Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Leuk Lymphoma ; 62(2): 438-445, 2021 02.
Article in English | MEDLINE | ID: mdl-33043739

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) mutations in Myeloid Neoplams (MNs) exhibit DNA hypermethylation via 2-hydroxyglutarate (2HG) over-production. Clinical impact of azacitidine (AZA) remains inconsistent in IDH1/2-mutated MNs and the potential of serum 2HG as a suitable marker of response to AZA is unknown. To address these questions, we retrospectively analyzed 93 MNs patients (78 AML, 11 MDS, 4 CMML) with IDH1/2 mutations treated with AZA. After a median of 5 cycles of AZA, overall response rate was 28% (including 15% complete remission) and median OS was 12.3 months (significantly shorter in AML compared to MDS/CMML patients). In multivariate analysis of AML patients, DNMT3A mutation was associated with shorter OS while IDH1/2 mutation subtypes had no independent impact. No difference was observed in serum 2HG levels upon AZA treatment between responding and refractory patients suggesting that serum 2HG cannot be used as a surrogate marker of AZA response.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Azacitidine/therapeutic use , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Retrospective Studies
3.
Drugs Today (Barc) ; 56(1): 21-32, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32055803

ABSTRACT

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key metabolic enzymes that convert isocitrate to alpha-ketoglutarate (alphaKG). Somatic point mutations in IDH1/2 that are found in rare distinct subsets of cancers confer a gain of function in cancer cells which results in the accumulation and secretion in vast excess of the oncometabolite D-2-hydroxyglutarate (D-2HG). Overproduction of D-2HG interferes with cellular metabolism and epigenetic regulation, contributing to oncogenesis. High levels of D-2HG inhibit alphaKG-dependent dioxygenases including histone, DNA and RNA demethylases, resulting in histone, DNA and RNA hypermethylation and cell differentiation blockade. In addition, D-2HG is a biomarker suitable for the detection of IDH1/2 mutations at diagnosis, and is also predictive of clinical response. The U.S. Food and Drug Administration (FDA) approved ivosidenib, a mutant-IDH1 enzyme inhibitor, for patients with relapsed or refractory IDH1-mutated acute myeloid leukemia (AML) in 2018, and also as front-line therapy for newly diagnosed elderly patients 75 years or older or who are ineligible to receive intensive chemotherapy in 2019. Ivosidenib represents a novel drug class for targeted therapy in AML.


Subject(s)
Glycine/analogs & derivatives , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Pyridines/therapeutic use , Adult , Aged , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic , Glycine/therapeutic use , Humans , Mutation
4.
Leuk Res ; 55: 58-64, 2017 04.
Article in English | MEDLINE | ID: mdl-28131982

ABSTRACT

BACKGROUND: Patients diagnosed with therapy-related myeloid neoplasms (TRMN) with concomitant active neoplastic disorder (CAND) are usually proposed for best supportive care (BSC). We evaluated the feasibility of using 5-azacytidine (AZA) in this setting. METHODS: All patients referred to Gustave Roussy between 2010 and 2015 for TRMN diagnosis (less than 30% blast) and eligible for AZA treatment were included. Patients with CAND proposed for BSC were also described. Patient's outcomes were analyzed based on the presence or not of a CAND. RESULTS: Fifty-two patients with TRMN were analyzed, including 19 patients with CAND (14 eligible for AZA) and 33 without CAND eligible for AZA. The 5 patients with CAND ineligible for AZA had a worst performance status (p=0.016) at diagnosis and a shorter overall survival (OS) (0.62 months). Baseline characteristics of patients eligible for AZA were similar in the 2 groups except a trend for best performance status in patients with CAND (p=0.06). Overall response rate (71.4% vs 60.3%), transfusion independence (50.0% vs 45.5%) and OS (12.7 months vs 10.8 months) were similar between patients with and without CAND respectively (p=ns). CONCLUSION: Here we report the feasibility and efficacy of AZA for selected patients with TRMN and a CAND.


Subject(s)
Azacitidine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Neoplasms, Second Primary/drug therapy , Neoplasms/complications , Adult , Aged , Aged, 80 and over , Azacitidine/administration & dosage , Female , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/mortality , Neoplasms/pathology , Neoplasms, Second Primary/complications , Neoplasms, Second Primary/mortality , Remission Induction , Retrospective Studies , Survival Rate , Treatment Outcome
5.
Med Mal Infect ; 44(4): 174-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24656841

ABSTRACT

PURPOSE: The effectiveness of posaconazole (PSZ) prophylaxis on invasive fungal infections, in patients presenting with acute myeloid leukemia (AML), seems to be correlated to its blood plasma concentration. Our goal was to identify the risk factors for underdosing. PATIENTS AND METHODS: We retrospectively reviewed the records of patients treated for AML treated with PSZ, during a 2-year period. Assays<500ng/mL were considered as under dosed. RESULTS: Fifty-nine assays (43 patients) were performed during induction (n=22) or consolidation (n=37) chemotherapy. PSZ treatment was initiated within a median of 3 days before neutropenia with a first assay performed at 8 days (3-28). The median PSZ blood plasma concentration was 375ng/mL (<200-1900). Forty-one (69%) treatment were maintained until the end of neutropenia. One patient presented with candidemia, 9 with possible invasive aspergillosis, without any significant association with underdosing. The univariate analysis showed that co-administration of proton pump inhibitors (PPIs) (P=0.01) and cause of hospitalization (induction chemotherapy vs consolidation, P=0.008) were associated with underdosing, contrary to feeding difficulties (P=0.07) and digestive disorders (P=0.5). The multivariate analysis confirmed the impact of PPI use (P=0.01) and the cause of hospitalization (P=0.003). CONCLUSION: This study highlights the major impact of PPI administration on PSZ blood plasma levels and stresses the risk of non-effective prophylaxis during induction treatment of AML.


Subject(s)
Antifungal Agents/administration & dosage , Antifungal Agents/blood , Aspergillosis/prevention & control , Drug Monitoring , Leukemia, Myeloid, Acute/blood , Triazoles/administration & dosage , Triazoles/blood , Adult , Aged , Aspergillosis/etiology , Female , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , Retrospective Studies , Young Adult
6.
Clin Microbiol Infect ; 20(7): O453-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24313354

ABSTRACT

Based on recommendations of the ECIL-4, we prospectively evaluated discontinuation of empirical antibiotic therapy in high-risk neutropenic acute myeloid leukaemia patients with fever of unknown origin. Seven patients (median neutropenia duration 30 days) were included. Four of them remained afebrile but quickly recovered from neutropenia. The other three had rapid recurrent fever. Two of these three patients had bacteraemia with susceptible strains and one of them was transferred to the ICU for septic shock. Median duration of sparing of antibiotics for the seven patients was 3 days (2-4). Because of these limited results the study was stopped.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Fever of Unknown Origin/drug therapy , Leukemia, Myeloid, Acute/complications , Neutropenia/complications , Withholding Treatment/ethics , Adult , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome
8.
Oncogene ; 32(37): 4331-42, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23085751

ABSTRACT

The term myelodysplastic syndrome (MDS) identifies a heterogeneous group of clonal disorders originating from bone marrow stem cells that often progress to acute myeloid leukemia (AML). The reference treatments for MDS include the DNA methyltransferase inhibitors azacytidine and decitabine. Recently, the epidermal growth factor receptor (EGFR) inhibitor erlotinib has been shown to exert antileukemic activity in vitro and in vivo, independent of the EGFR. Thanks to this feature, erlotinib is currently being tested as an antileukemic drug in clinical trials. Here, we report that azacytidine and erlotinib mediate synergistic antineoplastic effects in several primary or secondary (post-MDS) AML cell lines. The combination of azacytidine and erlotinib blocked cell-cycle progression and induced caspase-dependent apoptosis more consistently than either of the two agents alone. These effects were not a consequence of cellular differentiation and could be discriminated from each other, as the former depended on caspases whereas the latter did not. The synergy between azacitidine and erlotinib, which involved the proteasomal degradation of the anti-apoptotic Bcl-2 family members MCL-1 and BCL2L10 and the upregulation of their pro-apoptotic counterpart PUMA, was abolished when azacytidine was replaced by decitabine but persisted when erlotinib was substituted with gefitinib, another EGFR inhibitor. Of note, the intracellular accumulation of azacytidine was exacerbated by both erlotinib and gefitinib, pointing to a pharmacokinetic mechanism of synergy. In approximately half of the cases studied, marrow and circulating blasts from MDS and AML patients, respectively, exhibited hyperadditive cytotoxic responses to the combination of azacytidine and erlotinib. These results strongly suggest that the combination of azacytidine and erlotinib may exert clinically relevant antileukemic effects.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Leukemia, Myeloid, Acute , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Antimetabolites, Antineoplastic/toxicity , Apoptosis/drug effects , Azacitidine/toxicity , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Drug Synergism , Erlotinib Hydrochloride , Humans , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Protein Kinase Inhibitors/toxicity , Quinazolines/toxicity
10.
Cell Death Dis ; 3: e396, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23034331

ABSTRACT

The chemokine receptor CXCR4 favors the interaction of acute myeloid leukemia (AML) cells with their niche but the extent to which it participates in pathogenesis is unclear. Here, we show that CXCR4 expression at the surface of leukemic cells allowed distinguishing CXCR4 (high) from CXCR4(neg/low) AML patients. When high levels of CXCR4 are expressed at the surface of AML cells, blocking the receptor function with small molecule inhibitors could promote leukemic cell death and reduce NOD/Shi-scid/IL-2Rγ(null) (NOG) leukemia-initiating cells (LICs). Conversely, these drugs had no efficacy when AML cells do not express CXCR4 or when they do not respond to chemokine CXC motif ligand 12 (CXCL12). Functional analysis showed a greater mobilization of leukemic cells and LICs in response to drugs, suggesting that they target the interaction between leukemic cells and their supportive bone marrow microenvironment. In addition, increased apoptosis of leukemic cells in vitro and in vivo was observed. CXCR4 expression level on AML blast cells and their migratory response to CXCL12 are therefore predictive of the response to the inhibitors and could be used as biomarkers to select patients that could potentially benefit from the drugs.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Receptors, CXCR4/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Apoptosis/drug effects , Benzylamines , Chemokine CXCL12/metabolism , Child, Preschool , Cyclams , Disease Models, Animal , Female , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Middle Aged , Receptors, CXCR4/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Cell Death Dis ; 1: e104, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21368877

ABSTRACT

Experiments performed in mice revealed that anthracyclines stimulate immunogenic cell death that is characterized by the pre-apoptotic exposure of calreticulin (CRT) on the surface of dying tumor cells. Here, we determined whether CRT exposure at the cell surface (ecto-CRT) occurs in human cancer in response to anthracyclines in vivo, focusing on acute myeloid leukemia (AML), which is currently treated with a combination of aracytine and anthracyclines. Most of the patients benefit from the induction chemotherapy but relapse within 1-12 months. In this study, we investigated ecto-CRT expression on malignant blasts before and after induction chemotherapy. We observed that leukemic cells from some patients exhibited ecto-CRT regardless of chemotherapy and that this parameter was not modulated by in vivo chemotherapy. Ecto-CRT correlated with the presence of phosphorylated eIF2α within the blasts, in line with the possibility that CRT exposure results from an endoplasmic reticulum stress response. Importantly, high levels of ecto-CRT on malignant myeloblasts positively correlated with the ability of autologous T cells to secrete interferon-γ on stimulation with blast-derived dendritic cell. We conclude that the presence of ecto-CRT on leukemia cells facilitates cellular anticancer immune responses in AML patients.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Calreticulin/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Calreticulin/drug effects , Cohort Studies , Dendritic Cells/physiology , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Immunity, Cellular/drug effects , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Phagocytosis/drug effects , Phosphorylation , Survival Analysis , T-Lymphocytes/immunology
14.
Oncogene ; 28(22): 2205-18, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19398952

ABSTRACT

The molecular mechanisms responsible for the evolution from the preleukemic entities of low-risk myelodysplastic syndrome (MDS) to the less favorable forms of high-risk MDS, as well as those enabling transformation to acute myeloid leukemia (AML), are still incompletely understood. Abundant evidence from solid tumors demonstrates that preneoplastic lesions activate signaling pathways of a DNA damage response (DDR), which functions as an 'anticancer barrier' hindering tumorigenesis. Testing the hypothesis that subgroups of MDS and AML differ with respect to DDR, we first assessed markers of DDR (phosphorylation of ATM, Chk-1, Chk-2 and H2AX) in cell lines representing different entities of MDS (P39, MOLM-13) and AML (MV4-11, KG-1) before and after gamma-irradiation. Although gamma-irradiation induced apoptosis and G(2)/M arrest and a concomitant increase in the phosphorylation of ATM, Chk-1 and H2AX in MDS-derived cell lines, this radiation response was attenuated in the AML-derived cell lines. It is noteworthy that KG-1, but not P39 cells exhibit signs of an endogenous activation of the DDR. Similarly, we found that the frequency of P-ATM(+) cells detectable in bone marrow (BM) biopsies increased in samples from patients with AML as compared with high-risk MDS samples and significantly correlated with the percentage of BM blasts. In contrast, the frequency of gamma-H2AX(+) cells was heterogeneous in all subgroups of AML and MDS. Whereas intermediate-1 MDS samples contained as little P-Chk-1 and P-Chk-2 as healthy controls, staining for both checkpoint kinases increased in intermediate-2 and high-risk MDS, yet declined to near-to-background levels in AML samples. Thus the activation of Chk-1 and Chk-2 behaves in accord with the paradigm established for solid tumors, whereas ATM is activated during and beyond transformation. In conclusion, we demonstrate the heterogeneity of the DDR response in MDS and AML and provide evidence for its selective suppression in AML because of the uncoupling between activated ATM and inactive checkpoint kinases.


Subject(s)
DNA Damage , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Ataxia Telangiectasia Mutated Proteins , Biopsy , Bone Marrow/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 2 , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , Granulocyte Precursor Cells/radiation effects , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Proteins/metabolism
15.
Oncogene ; 28(8): 1099-109, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19079347

ABSTRACT

The anti-apoptotic transcription factor nuclear factor-kappaB (NF-kappaB) is constitutively activated in CD34(+) myeloblasts from high-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) patients. Inhibition of NF-kappaB by suppressing the canonical NF-kappaB activation pathway, for instance by knockdown of the three subunits of the inhibitor of NF-kappaB (I kappaB) kinase (IKK) complex (IKK1, IKK2 and NEMO) triggers apoptosis in such cells. Here, we show that an MDS/AML model cell line exhibits a constitutive interaction, within the nucleus, of activated, S1981-phosphorylated ataxia telangiectasia mutated (ATM) with NEMO. Inhibition of ATM with two distinct pharmacological inhibitors suppressed the activating autophosphorylation of ATM, blocked the interaction of ATM and NEMO, delocalized NEMO as well as another putative NF-kappaB activator, PIDD, from the nucleus, abolished the activating phosphorylation of the catalytic proteins of the IKK complex (IKK1/2 on serines 176/180), enhanced the expression of I kappaB alpha and caused the relocalization of NF-kappaB from the nucleus to the cytoplasm, followed by apoptosis. Knockdown of ATM with small-interfering RNAs had a similar effect that could not be enhanced by knockdown of NEMO, PIDD and the p65 NF-kappaB subunit, suggesting that an ATM inhibition/depletion truly induced apoptosis through inhibition of the NF-kappaB system. Pharmacological inhibition of ATM also induced the nucleocytoplasmic relocalization of p65 in malignant myeloblasts purified from patients with high-risk MDS or AML, correlating with the induction of apoptosis. Altogether, these results support the contention that constitutively active ATM accounts for the activation of NF-kappaB in high-risk MDS and AML.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Active Transport, Cell Nucleus , Adult , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins , Bone Marrow Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Damage , Death Domain Receptor Signaling Adaptor Proteins , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Leukemia, Myeloid, Acute/pathology , Middle Aged , Myelodysplastic Syndromes/pathology , NF-kappa B/genetics , Phosphorylation , Protein Transport , Risk Factors , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Cells, Cultured
16.
Leuk Res ; 32(7): 1049-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18191202

ABSTRACT

Anemia in MDS with 5q deletion was generally considered, until the advent of lenalidomide, unresponsive to available treatments. We analyzed erythroid response to erythropoetin (EPO) or darbepoetin (DAR) and thalidomide in MDS with 5q deletion treated by French centers (GFM) and in whom karyotype was successfully performed. Of 345 patients treated with EPO or DAR+/-G-CSF, 48 had 5q deletion. The response rate was 46% (31% major, 15% minor) according to International Working Group (IWG) 2000 criteria versus 64% in patients without 5q deletion (p=0.03). According to IWG 2006 criteria, the response rate in patients with 5q deletion was 39% versus 52% in patients without 5q deletion (p=0.10). Mean duration of response was 14 months versus 25 months (IWG 2000) and 13 months versus 27 months (IWG 2006) in 5q deletion and non-5q deletion patients (p=0.019 and 0.003, respectively). Of 120 MDS treated with thalidomide, all of whom had successful cytogenetic analysis, 37% of the 24 patients with 5q deletion responded (IWG 2000 criteria, 20% major, 17% minor) with a mean duration of 9.5 months, versus 32% (18% major, 14% minor) in MDS without 5q deletion and a mean response duration of 9 months (p=NS). Our results confirm that response rates to EPO or DAR and thalidomide are clearly inferior to those obtained with lenalidomide.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 5 , Erythropoietin/therapeutic use , Myelodysplastic Syndromes/drug therapy , Thalidomide/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/genetics
17.
Leukemia ; 21(1): 66-71, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17051246

ABSTRACT

Pivotal phase II studies in acute myeloblastic leukemia (AML) patients in first relapse have used gemtuzumab ozogamicin (GO) (Mylotarg) at a dose of 9 mg/m(2) on days 1 and 14. These studies showed a 26% response rate (13% complete remission (CR) and 13% CRp (complete remission with incomplete platelet recovery)) but with high degree of hematological and liver toxicities. Based on in vitro studies showing a re-expression of CD33 antigenic sites on the cell surface of blasts cells after exposure to GO, we hypothesized that fractionated doses of GO may be efficient and better tolerated. Fifty-seven patients with AML in first relapse received GO at a dose of 3 mg/m(2) on days 1, 4 and 7 for one course. Fifteen patients (26%) achieved CR and four (7%) CRp. Remission rate correlated strongly with P-glycoprotein and MRP1 activities. The median relapse-free survival was 11 months, similar for CR or CRp patients. Median duration of neutropenia < 500/microl and thrombocytopenia < 50,000/microl were, respectively, 23 and 21 days. No grade 3 or 4 liver toxicity was observed. No veno-occlusive disease occurred after GO or after hematopoietic stem cell transplantation given after GO in seven patients. Mylotarg administered in fractionated doses demonstrated an excellent efficacy/safety profile.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/blood , Adult , Aged , Aged, 80 and over , Aminoglycosides/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Antineoplastic Agents/adverse effects , Disease-Free Survival , Drug Administration Schedule , Gemtuzumab , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Middle Aged , Multidrug Resistance-Associated Proteins/blood , Recurrence , Remission Induction , Sialic Acid Binding Ig-like Lectin 3
18.
Leukemia ; 20(12): 2155-61, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17039234

ABSTRACT

Adult patients with acute lymphoblastic leukemia (ALL) and t(1;19)/E2A-PBX1 or t(4;11)/MLL-AF4 have a poor outcome. We have evaluated the impact of an intensified post-remission therapy using a high-dose chemotherapy course followed by allogeneic or autologous SCT on the outcome of 58 patients with t(1;19)/E2A-PBX1 (E2A group, n=24) or t(4;11)/MLL-AF4 (MLL group, n=34) treated in the LALA-94 multicenter prospective study. Patients in the MLL group had higher WBC counts and more frequent DIC. CR rates achieved by MLL and E2A groups were similar to other B-cell ALL (87, 82 and 86% respectively). While in CR, patients with a donor were assigned to alloSCT (n=22), the remaining patients with were randomized between autoSCT (n=15) or chemotherapy (n=8). Five-year overall survival was 31 and 45% for E2A and MLL groups, respectively. In both groups, DFS was higher in the alloSCT arm as compared to autoSCT and chemotherapy arms. The results of this study show that chemotherapy intensification did not overcome the poor prognosis of adults with t(1;19)/E2A-PBX1. Allogeneic SCT should thus be offered in first CR to patients with t(1;19)/E2A-PBX1 or t(4;11)/MLL-AF4. New therapeutic approaches are needed for patients without donor.


Subject(s)
Burkitt Lymphoma/genetics , Burkitt Lymphoma/therapy , Hematopoietic Stem Cell Transplantation , Translocation, Genetic , Adolescent , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 4/genetics , DNA-Binding Proteins/genetics , Female , Histone-Lysine N-Methyltransferase , Humans , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , Prospective Studies , Proto-Oncogene Proteins/genetics , Transcriptional Elongation Factors , Transplantation, Homologous
20.
Leukemia ; 20(6): 965-70, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16598313

ABSTRACT

In core binding factors (CBF) acute myeloid leukemia (AML), the disruption of CBFalpha/beta genes impairs normal hematopoietic differentiation and is supposed to cooperate with additional mutations promoting proliferation. The incidence and the prognosis of receptor tyrosine kinase (RTK) c-Kit and FLT3 mutations and Ras mutations were evaluated in 103 pediatric and adult patients with CBF-AML. c-Kit mutations were present in 17% patients. c-Kit exon 8 mutations were more frequent in inv(16) than in t(8;21) subset (20 versus 6%). Only one patient had FLT3-ITD but FLT3-D835 was as frequent as reported in AML population (7%). Ras mutations were significantly more frequent in inv(16) than in t(8;21) subset (36 versus 8%, P=0.001). RTK mutations were associated with a higher white blood cell count (WBC) (36 versus 21 G/L, P=0.05). FLT3 mutations were significantly associated with a shorter EFS and survival (P<0.0001 and P=0.0002) owing to an excess of early events. c-Kit mutations were associated with a shorter EFS and RFS (P=0.002 and P=0.003) in t(8;21) but not inv(16) patients. As previously observed, Ras mutations did not affect prognosis. Screening for RTK mutations may help to identify patients with a more adverse outcome and thus susceptible to benefit from intensified protocols or RTK inhibitors.


Subject(s)
Core Binding Factors/genetics , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins p21(ras)/genetics , fms-Like Tyrosine Kinase 3/genetics , Acute Disease , Adolescent , Adult , Aged , Child , Child, Preschool , Exons , Female , Humans , Infant , Leukemia, Myeloid/diagnosis , Male , Middle Aged , Mutation , Prognosis , Retrospective Studies , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...