Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
Blood ; 138(10): 898-911, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34019641

ABSTRACT

Germline pathogenic TERT variants are associated with short telomeres and an increased risk of developing myelodysplastic syndrome (MDS) among patients with a telomere biology disorder. We identified TERT rare variants in 41 of 1514 MDS patients (2.7%) without a clinical diagnosis of a telomere biology disorder who underwent allogeneic transplantation. Patients with a TERT rare variant had shorter telomere length (P < .001) and younger age at MDS diagnosis (52 vs 59 years, P = .03) than patients without a TERT rare variant. In multivariable models, TERT rare variants were associated with inferior overall survival (P = .034) driven by an increased incidence of nonrelapse mortality (NRM; P = .015). Death from a noninfectious pulmonary cause was more frequent among patients with a TERT rare variant. Most variants were missense substitutions and classified as variants of unknown significance. Therefore, we cloned all rare missense variants and quantified their impact on telomere elongation in a cell-based assay. We found that 90% of TERT rare variants had severe or intermediate impairment in their capacity to elongate telomeres. Using a homology model of human TERT bound to the shelterin protein TPP1, we inferred that TERT rare variants disrupt domain-specific functions, including catalysis, protein-RNA interactions, and recruitment to telomeres. Our results indicate that the contribution of TERT rare variants to MDS pathogenesis and NRM risk is underrecognized. Routine screening for TERT rare variants in MDS patients regardless of age or clinical suspicion may identify clinically inapparent telomere biology disorders and improve transplant outcomes through risk-adapted approaches.


Subject(s)
Genetic Variation , Myelodysplastic Syndromes , Telomerase/genetics , Adult , Disease-Free Survival , Female , Humans , Male , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Survival Rate
2.
Br J Haematol ; 192(6): 1006-1010, 2021 03.
Article in English | MEDLINE | ID: mdl-32307695

ABSTRACT

We screened 47 subjects with DDX41 variants among 1529 subjects with myeloid neoplasms. The most common germline variants included Splice c.935 + 4A>T, p.T360Ifs*33, p.V152G, p.S217Ifs*4, p.R311* and p.R369*. Except for the p.R369*, no other variants have been previously reported. Clinical covariates of subjects with simple DDX41 somatic variants and germline/somatic biallelic variants are similar. The two-year overall survival (OS) of subjects with DDX41 variants was 85%. Overall response rate to demethylation therapy in subjects with DDX41 variants was 69%. The response did not correlate with the presence of a germline variant.


Subject(s)
DEAD-box RNA Helicases/genetics , Hematologic Neoplasms , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Neoplasm Proteins/genetics , Adult , Aged , DEAD-box RNA Helicases/metabolism , Disease-Free Survival , Female , Hematologic Neoplasms/enzymology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Neoplasm Proteins/metabolism , Survival Rate
3.
Ann Hematol ; 100(5): 1149-1158, 2021 May.
Article in English | MEDLINE | ID: mdl-33569703

ABSTRACT

Suspicion of myelodysplastic syndromes (MDS) is the most common reason for bone marrow aspirate in elderly patients. This study aimed to prospectively validate the accuracy for flow cytometric analysis of peripheral blood neutrophil myeloperoxidase expression in ruling out MDS. We enrolled 62 consecutive patients who were referred for suspected MDS, based on medical history and peripheral blood cytopenia. The accuracy of intra-individual robust coefficient of variation (RCV) for peripheral blood neutrophil myeloperoxidase expression was assessed with a prespecified 30% threshold. Cytomorphological evaluation of bone marrow aspirate performed by experienced hematopathologists confirmed MDS in 23 patients (prevalence, 37%), unconfirmed MDS in 32 patients (52%, including 3 patients with idiopathic cytopenia of undetermined significance (ICUS)), and was uninterpretable in 7 patients (11%). The median intra-individual RCV values for neutrophil myeloperoxidase expression in peripheral blood were 37.4% (range, 30.7-54.1), 29.2% (range, 28.1-32.1), and 29.1% (range, 24.7-37.8) for patients with confirmed suspicion of MDS, ICUS, and unconfirmed suspicion of MDS, respectively (P<0.001). The area under the ROC curve was 0.92 (95% confidence interval, 0.86-0.99). An intra-individual RCV value lower than 30% ruled out MDS for 35% (i.e., 19/55) patients referred for suspected disease, with 100% sensitivity (95% CI, 85-100%) and 100% negative predictive value (95% CI, 82-100%) estimates. This study shows that flow cytometric analysis of peripheral blood neutrophil myeloperoxidase expression might obviate the need for bone marrow aspirate for 35% of patients with suspected MDS. Trial registration: ClinicalTrials.gov identifier: NCT03363399 (first posted on December 6, 2017).


Subject(s)
Myelodysplastic Syndromes/diagnosis , Neutrophils/enzymology , Peroxidase/analysis , Aged , Aged, 80 and over , Bone Marrow/pathology , Female , Flow Cytometry/methods , Humans , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/enzymology , Prospective Studies
4.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344823

ABSTRACT

Iron overload is related to leukemia transformation in myelodysplastic syndrome (MDS) patients. Siderophores help to transport iron. Type 2-hydroxybutyrate dehydrogenase (BDH2) is a rate-limiting factor in the biogenesis of siderophores. Using qRT-PCR, we analyze BDH2mRNA expression in the bone marrow (BM) of 187 MDS patients, 119 de novo acute myeloid leukemia (AML) patients, and 43 lymphoma patients with normal BM. Elevated BDH2mRNA expression in BM is observed in MDS patients (n = 187 vs. 43, normal BM; P = 0.009), and this is related to ferritin levels. Patients with higher BDH2 expression show a greater risk of leukemia progression (15.25% vs. 3.77%, lower expression; P = 0.017) and shorter leukemia-free-survival (medium LFS, 9 years vs. 7 years; P = 0.024), as do patients with a ferritin level ≥350 ng/mL. Additionally, we investigate the mechanisms related to the prognostic ability of BDH2 by using BDH2-KD THP1. The cell cycle analysis, surface markers, and special stain studies indicate that BDH2-KD induces differentiation and decreases the growth rate of THP1 cells, which is associated with the retardation of the cell cycle. Moreover, many genes, including genes related to mitochondrial catabolism, oncogenes, tumor suppressor genes, and genes related to cell differentiation and proliferation influence BDH2-KD THP1 cells. Herein, we demonstrate that BDH2 is involved in cell cycle arrest and the inhibition of differentiation in malignant cells. Furthermore, the high BDH2 expression in MDS patients could be suggestive of a poor prognostic factor. This study provides a foundation for further research on the roles of BDH2 and iron metabolism in the pathogenesis of MDS.


Subject(s)
Bone Marrow/pathology , Gene Expression Regulation/genetics , Hydroxybutyrate Dehydrogenase/physiology , Leukemia, Myeloid, Acute/enzymology , Myelodysplastic Syndromes/enzymology , Preleukemia/enzymology , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Bone Marrow/metabolism , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Female , Ferritins/blood , Gene Expression Regulation, Leukemic , Humans , Hydroxybutyrate Dehydrogenase/biosynthesis , Hydroxybutyrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lipocalin-2/biosynthesis , Lipocalin-2/genetics , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Preleukemia/genetics , Preleukemia/pathology , Prognosis , Progression-Free Survival , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/genetics , THP-1 Cells , Young Adult
5.
Haematologica ; 104(12): 2382-2390, 2019 12.
Article in English | MEDLINE | ID: mdl-31004030

ABSTRACT

Suspicion of myelodysplastic syndromes (MDS) is one of the commonest reasons for bone marrow aspirate in elderly patients presenting with persistent peripheral blood (PB) cytopenia of unclear etiology. A PB assay that accurately rules out MDS would have major benefits. The diagnostic accuracy of the intra-individual robust coefficient of variation (RCV) for neutrophil myeloperoxidase (MPO) expression measured by flow cytometric analysis in PB was evaluated in a retrospective derivation study (44 MDS cases and 44 controls) and a prospective validation study (68 consecutive patients with suspected MDS). Compared with controls, MDS cases had higher median RCV values for neutrophil MPO expression (40.2% vs 30.9%; P<0.001). The area under the receiver operating characteristic curve estimates were 0.94 [95% confidence interval (CI): 0.86-0.97] and 0.87 (95%CI: 0.76-0.94) in the derivation and validation studies, respectively. A RCV lower than 30% ruled out MDS with 100% sensitivity (95%CI: 78-100%) and 100% negative predictive value (95%CI: 83-100%) in the prospective validation study. Neutrophil MPO expression measured by flow cytometric analysis in PB might obviate the need for invasive bone marrow aspirate and biopsy for up to 29% of patients with suspected MDS.


Subject(s)
Biomarkers, Tumor/analysis , Flow Cytometry/methods , Leukemia, Myelomonocytic, Chronic/diagnosis , Myelodysplastic Syndromes/diagnosis , Neutrophils/enzymology , Peroxidase/metabolism , Aged , Case-Control Studies , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Leukemia, Myelomonocytic, Chronic/enzymology , Male , Myelodysplastic Syndromes/enzymology , Prognosis , Prospective Studies , ROC Curve , Retrospective Studies
6.
FASEB J ; 32(2): 681-692, 2018 02.
Article in English | MEDLINE | ID: mdl-28970249

ABSTRACT

PI-PLCß1 is involved in cell proliferation, differentiation, and myelodysplastic syndrome (MDS) pathogenesis. Moreover, the increased activity of PI-PLCß1 reduces the expression of PKC-α, which, in turn, delays the cell proliferation and is linked to erythropoiesis. Lenalidomide is currently used in low-risk patients with MDS and del(5q), where it can suppress the del(5q) clone and restore normal erythropoiesis. In this study, we analyzed the effect of lenalidomide on 16 patients with low-risk del(5q) MDS, as well as del(5q) and non-del(5q) hematopoietic cell lines, mainly focusing on erythropoiesis, cell cycle, and PI-PLCß1/PKC-α signaling. Overall, 11 patients were evaluated clinically, and 10 (90%) had favorable responses; the remaining case had a stable disease. At a molecular level, both responder patients and del(5q) cells showed a specific induction of erythropoiesis, with a reduced γ/ß-globin ratio, an increase in glycophorin A, and a nuclear translocation of PKC-α. Moreover, lenalidomide could induce a selective G0/G1 arrest of the cell cycle in del(5q) cells, slowing down the rate proliferation in those cells. Altogether, our results could not only better explain the role of PI-PLCß1/PKC-α signaling in erythropoiesis but also lead to a better comprehension of the lenalidomide effect on del(5q) MDS and pave the way to innovative, targeted therapies.-Poli, A., Ratti, S., Finelli, C., Mongiorgi, S., Clissa, C., Lonetti, A., Cappellini, A., Catozzi, A., Barraco, M., Suh, P.-G., Manzoli, L., McCubrey, J. A., Cocco, L., Follo, M. Y. Nuclear translocation of PKC-α is associated with cell cycle arrest and erythroid differentiation in myelodysplastic syndromes (MDSs).


Subject(s)
Cell Differentiation , Cell Nucleus/enzymology , Erythroid Cells/enzymology , Erythropoiesis , G1 Phase Cell Cycle Checkpoints , Myelodysplastic Syndromes/enzymology , Protein Kinase C-alpha/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Aged , Aged, 80 and over , Cell Line , Cell Nucleus/genetics , Cell Nucleus/pathology , Erythroid Cells/pathology , Female , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Protein Kinase C-alpha/genetics , Resting Phase, Cell Cycle
7.
Br J Haematol ; 178(2): 240-249, 2017 07.
Article in English | MEDLINE | ID: mdl-28486748

ABSTRACT

Telomere dysfunction is implicated in the generation of large-scale genomic rearrangements that drive progression to malignancy. In this study we used high-resolution single telomere length analysis (STELA) to examine the potential role of telomere dysfunction in 80 myelodysplastic syndrome (MDS) and 95 de novo acute myeloid leukaemia (AML) patients. Despite the MDS cohort being older, they had significantly longer telomeres than the AML cohort (P < 0·0001) where telomere length was also significantly shorter in younger AML patients (age <60 years) (P = 0·02) and in FLT3 internal tandem duplication-mutated AML patients (P = 0·03). Using a previously determined telomere length threshold for telomere dysfunction (3·81 kb) did not provide prognostic resolution in AML [Hazard ratio (HR) = 0·68, P = 0·2]. In contrast, the same length threshold was highly prognostic for overall survival in the MDS cohort (HR = 5·0, P < 0·0001). Furthermore, this telomere length threshold was an independent parameter in multivariate analysis when adjusted for age, gender, cytogenetic risk group, number of cytopenias and International Prognostic Scoring System (IPSS) score (HR = 2·27, P < 0·0001). Therefore, telomere length should be assessed in a larger prospective study to confirm its prognostic role in MDS with a view to integrating this variable into a revised IPSS.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/pathology , Telomere/pathology , Adult , Aged , Biomarkers, Tumor/metabolism , Disease-Free Survival , Female , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/mortality , Prognosis , Prospective Studies , Risk Factors , Telomerase/metabolism , Young Adult
8.
J Cell Physiol ; 231(8): 1645-55, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26626942

ABSTRACT

Phosphatidylinositol (PI) metabolism represents the core of a network of signaling pathways which modulate many cellular functions including cell proliferation, cell differentiation, apoptosis, and membrane trafficking. An array of kinases, phosphatases, and lipases acts on PI creating an important number of second messengers involved in different cellular processes. Although, commonly, PI signaling was described to take place at the plasma membrane, many evidences indicated the existence of a PI cycle residing in the nuclear compartment of eukaryotic cells. The discovery of this mechanism shed new light on many nuclear functions, such as gene transcription, DNA modifications, and RNA expression. As these two PI cycles take place independently of one another, understanding how nuclear lipid signaling functions and modulates nuclear output is fundamental in the study of many cellular processes. J. Cell. Physiol. 231: 1645-1655, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cell Nucleus/enzymology , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Second Messenger Systems , Type C Phospholipases/metabolism , Animals , Cell Cycle Checkpoints , Cell Differentiation , Cell Nucleus/pathology , Cell Proliferation , Humans , Hydrolysis , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology , Neoplasms/enzymology , Neoplasms/pathology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Transcription, Genetic
9.
Cancer ; 122(12): 1871-9, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26990290

ABSTRACT

BACKGROUND: RAS/RAF/mitogen-activated protein kinase activation is common in myeloid malignancies. Trametinib, a mitogen-activated protein kinase kinase 1 (MEK1)/MEK2 inhibitor with activity against multiple myeloid cell lines at low nanomolar concentrations, was evaluated for safety and clinical activity in patients with relapsed/refractory leukemias. METHODS: This phase 1/2 study accrued patients with any relapsed/refractory leukemia in phase 1. In phase 2, this study accrued patients with relapsed/refractory acute myeloid leukemia (AML) or high-risk myelodysplastic syndromes (MDS) with NRAS or KRAS mutations (cohort 1); patients with AML, MDS, or chronic myelomonocytic leukemia (CMML) with a RAS wild-type mutation or an unknown mutation status (cohort 2); and patients with CMML with an NRAS or KRAS mutation (cohorts 3). RESULTS: The most commonly reported treatment-related adverse events were diarrhea, rash, nausea, and increased alanine aminotransferase levels. The phase 2 recommended dose for Trametinib was 2 mg orally daily. The overall response rates were 20%, 3%, and 27% for cohorts 1, 2, and 3, respectively, and this indicated preferential activity among RAS-mutated myeloid malignancies. Repeated cycles of trametinib were well tolerated with manageable or reversible toxicities; these results were similar to those of other trametinib studies. CONCLUSIONS: The selective, single-agent activity of trametinib against RAS-mutated myeloid malignancies validates its therapeutic potential. Combination strategies based on a better understanding of the hierarchical role of mutations and signaling in myeloid malignancies are likely to improve the response rate and duration. Cancer 2016;122:1871-9. © 2016 American Cancer Society.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia/drug therapy , MAP Kinase Signaling System/drug effects , Myelodysplastic Syndromes/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Humans , Kaplan-Meier Estimate , Leukemia/blood , Leukemia/enzymology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Middle Aged , Mutation , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/adverse effects , Pyridones/blood , Pyrimidinones/adverse effects , Pyrimidinones/blood , Recurrence , Young Adult
10.
Ann Hematol ; 95(10): 1571-82, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26983918

ABSTRACT

DNA methylation is the most common epigenetic modification in the mammalian genome. DNA methylation is governed by the DNA methyltransferases mainly DNMT1, DNMT3A, and DNMT3B. DNMT1 methylates hemimethylated DNA ensuring accurate DNA methylation maintenance. DNMT1 is involved in the proper differentiation of hematopoietic stem cells (HSCs) through the interaction with effector molecules. DNMT1 is deregulated in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) as early as the leukemic stem cell stage. Through the interaction with fundamental transcription factors, non-coding RNAs, fusion oncogenes and by modulating core members of signaling pathways, it can affect leukemic cells biology. DNMT1 action might be also catalytic-independent highlighting a methylation-independent mode of action. In this review, we have gathered some current facts of DNMT1 role in AML and MDS and we also propose some perspectives for future studies.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Gene Expression Regulation, Leukemic/genetics , Leukemia, Myeloid, Acute/enzymology , Myelodysplastic Syndromes/enzymology , Neoplasm Proteins/physiology , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methyltransferase 3A , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Transcription Factors/physiology , Transcription, Genetic , DNA Methyltransferase 3B
11.
Vopr Onkol ; 62(1): 101-3, 2016.
Article in Russian | MEDLINE | ID: mdl-30444586

ABSTRACT

There was studied the level of matrix metalloproteinase-9 (MMP-9) in plasma of bone marrow aspirates in 87 patients: 39 with acute myeloid leukemia (AML) and 48 myelodysplastic syndrome (MDS). It has been found out an association of the level of MMP-9 in plasma of bone marrow aspirates in patients with AML and MDS with a volume of leukemic mass.


Subject(s)
Bone Marrow/enzymology , Leukemia, Myeloid, Acute/enzymology , Matrix Metalloproteinase 9/metabolism , Myelodysplastic Syndromes/enzymology , Neoplasm Proteins/metabolism , Adolescent , Adult , Aged , Biopsy, Needle , Bone Marrow/pathology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/pathology
12.
Hematol Oncol ; 33(2): 57-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24777753

ABSTRACT

Rigosertib (ON 01910.Na) is an inhibitor of the phosphoinositide 3-kinase and polo-like kinase pathways that induces mitotic arrest and apoptosis in neoplastic cells, while sparing normal cells. Our purpose is to summarize the clinical activity and safety of intravenous (IV) rigosertib delivered by an external ambulatory infusion pump in patients with refractory anemia with excess blasts-1, -2, or, -t myelodysplastic syndromes (MDS) following prior treatment with DNA methyltransferase (DNMT) inhibitors. A total of 39 patients with MDS who fulfilled these criteria were enrolled in four phase 1-2 clinical trials of IV rigosertib. Thirty five (88%) had higher risk disease according to the Revised International Prognostic Scoring System. Median overall survival for this group of 39 patients was 35 weeks. Of 30 evaluable patients with follow-up bone marrow biopsies, 12 (40%) achieved complete (n = 5) or partial (n = 7) bone marrow blast responses. In addition, 15 patients achieved stabilization of bone marrow blasts. One patient with a complete bone marrow response also achieved a complete cytogenetic response. A second patient with stable bone marrow blasts achieved a partial cytogenetic response. Two of the responding patients and three patients with stable disease had hematological improvements. Rigosertib-induced bone marrow blast decreases and stability appeared to be predictive of prolonged survival. IV rigosertib had a favorable safety profile without significant myelosuppression. Most common drug-related toxicities included fatigue, diarrhea, nausea, dysuria, and hematuria. In summary, IV rigosertib is well tolerated and has clinical activity in patients with higher risk MDS following DNMT inhibitor treatment. A multinational pivotal phase 3 randomized clinical trial of rigosertib versus best supportive care for patients with MDS with excess blasts following prior treatment with DNMT inhibitors (ONTIME: ON 01910.Na Trial In Myelodysplastic SyndromE) has recently completed enrollment.


Subject(s)
Clinical Trials, Phase I as Topic/statistics & numerical data , Clinical Trials, Phase II as Topic/statistics & numerical data , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Glycine/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Sulfones/therapeutic use , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/drug therapy , Anemia, Refractory, with Excess of Blasts/enzymology , Anemia, Refractory, with Excess of Blasts/pathology , Bone Marrow/pathology , Cell Cycle Proteins/antagonists & inhibitors , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/pharmacology , Female , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacology , Glycine/therapeutic use , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Risk , Signal Transduction/drug effects , Sulfones/administration & dosage , Sulfones/adverse effects , Sulfones/pharmacology , Polo-Like Kinase 1
14.
Am J Hematol ; 90(4): 276-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25530214

ABSTRACT

We investigated the combination of midostaurin and azacitidine (AZA) in patients with acute myeloid leukemia (AML) and high risk myelodysplastic syndrome (MDS). Patients received AZA 75 mg m(-2) on days 1-7 and midostaurin 25 mg bid (in cohort 1 of phase I) or 50 mg bid (in cohort 2 of Phase I and in Phase II) orally on day 8-21 during the first cycle and continuously thereafter. Fourteen patients were enrolled in the phase I and 40 in the phase II. Overall response rate was 26%. The median remission duration (RD) was 20 weeks and was significantly longer in patients with FLT3 mutations not previously exposed to other FLT3 inhibitors (P = 0.05) and in patients not previously transplanted (P = 0.01). Thirty-two (59%) patients have died, all of complications related to disease progression. G3-4 nonhematological toxicity was reported in 38 (70%) patients, most frequently infections (56%), ejection fraction reduction (11%), and diarrhea or nausea/vomiting (9% each). The combination of midostaurin and AZA is an effective and safe regimen in patients with AML and high-risk MDS. Patients with FLT3 mutations but not previously exposed to other FLT3 inhibitors and patients not previously transplanted derived the greatest benefit. Further studies with this combination are warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/therapeutic use , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/mortality , Staurosporine/administration & dosage , Staurosporine/adverse effects , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , Young Adult , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
17.
Br J Haematol ; 167(5): 664-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25160658

ABSTRACT

Resistance to temozolomide is largely mediated by the DNA repair enzyme O(6) -methylguanine DNA methyltransferase (MGMT). We conducted a prospective multicentre study of patients with previously untreated acute myeloid leukaemia (AML) or high-risk myelodysplastic syndrome (MDS) who were not candidates for intensive therapy. Patient selection was based on MGMT expression by Western blot. Patients with MGMT:ACTB (ß-actin) ratio <0·2 were eligible to receive temozolomide 200 mg/m(2) /d ×7 d. Patients achieving a complete response (CR) could receive up to 12 monthly cycles of temozolomide ×5/28 d. Of 166 patients screened, 81 (49%) demonstrated low MGMT expression; 45 of these were treated with temozolomide. The overall response rate was 53%; 36% achieved complete clearance of blasts, with 27% achieving a CR/CR with incomplete platelet recovery (CRp). Factors associated with a trend toward a higher response rate included MDS, methylated MGMT promoter and standard cytogenetic risk group. Induction and post-remission cycles were well-tolerated and most patients were treated on an outpatient basis. Patient who achieved CR/CRp had a superior overall survival compared to partial or non-responders. In conclusion, targeted therapy based on pre-selection for low MGMT expression was associated with a higher response rate to temozolomide compared to previous reports of unselected patients.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , DNA Modification Methylases/biosynthesis , DNA Repair Enzymes/biosynthesis , Dacarbazine/analogs & derivatives , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Tumor Suppressor Proteins/biosynthesis , Aged , Aged, 80 and over , Animals , Dacarbazine/administration & dosage , Disease-Free Survival , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/mortality , Prospective Studies , Risk Factors , Survival Rate , Temozolomide
19.
Br J Haematol ; 162(4): 517-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23789936

ABSTRACT

The multi-kinase inhibitor rigosertib (ON 01910.Na) induces mitotic arrest and apoptosis in myeloblasts, while sparing normal cells. The purpose of this study was to determine the pharmacokinetic profile, maximum-tolerated dose (MTD), safety, and clinical activity of an oral formulation of rigosertib in patients with myelodysplastic syndromes (MDS). For pharmacokinetic studies, patients received rigosertib in single escalating weekly doses. To determine the MTD, patient cohorts received escalating doses of rigosertib twice daily for 14 d of a 21-d cycle. Overall, 37 patients were treated. Rigosertib exposure increased with escalating oral doses. Mean absolute oral bioavailability ranged from 13·9% (fed) to 34·8% (fasting) in 12 patients treated at the 560 mg b.i.d. dose level. Dose-limiting toxicity (grade 3 dysuria and shortness of breath) occurred at the 700 mg b.i.d. dose. Five patients experienced grade 3 non-haematological toxicity, including symptoms of urothelial inflammation, hypotension and syncope, fatigue and abdominal pain. Encouraging signs of clinical activity included two bone marrow complete remissions in refractory anaemia with excess blasts type 1 patients previously treated with azacitidine. In addition, four patients each achieved transfusion independence and haematological improvements. In conclusion, oral rigosertib is bioavailable and well tolerated, and has clinical activity in patients with MDS.


Subject(s)
Glycine/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Protein Kinase Inhibitors/therapeutic use , Sulfones/therapeutic use , Administration, Oral , Aged , Aged, 80 and over , Biological Availability , Capsules , Disease Progression , Dose-Response Relationship, Drug , Dyspnea/chemically induced , Female , Food-Drug Interactions , Gastrointestinal Diseases/chemically induced , Glycine/administration & dosage , Glycine/adverse effects , Glycine/blood , Glycine/pharmacokinetics , Glycine/therapeutic use , Humans , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/enzymology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Remission Induction , Sulfones/administration & dosage , Sulfones/adverse effects , Sulfones/blood , Sulfones/pharmacokinetics , Treatment Outcome , Urologic Diseases/chemically induced
20.
Biochem Biophys Res Commun ; 441(2): 431-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24369899

ABSTRACT

Although the kinase receptor TrkA may play an important role in acute myeloid leukemia (AML), its involvement in other types of leukemia has not been reported. Furthermore, how it contributes to leukemogenesis is unknown. Here, we describe a molecular network that is important for TrkA function in leukemogenesis. We found that TrkA is frequently overexpressed in other types of leukemia such as acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), and myelodysplastic syndrome (MDS) including AML. In addition, TrkA was overexpressed in patients with MDS or secondary AML evolving from MDS. TrkA induced significant hematological malignancies by inducing PLK-1 and Twist-1, and enhanced survival and proliferation of leukemia, which was correlated with activation of the phosphatidylinositol 3-kinase/Akt/mTOR pathway. Moreover, endogenous TrkA associated with c-Src complexes was detected in leukemia. Suppression of c-Src activation by TrkA resulted in markedly decreased expression of PLK-1 and Twist-1 via suppressed activation of Akt/mTOR cascades. These data suggest that TrkA plays a key role in leukemogenesis and reveal an unexpected physiological role for TrkA in the pathogenesis of leukemia. These data have important implications for understanding various hematological malignancies.


Subject(s)
Leukemia/enzymology , Leukemia/pathology , Receptor, trkA/metabolism , src-Family Kinases/metabolism , CSK Tyrosine-Protein Kinase , Cell Cycle Proteins/biosynthesis , Cell Proliferation , Enzyme Activation , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Metabolic Networks and Pathways , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology , Nuclear Proteins/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured , Twist-Related Protein 1/biosynthesis , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL