Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 97
2.
Ann Hematol ; 98(6): 1421-1426, 2019 Jun.
Article En | MEDLINE | ID: mdl-30919072

Hydroxyurea (HU) resistance or intolerance occurs in 15 to 24% of patients with polycythemia vera (PV). Resistance to HU is associated with a shortened life expectancy, intolerance has no prognostic value. We assessed the occurrence of HU resistance or intolerance comparing the original (ELNo) versus the modified European Leukemia Net (ELNm) criteria as applied in recent large clinical trials including PV patients. We retrospectively analyzed 106 patients with PV treated with HU at the University Hospitals of Leuven between 1990 and 2016 for occurrence of HU resistance/intolerance when using both ELNo as ELNm. After a mean duration of treatment of 5.1 years, when applying the ELNo 20.7% of patients had shown resistance or intolerance to HU in comparison to 39.6% when using the ELNm. When using the ELNo 4.7% of patients were resistant to HU versus 23.6% when applying the ELNm. In total, 16.0% of patients were HU intolerant. This rate was identical when using both ELNo and ELNm. 20.7% of PV patients were considered as HU-resistant or intolerant when using the original ELN criteria. However, when applying the modified ELN criteria 39.6% of PV patients were resistant or intolerant to HU. In our hands, no patient received a minimum dose of 2 g HU a day, as such the ELNm seem better adapted for daily clinical use. However, the prognostic value of HU-resistance in PV, when defined by the ELNm, still needs to be confirmed.


Drug Resistance , Hydroxyurea/therapeutic use , Polycythemia Vera/drug therapy , Adult , Aged , Aged, 80 and over , Female , Fever/chemically induced , Follow-Up Studies , Humans , Hydroxyurea/adverse effects , Hydroxyurea/pharmacology , Leg Ulcer/chemically induced , Male , Middle Aged , Mucositis/chemically induced , Polycythemia Vera/complications , Polycythemia Vera/mortality , Prognosis , Retrospective Studies , Thrombosis/epidemiology , Thrombosis/etiology
3.
Leukemia ; 33(8): 1910-1922, 2019 08.
Article En | MEDLINE | ID: mdl-30858550

Minimal residual disease (MRD) is a powerful prognostic factor in acute lymphoblastic leukemia (ALL) and is used for patient stratification and treatment decisions, but its precise role in Philadelphia chromosome positive ALL is less clear. This uncertainty results largely from methodological differences relating to the use of real-time quantitative PCR (qRT-PCR) to measure BCR-ABL1 transcript levels for MRD analysis. We here describe the first results by the EURO-MRD consortium on standardization of qRT-PCR for the e1a2 BCR-ABL1 transcript in Ph + ALL, designed to overcome the lack of standardisation of laboratory procedures and data interpretation. Standardised use of EAC primer/probe sets and of centrally prepared plasmid standards had the greatest impact on reducing interlaboratory variability. In QC1 the proportion of analyses with BCR-ABL1/ABL1 ratios within half a log difference were 40/67 (60%) and 52/67 (78%) at 10-3 and 36/67 (53%) and 53/67 (79%) at 10-4BCR-ABL1/ABL1. Standardized RNA extraction, cDNA synthesis and cycler platforms did not improve results further, whereas stringent application of technical criteria for assay quality and uniform criteria for data interpretation and reporting were essential. We provide detailed laboratory recommendations for the standardized MRD analysis in routine diagnostic settings and in multicenter clinical trials for Ph + ALL.


Fusion Proteins, bcr-abl/genetics , Philadelphia Chromosome , Practice Guidelines as Topic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Real-Time Polymerase Chain Reaction/methods , Consensus , Humans , Neoplasm, Residual , RNA, Messenger/analysis
4.
Ann Oncol ; 30(1): 85-95, 2019 01 01.
Article En | MEDLINE | ID: mdl-30371735

Background: Early cancer diagnosis might improve survival rates. As circulating tumor DNA (ctDNA) carries cancer-specific modifications, it has great potential as a noninvasive biomarker for detection of incipient tumors. Patients and methods: We collected cell-free DNA (cfDNA) samples of 1002 elderly without a prior malignancy, carried out whole-genome massive parallel sequencing and scrutinized the mapped sequences for the presence of (sub)chromosomal copy number alterations (CNAs) predictive for a malignancy. When imbalances were detected, 6-monthly clinical follow-up was carried out. Results: In 3% of participants chromosomal imbalances were detected. Follow-up analyses, including whole-body MRI screening, confirmed the presence of five hematologic malignancies: one Hodgkin lymphoma (HL), stage II; three non-HL (type chronic lymphocytic leukemia, Rai I-Binet A; type SLL, stage III; type mucosa-associated lymphoid tissue, stage I) and one myelodysplastic syndrome with excess blasts, stage II. The CNAs detected in cfDNA were tumor-specific. Furthermore, one case was identified with monoclonal B-cell lymphocytosis, a potential precursor of B-cell malignancy. In 24 additional individuals, CNAs were identified but no cancer diagnosis was made. For 9 of them, the aberrant cfDNA profile originated from peripheral blood cells. For 15 others the origin of aberrations in cfDNA remains undetermined. Conclusion(s): Genomewide profiling of cfDNA in apparently healthy individuals enables the detection of incipient hematologic malignancies as well as clonal mosaicism with unknown clinical significance. CNA screening of cellular DNA of peripheral blood in elderly has established that clonal mosaicism for these chromosomal anomalies predicts a 5- to 10-fold enhanced risk of a subsequent cancer. We demonstrate that cfDNA screening detects CNAs, which are not only derived from peripheral blood, but even more from other tissues. Since the clinical relevance of clonal mosaics in other tissues remains unknown, long-term follow-up is warranted. Taken together, this study demonstrates that genomewide cfDNA analysis has potential as an unbiased screening approach for hematological malignancies and premalignant conditions.


Circulating Tumor DNA/analysis , DNA Copy Number Variations , DNA, Neoplasm/analysis , Early Detection of Cancer/methods , Neoplasms/diagnosis , Neoplasms/genetics , Whole Genome Sequencing/methods , Aged , Aged, 80 and over , Circulating Tumor DNA/genetics , Cohort Studies , DNA, Neoplasm/genetics , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Neoplasms/blood , Prognosis
5.
Hematology ; 23(10): 785-792, 2018 Dec.
Article En | MEDLINE | ID: mdl-29993347

OBJECTIVES: CML, PV, ET and PMF are so called classical MPN with distinct clinical phenotypes. The discovery of the BCR-ABL1 translocation and mutations in driver genes JAK2, MPL and CALR has provided novel insights in their pathogenesis. While these mutations are thought to be mutually exclusive, rare cases of MPN with coexisting driver mutations have been reported. However, little is known about the clinical, biological and molecular characteristics of these patients and the interaction of the neoplastic clones. METHODS: We retrospectively studied 11 MPN patients with coexisting driver mutations (JAK2 V617F + BCR-ABL1: n = 8; CALR type 2 + BCR-ABL1: n = 1; JAK2 V617F + MPL W515: n = 1; JAK2 V617F + CALR type 1: n = 1). To assess possible associated molecular aberrations, we analysed DNA of six patients using NGS. RESULTS: In four CML patients, decreasing BCR-ABL1 transcript levels with increasing JAK2 V617F allele burden under TKI were observed. This strongly suggests that the coexistence of driver mutations originates from two different clones growing independently. Additional somatic mutations were detected in 5 out of 6 (83%) patients affecting 4 different genes, confirming the heterogeneity of this study cohort. Suboptimal response to TKI was observed with a higher frequency (4/8 patients) than reported in conventional series of CML and the overall tolerance of treatment with hydroxyurea and/or imatinib in our series was poor. CONCLUSION: Given the emergence of NGS in clinical practice, more similar cases will be identified in the coming years. The optimal treatment strategy for this rare group of patients is uncertain and toxicity of combination treatment may have to be considered.


Drug Resistance, Neoplasm/genetics , Hematologic Neoplasms/genetics , Mutation , Myeloproliferative Disorders/genetics , Neoplasm Proteins/genetics , Adult , Aged , Female , Hematologic Neoplasms/drug therapy , Humans , Hydroxyurea , Male , Middle Aged , Myeloproliferative Disorders/drug therapy
6.
Am J Transplant ; 18(12): 3007-3020, 2018 12.
Article En | MEDLINE | ID: mdl-29734503

Acute graft-versus-host disease (GVHD) after liver transplant (LTx) is a rare complication with a high mortality rate. Recently, monoclonal antibody (mAb) treatment, specifically with anti-interleukin 2 receptor antibodies (IL2RAb) and anti-tumor necrosis factor-α antibodies (TNFAb), has gained increasing interest. However, evidence is mostly limited to case reports and the efficacy remains unclear. Here, we describe 5 patients with LTx-associated GVHD from our center and provide the results of our systematic literature review to evaluate the potential therapeutic benefit of IL2RAb/TNFAb treatment. Of the combined population of 155 patients (5 in our center and 150 through systematic search), 24 were given mAb (15.5%)-4 with TNFAb (2.6%) and 17 with IL2RAb (11%) ("mAb group")-and compared with patients who received other treatments (referred to as "no-mAb group"). Two-sided Fisher exact tests revealed a better survival when comparing treatment with mAb versus no-mAb (11/24 vs 27/131; P = .018), TNFAb versus no-mAb (3/4 vs 27/131; P = .034), and IL2RAb versus no-mAb (8/17 vs 27/131; P = .029). This systematic review suggests a beneficial effect of mAb treatment and a promising role for TNFAb and IL2RAb as a first-line strategy to treat LTx-associated acute GVHD.


Antibodies, Monoclonal/therapeutic use , Graft Rejection/mortality , Graft vs Host Disease/mortality , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Liver Transplantation/mortality , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Female , Follow-Up Studies , Graft Rejection/drug therapy , Graft Rejection/etiology , Graft Survival , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Humans , Liver Transplantation/adverse effects , Male , Middle Aged , Postoperative Complications , Prognosis , Retrospective Studies , Risk Factors , Survival Rate
7.
Ann Oncol ; 29(1): 200-208, 2018 01 01.
Article En | MEDLINE | ID: mdl-29186353

Background: Reported prevalence of driver gene mutations in non-small-cell lung cancer (NSCLC) is highly variable and clinical correlations are emerging. Using NSCLC biomaterial and clinical data from the European Thoracic Oncology Platform Lungscape iBiobank, we explore the epidemiology of mutations and association to clinicopathologic features and patient outcome (relapse-free survival, time-to-relapse, overall survival). Methods: Clinically annotated, resected stage I-III NSCLC FFPE tissue was assessed for gene mutation using a microfluidics-based multiplex PCR platform. Mutant-allele detection sensitivity is >1% for most of the ∼150 (13 genes) mutations covered in the multiplex test. Results: Multiplex testing has been carried out in 2063 (76.2%) of the 2709 Lungscape cases (median follow-up 4.8 years). FFPE samples mostly date from 2005 to 2008, yet recently extracted DNA quality and quantity was generally good. Average DNA yield/case was 2.63 µg; 38 cases (1.4%) failed QC and were excluded from study; 95.1% of included cases allowed the complete panel of mutations to be tested. Most common were KRAS, MET, EGFR and PIK3CA mutations with overall prevalence of 23.0%, 6.8%, 5.4% and 4.9%, respectively. KRAS and EGFR mutations were significantly more frequent in adenocarcinomas: PIK3CA in squamous cell carcinomas. MET mutation prevalence did not differ between histology groups. EGFR mutations were found predominantly in never smokers; KRAS in current/former smokers. For all the above mutations, there was no difference in outcome between mutated and non-mutated cases. Conclusion: Archival FFPE NSCLC material is adequate for multiplex mutation analysis. In this large, predominantly European, clinically annotated stage I-III NSCLC cohort, none of the mutations characterized showed prognostic significance.


Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Mutation , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase/biosynthesis , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/pathology , DNA Mutational Analysis/methods , Female , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Male , Middle Aged , Multiplex Polymerase Chain Reaction/methods , Neoplasm Staging , Prevalence , Progression-Free Survival , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/genetics , Smoking/genetics , Young Adult
8.
Leukemia ; 31(8): 1706-1714, 2017 08.
Article En | MEDLINE | ID: mdl-27909306

Chromosomal region 1p22 is deleted in ⩾20% of multiple myeloma (MM) patients, suggesting the presence of an unidentified tumor suppressor. Using high-resolution genomic profiling, we delimit a 58 kb minimal deleted region (MDR) on 1p22.1 encompassing two genes: ectopic viral integration site 5 (EVI5) and ribosomal protein L5 (RPL5). Low mRNA expression of EVI5 and RPL5 was associated with worse survival in diagnostic cases. Patients with 1p22 deletion had lower mRNA expression of EVI5 and RPL5, however, 1p22 deletion status is a bad predictor of RPL5 expression in some cases, suggesting that other mechanisms downregulate RPL5 expression. Interestingly, RPL5 but not EVI5 mRNA levels were significantly lower in relapsed patients responding to bortezomib and; both in newly diagnosed and relapsed patients, bortezomib treatment could overcome their bad prognosis by raising their progression-free survival to equal that of patients with high RPL5 expression. In conclusion, our genetic data restrict the MDR on 1p22 to EVI5 and RPL5 and although the role of these genes in promoting MM progression remains to be determined, we identify RPL5 mRNA expression as a biomarker for initial response to bortezomib in relapsed patients and subsequent survival benefit after long-term treatment in newly diagnosed and relapsed patients.


Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 1 , Multiple Myeloma/genetics , Ribosomal Proteins/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Humans , MDS1 and EVI1 Complex Locus Protein , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Mutation , Proto-Oncogenes/genetics , RNA, Messenger/analysis , Transcription Factors/genetics
9.
Leukemia ; 30(9): 1824-31, 2016 09.
Article En | MEDLINE | ID: mdl-27168466

Intrachromosomal amplification of chromosome 21 (iAMP21) identifies a high-risk subtype of acute lymphoblastic leukaemia (ALL), requiring intensive treatment to reduce their relapse risk. Improved understanding of the genomic landscape of iAMP21-ALL will ascertain whether these patients may benefit from targeted therapy. We performed whole-exome sequencing of eight iAMP21-ALL samples. The mutation rate was dramatically disparate between cases (average 24.9, range 5-51) and a large number of novel variants were identified, including frequent mutation of the RAS/MEK/ERK pathway. Targeted sequencing of a larger cohort revealed that 60% (25/42) of diagnostic iAMP21-ALL samples harboured 42 distinct RAS pathway mutations. High sequencing coverage demonstrated heterogeneity in the form of multiple RAS pathway mutations within the same sample and diverse variant allele frequencies (VAFs) (2-52%), similar to other subtypes of ALL. Constitutive RAS pathway activation was observed in iAMP21 samples that harboured mutations in the predominant clone (⩾35% VAF). Viable iAMP21 cells from primary xenografts showed reduced viability in response to the MEK1/2 inhibitor, selumetinib, in vitro. As clonal (⩾35% VAF) mutations were detected in 26% (11/42) of iAMP21-ALL, this evidence of response to RAS pathway inhibitors may offer the possibility to introduce targeted therapy to improve therapeutic efficacy in these high-risk patients.


Chromosome Aberrations , Chromosomes, Human, Pair 21 , MAP Kinase Signaling System/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , ras Proteins/metabolism , Animals , Benzimidazoles/pharmacology , Cell Survival , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Mice , Mutation Rate , Sequence Analysis, DNA
10.
Am J Transplant ; 16(2): 414-25, 2016 Feb.
Article En | MEDLINE | ID: mdl-26780579

The molecular pathogenesis of posttransplant diffuse large B cell lymphoma (PT-DLBCL) is largely unknown. We have recently shown that Epstein-Barr virus-positive (EBV(+)) and -negative (EBV(-)) PT-DLBCL have distinct gene expression profiles, and the transcriptomic profile of EBV(-) PT-DLBCL is similar to that of DLBCL in immunocompetent individuals (IC-DLBCL). To validate these observations at the genomic level, we performed array-comparative genome hybridization (aCGH) analysis of 21 EBV(+) PT-DLBCL, 6 EBV(-) PT-DLBCL, and 11 control IC-DLBCL, and subsequently combined genomic and transcriptomic data. The analysis showed that EBV(+) and EBV(-) PT-DLBCL have distinct aCGH profiles and shared only one recurrent imbalance. EBV(-) PT-DLBCL, however, displayed at least 10 aberrations recurrent in IC-DLBCL, among which characteristic gain of 3/3q and 18q, and loss of 6q23/TNFAIP3 as well as 9p21/CDKN2A. The most prevalent aberration in EBV(+) PT-DLBCL was gain/amplification of 9p24.1 targeting PDCD1LG2/PDL2. Our data indicate that the FOXP1 oncogene and the tumor suppressor CDKNA2 implicated in EBV(-) DLBCL, do not play a critical role in the pathogenesis of EBV(+) PT-DLBCL. Altogether, genomic profiling of PT-/IC-DLBCL confirms that EBV(-) and EBV(+) PT-DLBCL are distinct entities, while EBV(-) PT-DLBCL has features in common with IC-DLBCL. These findings support the hypothesis that EBV(-) PT-DLBCL are de novo lymphomas in transplant recipients.


Biomarkers, Tumor/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Profiling , Genomics/methods , Lymphoma, Large B-Cell, Diffuse/genetics , Postoperative Complications/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Child , Child, Preschool , Comparative Genomic Hybridization , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Female , Follow-Up Studies , Graft Rejection , Graft Survival , Herpesvirus 4, Human , Humans , Immunoenzyme Techniques , In Situ Hybridization , Lymphoma, Large B-Cell, Diffuse/surgery , Lymphoma, Large B-Cell, Diffuse/virology , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
12.
Leukemia ; 29(5): 1092-103, 2015 May.
Article En | MEDLINE | ID: mdl-25428262

The splicing factor SF3B1 is the most commonly mutated gene in the myelodysplastic syndrome (MDS), particularly in patients with refractory anemia with ring sideroblasts (RARS). We investigated the functional effects of SF3B1 disruption in myeloid cell lines: SF3B1 knockdown resulted in growth inhibition, cell cycle arrest and impaired erythroid differentiation and deregulation of many genes and pathways, including cell cycle regulation and RNA processing. MDS is a disorder of the hematopoietic stem cell and we thus studied the transcriptome of CD34(+) cells from MDS patients with SF3B1 mutations using RNA sequencing. Genes significantly differentially expressed at the transcript and/or exon level in SF3B1 mutant compared with wild-type cases include genes that are involved in MDS pathogenesis (ASXL1 and CBL), iron homeostasis and mitochondrial metabolism (ALAS2, ABCB7 and SLC25A37) and RNA splicing/processing (PRPF8 and HNRNPD). Many genes regulated by a DNA damage-induced BRCA1-BCLAF1-SF3B1 protein complex showed differential expression/splicing in SF3B1 mutant cases. This is the first study to determine the target genes of SF3B1 mutation in MDS CD34(+) cells. Our data indicate that SF3B1 has a critical role in MDS by affecting the expression and splicing of genes involved in specific cellular processes/pathways, many of which are relevant to the known RARS pathophysiology, suggesting a causal link.


Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Stem Cells/cytology , Alternative Splicing , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Antigens, CD34/metabolism , Cell Cycle , Cell Differentiation , Cell Proliferation , Exons , Female , Gene Expression Profiling , Gene Knockdown Techniques , Heterozygote , Homeostasis , Humans , K562 Cells , Male , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , Point Mutation , RNA/genetics , RNA Splicing , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/metabolism , Sequence Analysis, RNA
14.
Leukemia ; 28(5): 1015-21, 2014 May.
Article En | MEDLINE | ID: mdl-24166298

Intrachromosomal amplification of chromosome 21 (iAMP21) defines a distinct cytogenetic subgroup of childhood B-cell precursor acute lymphoblastic leukaemia (BCP-ALL). To date, fluorescence in situ hybridisation (FISH), with probes specific for the RUNX1 gene, provides the only reliable detection method (five or more RUNX1 signals per cell). Patients with iAMP21 are older (median age 9 years) with a low white cell count. Previously, we demonstrated a high relapse risk when these patients were treated as standard risk. Recent studies have shown improved outcome on intensive therapy. In view of these treatment implications, accurate identification is essential. Here we have studied the cytogenetics and outcome of 530 iAMP21 patients that highlighted the association of specific secondary chromosomal and genetic changes with iAMP21 to assist in diagnosis, including the gain of chromosome X, loss or deletion of chromosome 7, ETV6 and RB1 deletions. These iAMP21 patients when treated as high risk showed the same improved outcome as those in trial-based studies regardless of the backbone chemotherapy regimen given. This study reinforces the importance of intensified treatment to reduce the risk of relapse in iAMP21 patients. This now well-defined patient subgroup should be recognised by World Health Organisation (WHO) as a distinct entity of BCP-ALL.


Chromosomes, Human, Pair 21 , Cytogenetic Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Treatment Outcome , Young Adult
17.
Leukemia ; 25(12): 1840-8, 2011 Dec.
Article En | MEDLINE | ID: mdl-21681191

Massively parallel pyrosequencing allows sensitive deep sequencing to detect molecular aberrations. Thus far, data are limited on the technical performance in a clinical diagnostic setting. Here, we investigated as an international consortium the robustness, precision and reproducibility of amplicon next-generation deep sequencing across 10 laboratories in eight countries. In a cohort of 18 chronic myelomonocytic leukemia patients, mutational analyses were performed on TET2, a frequently mutated gene in myeloproliferative neoplasms. Additionally, hotspot regions of CBL and KRAS were investigated. The study was executed using GS FLX sequencing instruments and the small volume 454 Life Sciences Titanium emulsion PCR setup. We report a high concordance in mutation detection across all laboratories, including a robust detection of novel variants, which were undetected by standard Sanger sequencing. The sensitivity to detect low-level variants present with as low as 1-2% frequency, compared with the 20% threshold for Sanger-based sequencing is increased. Together with the output of high-quality long reads and fast run time, we demonstrate the utility of deep sequencing in clinical applications. In conclusion, this multicenter analysis demonstrated that amplicon-based deep sequencing is technically feasible, achieves high concordance across multiple laboratories and allows a broad and in-depth molecular characterization of cancer specimens with high diagnostic sensitivity.


DNA-Binding Proteins/genetics , High-Throughput Nucleotide Sequencing , Leukemia, Myelomonocytic, Chronic/genetics , Mutation/genetics , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Aged , Aged, 80 and over , Cohort Studies , DNA Mutational Analysis , Dioxygenases , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prognosis , Proto-Oncogene Proteins p21(ras)
18.
Leukemia ; 25(1): 130-4, 2011 Jan.
Article En | MEDLINE | ID: mdl-21030981

Loss of function mutations and deletions encompassing the plant homeodomain finger 6 (PHF6) gene are present in about 20% of T-cell acute lymphoblastic leukemias (ALLs). Here, we report the identification of recurrent mutations in PHF6 in 10/353 adult acute myeloid leukemias (AMLs). Genetic lesions in PHF6 found in AMLs are frameshift and nonsense mutations distributed through the gene or point mutations involving the second plant homeodomain (PHD)-like domain of the protein. As in the case of T-ALL, where PHF6 alterations are found almost exclusively in males, mutations in PHF6 were seven times more prevalent in males than in females with AML. Overall, these results identify PHF6 as a tumor suppressor gene mutated in AML and extend the role of this X-linked tumor suppressor gene in the pathogenesis of hematologic tumors.


Carrier Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , Animals , Female , Genes, Tumor Suppressor , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/etiology , Male , Mice , Middle Aged , Myeloid Cells/metabolism , Repressor Proteins , Sex Characteristics
19.
Pediatr Blood Cancer ; 53(2): 220-2, 2009 Aug.
Article En | MEDLINE | ID: mdl-19405140

We report on an acute myeloid leukemia in a neonate whose mother was exposed to diethylstilboestrol in utero. The newborn presented with leukemia cutis, hemorrhagic skin lesions, hyperleucocytosis and disseminated intravascular coagulation. A bone marrow examination confirmed the diagnosis of acute monocytic leukemia with a t(11;19) MLL-ELL fusion transcript. Chemotherapy was initiated but the child developed a bilateral pulmonary infection that led to fatal respiratory distress. This case shows acute myeloid leukemia and the third pediatric leukemia reported after maternal diethylstilboestrol exposure.


Diethylstilbestrol/adverse effects , Estrogens, Non-Steroidal/adverse effects , Leukemia, Myeloid, Acute/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Infertility, Female/chemically induced , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Mothers , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion , Pedigree , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
...