcript was identified by additional RT-PCR analyses using other primers in a NUP98-NSD1-like patient, revealing the significance of this signature to detect NUP98-NSD1 gene fusions and to identify a new poor prognostic subgroup in AML.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Translocation, Genetic/genetics , Adolescent , Child , Child, Preschool , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 5/genetics , Disease-Free Survival , Female , Follow-Up Studies , Homeodomain Proteins , Humans , Leukemia, Myeloid, Acute/pathology , Male , Prognosis
14.
Nephrol Dial Transplant ; 28(8): 2123-30, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23729491

ABSTRACT

BACKGROUND: Mutations in the ATP6V1B1 and the ATP6V0A4 genes cause primary autosomal-recessive distal renal tubular acidosis (dRTA). Large deletions of either gene in patients with dRTA have not been described. METHODS: The ATP6V1B1 and ATP6V0A4 genes were directly sequenced in 11 Japanese patients with primary dRTA from nine unrelated kindreds. Large heterozygous deletions were analyzed by quantitative real-time polymerase chain reaction (PCR). The clinical features of the 11 patients were also investigated. RESULTS: Novel mutations in the ATP6V1B1 gene were identified in two kindreds, including frameshift, in-frame insertion and nonsense mutations. Large deletions in the ATP6V0A4 gene were identified in two kindreds. Exon 15 of ATP6V0A4 was not amplified in one patient, with a long PCR confirming compound heterozygous deletions of 3.7- and 6.9-kb nucleotides, including all of exon 15. Direct DNA sequencing revealed a heterozygous frameshift mutation in ATP6V0A4 in another patient, with quantitative real-time PCR indicating that all exons up to exon 8 were deleted in one allele. Clinical investigation showed that four of the six patients with available clinical data presented with hyperammonemia at onset. CONCLUSIONS: To our knowledge, these dRTA patients are the first to show large deletions involving one or more entire exons of the ATP6V0A4 gene. Quantitative PCR amplification may be useful in detecting heterozygous large deletions. These results expand the spectrum of mutations in the ATP6V0A4 and ATP6V1B1 genes associated with primary dRTA and provide insight into possible structure-function relationships.


Subject(s)
Acidosis, Renal Tubular/genetics , Exons/genetics , Mutation/genetics , Vacuolar Proton-Translocating ATPases/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Family , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Young Adult
15.
J Pediatr Hematol Oncol ; 35(7): 554-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23389504

ABSTRACT

BACKGROUND: Kasabach-Merritt phenomenon (KMP) is a rare condition and optimal treatments have not yet been established, especially for cases that are unresponsive to first-line therapy. We retrospectively reviewed 11 KMP cases treated over the past 13 years in our institute. OBSERVATIONS: With the exception of 1 case, steroids were administered as the first-line therapy. Eight cases required second-line or third-line therapy. The effective salvage therapies include interferon (n=1), radiotherapy (n=1), and chemotherapy (n=5). One case continues to depend upon chemotherapy. Three refractory cases were therapy dependent over 1 year of age, whereas 8 were treated effectively by 6 months of age. CONCLUSIONS: Chemotherapy seems to be the most effective therapy for steroid-resistant KMP cases.


Subject(s)
Kasabach-Merritt Syndrome/diagnosis , Combined Modality Therapy/adverse effects , Female , Humans , Infant , Infant, Newborn , Kasabach-Merritt Syndrome/blood , Kasabach-Merritt Syndrome/therapy , Male , Retrospective Studies , Treatment Outcome
16.
Rinsho Ketsueki ; 53(12): 1997-2002, 2012 Dec.
Article in Japanese | MEDLINE | ID: mdl-23318965

ABSTRACT

Recently, bone marrow transplantation from an unrelated donor has been recommended as an option for the treatment of very severe aplastic anemia (vSAA) refractory to immunosuppressive therapy (IST) in the absence of a human leukocyte antigen-matched related donor (MRD). For SAA patients with complications such as bacterial infections, prompt transplantation using either a mismatched related donor or cord blood (CB) becomes necessary. However, the former option is associated with graft-versus-host disease, whereas the latter option is associated with a more significant risk of graft failure. We report a patient with vSAA refractory to IST that was repeatedly complicated by bacterial infection. An MRD was unavailable for this patient, so we decided on emergent transplantation and successfully performed CB transplantation (CBT) using a low-dose TBI conditioning regimen. Although it may be necessary to examine more clinical cases in the future, a conditioning regimen such as that involving low-dose TBI may decrease the risk of graft failure, and CBT may become a treatment option for vSAA in the absence of a suitable donor.


Subject(s)
Anemia, Aplastic/therapy , Fetal Blood/transplantation , Child , Graft vs Host Disease/immunology , HLA Antigens/blood , Humans , Male , Transplantation, Heterologous , Treatment Outcome
17.
Int J Hematol ; 113(5): 662-667, 2021 May.
Article in English | MEDLINE | ID: mdl-33394336

ABSTRACT

Transient abnormal myelopoiesis (TAM) is a unique clonal myeloproliferation characterized by immature megakaryoblasts that occurs in 5-10% of neonates with Down syndrome (DS). Although TAM regresses spontaneously in most patients, approximately 20% of TAM cases result in early death, and approximately 20% of survivors develop acute megakaryoblastic leukemia (AMKL). We retrospectively reviewed records of 35 DS patients with TAM to determine the correlation between clinical characteristics and blast percentage. Thirteen of the 35 patients were classified as low blast percentage TAM (LBP-TAM), defined as TAM with a peak peripheral blast percentage ≤ 10%. Although no patient with LBP-TAM experienced systemic edema, disseminated intravascular coagulation, or early death, eight patients had elevated direct bilirubin levels (> 2 mg/dl) and one developed AMKL. All patients with LBP-TAM had serum markers of liver fibrosis that exceeded the normal limits, and two patients underwent liver biopsy to clarify the etiology of pathological jaundice. Taken together, our results suggest that patients with LBP-TAM may be at risk of liver fibrosis and liver failure, similarly to patients with classical TAM. Although these patients generally have a good prognosis, they should be carefully monitored for potential development of liver disease and leukemia.


Subject(s)
Down Syndrome/complications , Leukemoid Reaction/complications , Adolescent , Adult , Child , Child, Preschool , Down Syndrome/blood , Female , Humans , Infant , Leukemoid Reaction/blood , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Male , Retrospective Studies , Young Adult
18.
Haematologica ; 95(8): 1293-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20378560

ABSTRACT

BACKGROUND: Diamond-Blackfan anemia is a rare, clinically heterogeneous, congenital red cell aplasia: 40% of patients have congenital abnormalities. Recent studies have shown that in western countries, the disease is associated with heterozygous mutations in the ribosomal protein (RP) genes in about 50% of patients. There have been no studies to determine the incidence of these mutations in Asian patients with Diamond-Blackfan anemia. DESIGN AND METHODS: We screened 49 Japanese patients with Diamond-Blackfan anemia (45 probands) for mutations in the six known genes associated with Diamond-Blackfan anemia: RPS19, RPS24, RPS17, RPL5, RPL11, and RPL35A. RPS14 was also examined due to its implied involvement in 5q- syndrome. RESULTS: Mutations in RPS19, RPL5, RPL11 and RPS17 were identified in five, four, two and one of the probands, respectively. In total, 12 (27%) of the Japanese Diamond-Blackfan anemia patients had mutations in ribosomal protein genes. No mutations were detected in RPS14, RPS24 or RPL35A. All patients with RPS19 and RPL5 mutations had physical abnormalities. Remarkably, cleft palate was seen in two patients with RPL5 mutations, and thumb anomalies were seen in six patients with an RPS19 or RPL5 mutation. In contrast, a small-for-date phenotype was seen in five patients without an RPL5 mutation. CONCLUSIONS: We observed a slightly lower frequency of mutations in the ribosomal protein genes in patients with Diamond-Blackfan anemia compared to the frequency reported in western countries. Genotype-phenotype data suggest an association between anomalies and RPS19 mutations, and a negative association between small-for-date phenotype and RPL5 mutations.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Mutation , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/drug therapy , Anemia, Diamond-Blackfan/ethnology , Asian People/genetics , Child , Female , Genetic Association Studies , Humans , Japan , Male , Steroids/therapeutic use , Treatment Outcome
19.
Pediatr Blood Cancer ; 55(7): 1406-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20730882

ABSTRACT

A 6-year-old Japanese female was diagnosed as having myeloid/NK cell precursor acute leukemia (MNKL) using immunocytochemical analysis. The patient was treated by cord blood transplantation from an HLA 1-locus mismatched unrelated donor after chemotherapy comprising cytosine arabinoside, idarubicin, etoposide, and L-asparaginase. We detected a nonsense mutation, C7412A, resulting in S2471X, where X is a terminal codon, in the PEST domain of NOTCH1 in this patient. The presence of the NOTCH1 activating mutation in MNKL might suggest a possible role in the leukemogenesis of MNKL.


Subject(s)
Codon, Nonsense , Killer Cells, Natural , Leukemia, Myeloid, Acute/genetics , Receptor, Notch1/genetics , Antineoplastic Agents , Asparaginase/therapeutic use , Child , Cord Blood Stem Cell Transplantation , Female , Humans , Leukemia, Myeloid, Acute/therapy
SELECTION OF CITATIONS
SEARCH DETAIL