Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Hematol Oncol ; 38(4): 531-540, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32469434

ABSTRACT

Patients with acute myeloid leukemia (AML) evolving from myelodysplastic syndrome (MDS) or higher-risk MDS have limited treatment options and poor prognosis. Our previous single-center study of decitabine followed by low dose idarubicin and cytarabine (D-IA) in patients with myeloid neoplasms showed promising primary results. We therefore conducted a multicenter study of D-IA regimen in AML evolving from MDS and higher-risk MDS. Patients with AML evolving from MDS or refractory anemia with excess blasts type 2 (RAEB-2) (based on the 2008 WHO classification) were included. The D-IA regimen (decitabine, 20 mg/m2 daily, days 1 to 3; idarubicin, 6 mg/m2 daily, days 4 to 6; cytarabine 25 mg/m2 every 12 hours, days 4 to 8; granulocyte colony stimulating factor [G-CSF], 5 µg/kg, from day 4 until neutrophil count increased to 1.0 × 109 /L) was administered as induction chemotherapy. Seventy-one patients were enrolled and treated, among whom 44 (62.0%) had AML evolving from MDS and 27 (38.0%) had RAEB-2. Twenty-eight (63.6%) AML patients achieved complete remission (CR) or complete remission with incomplete blood count recovery (CRi): 14 (31.8%) patients had CR and 14 (31.8%) had CRi. Six (22.2%) MDS patients had CR and 15 (55.6%) had marrow complete remission. The median overall survival (OS) was 22.4 months for the entire group, with a median OS of 24.2 months for AML and 20.0 months for MDS subgroup. No early death occurred. In conclusion, the D-IA regimen was effective and well tolerated, representing an alternative option for patients with AML evolving from MDS or MDS subtype RAEB-2.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Clonal Evolution , Epigenesis, Genetic , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/pathology , Cytarabine/administration & dosage , Decitabine/administration & dosage , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Idarubicin/administration & dosage , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Prognosis , Prospective Studies , Survival Rate , Young Adult
2.
Blood ; 135(25): 2271-2285, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32202636

ABSTRACT

SETD2, the histone H3 lysine 36 methyltransferase, previously identified by us, plays an important role in the pathogenesis of hematologic malignancies, but its role in myelodysplastic syndromes (MDSs) has been unclear. In this study, low expression of SETD2 correlated with shortened survival in patients with MDS, and the SETD2 levels in CD34+ bone marrow cells of those patients were increased by decitabine. We knocked out Setd2 in NUP98-HOXD13 (NHD13) transgenic mice, which phenocopies human MDS, and found that loss of Setd2 accelerated the transformation of MDS into acute myeloid leukemia (AML). Loss of Setd2 enhanced the ability of NHD13+ hematopoietic stem and progenitor cells (HSPCs) to self-renew, with increased symmetric self-renewal division and decreased differentiation and cell death. The growth of MDS-associated leukemia cells was inhibited though increasing the H3K36me3 level by using epigenetic modifying drugs. Furthermore, Setd2 deficiency upregulated hematopoietic stem cell signaling and downregulated myeloid differentiation pathways in the NHD13+ HSPCs. Our RNA-seq and chromatin immunoprecipitation-seq analysis indicated that S100a9, the S100 calcium-binding protein, is a target gene of Setd2 and that the addition of recombinant S100a9 weakens the effect of Setd2 deficiency in the NHD13+ HSPCs. In contrast, downregulation of S100a9 leads to decreases of its downstream targets, including Ikba and Jnk, which influence the self-renewal and differentiation of HSPCs. Therefore, our results demonstrated that SETD2 deficiency predicts poor prognosis in MDS and promotes the transformation of MDS into AML, which provides a potential therapeutic target for MDS-associated acute leukemia.


Subject(s)
Anemia, Refractory, with Excess of Blasts/pathology , Calgranulin B/physiology , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/physiology , Leukemia, Myeloid, Acute/etiology , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Animals , Calgranulin B/biosynthesis , Calgranulin B/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Decitabine/pharmacology , Down-Regulation , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Histone Code/drug effects , Histone-Lysine N-Methyltransferase/biosynthesis , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelodysplastic Syndromes/pathology , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Prognosis , Recombinant Proteins/therapeutic use , Time Factors , Tissue Array Analysis , Transcriptome
4.
J Postgrad Med ; 63(3): 194-196, 2017.
Article in English | MEDLINE | ID: mdl-28272065

ABSTRACT

Erythrophagocytosis by leukemic blasts is a rare phenomenon. We report a case of a female diagnosed with acute monoblastic leukemia with leukemic blasts that were CD34 and CD117 negative, showing erythrophagocytosis, vacoulations, and a rare t(8;16) on bone marrow karyotype which is associated with a poor prognosis despite intensive chemotherapy. Meticulous bone marrow examination in such a scenario may point towards the presence of t(8;16) and help clinicians take a well-informed clinical decision.


Subject(s)
Anemia, Refractory, with Excess of Blasts/pathology , Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Adult , Anemia, Refractory, with Excess of Blasts/mortality , Chromosome Aberrations , Fatal Outcome , Female , Humans , Karyotype , Leukemia, Myeloid, Acute/mortality , Mutation
5.
Leuk Res ; 45: 82-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27107658

ABSTRACT

The present study aimed to compare the efficacy and safety of azacitidine and decitabine in patients with myelodysplastic syndrome (MDS). A total of 88 patients diagnosed with refractory anemia with excess blast (RAEB) treated with azacitidine (n=57) or decitabine (n=31) were evaluated. Comparisons between azacitidine and decitabine groups were performed in the whole cohort, and in a 1:1 propensity score-matched cohort in order to reduce the simple selection bias. Patients who received azacitidine or decitabine had comparable overall response rates in both the unmatched (49.1% vs. 64.5%, p=0.166) and the propensity-matched cohorts (52% vs. 68%, p=0.248). The cumulative incidence of AML transformation at one year was comparable between azacitidine and decitabine in the unmatched (24.0% vs. 31.3%, p=0.26) and in the propensity-matched cohorts (18.7% vs. 31.5%, p=0.11). There was no difference in terms of transfusion requirement, febrile neutropenia episodes or the need for antifungal use during the treatment cycles in the propensity-matched cohort. The median overall survival was 20.4 months for azacitidine and 16.8 months for decitabine (p=0.59). Finally, we found that at least a four-cycle treatment with any HMA was a favorable factor. In conclusion, both azacitidine and decitabine have similar efficacy and toxicity profiles in the treatment of MDS-RAEB.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Aged , Anemia, Refractory, with Excess of Blasts/complications , Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Refractory, with Excess of Blasts/pathology , Antimetabolites, Antineoplastic/adverse effects , Azacitidine/adverse effects , Blood Component Transfusion/statistics & numerical data , Cell Transformation, Neoplastic , Decitabine , Drug Evaluation , Female , Humans , Leukemia, Myeloid, Acute , Male , Middle Aged , Myelodysplastic Syndromes , Patient Selection , Propensity Score , Remission Induction , Retrospective Studies , Survival Rate
7.
Dermatol Online J ; 22(12)2016 Dec 15.
Article in English | MEDLINE | ID: mdl-28329538

ABSTRACT

HTLV-1 is a virus that is endemic in southwesternJapan and the Caribbean and has been implicatedin the development of ATLL. ATLL, which is anuncommon malignant condition of peripheralT-lymphocytes, is characterized by four clinicalsubtypes, which include acute, lymphomatous,chronic, and smoldering types, that are based onLDH levels, calcium levels, and extent of organinvolvement. We present a 52-year- old woman withpruritic patches with scale on the buttocks and withtender, hyperpigmented macules and papules oftwo-years duration. Histopathologic examinationwas suggestive of mycosis fungoides, laboratoryresults showed HTLV-I and II, and the patient wasdiagnosed with primary cutaneous ATLL. We reviewthe literature on HTLV-1 and ATLL and specifically theprognosis of cutaneous ATLL. The literature suggeststhat a diagnosis of ATLL should be considered amongpatients of Caribbean origin or other endemicareas with skin lesions that suggest a cutaneousT-cell lymphoma, with clinicopathologic features ofmycosis fungoides. Differentiation between ATLLand cutaneous T-cell lymphoma is imperative as theyhave different prognoses and treatment approaches.


Subject(s)
Anemia, Refractory, with Excess of Blasts/diagnosis , Leukemia-Lymphoma, Adult T-Cell/diagnosis , Skin Neoplasms/diagnosis , Anemia, Refractory, with Excess of Blasts/pathology , Anemia, Refractory, with Excess of Blasts/virology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , HTLV-I Antibodies/immunology , HTLV-II Antibodies/immunology , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 2/immunology , Humans , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Middle Aged , Skin Neoplasms/pathology , Skin Neoplasms/virology
8.
Eur J Haematol ; 94(5): 413-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25200248

ABSTRACT

TET2, a member of the ten-eleven-translocation (TET) family genes that modify DNA by converting 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC), is located in chromosome 4q24 and is frequently mutated in myeloid malignancies. The impact of TET2 mutation on survival outcomes is still controversial; however, functional studies have proved that it is a loss-of-function mutation that impairs myeloid cell differentiation and contributes to the phenotype of myeloid neoplasia. We, herein, aimed to investigate TET2 expression in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). A significantly decreased TET2 expression was observed in bone marrow cells from AML (n = 53) and patients with MDS (n = 64), compared to normal donors (n = 22). In MDS, TET2 expression was significantly reduced in RAEB-1/RAEB-2 compared to other WHO 2008 classifications, and a lower TET2 expression was observed at the time of MDS disease progression in four of five patients. In multivariate analysis, low TET2 expression (P = 0.03), male gender (P = 0.02), and WHO 2008 classification (P < 0.0001) were independent predictors of poorer overall survival. These results suggest that defective TET2 expression plays a role in the MDS pathophysiology and predicts survival outcomes in this disease.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Sideroblastic/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Refractory, with Excess of Blasts/pathology , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/mortality , Anemia, Sideroblastic/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Case-Control Studies , Chromosomes, Human, Pair 4 , Dioxygenases , Down-Regulation , Female , Gene Expression , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Multivariate Analysis , Mutation , Prognosis , Survival Analysis
9.
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
10.
Indian J Pathol Microbiol ; 57(3): 476-9, 2014.
Article in English | MEDLINE | ID: mdl-25118751

ABSTRACT

Use of automated hematology analyzers for routine blood count reporting has increased the reproducibility and accuracy of test results. However, at times, these instruments may generate spurious test results. Such results can result in inappropriate investigations or treatment decisions in patients. Spuriously normal or high platelet counts carry the risk of under diagnosis of the true thrombocytopenia with adverse clinical implications. We present a patient with smoldering myeloma with spurious platelet count due to cryoglobulins.


Subject(s)
Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/pathology , Cryoglobulins/metabolism , Diagnostic Errors , Platelet Count/methods , Automation, Laboratory/methods , Humans , Male , Middle Aged
12.
Am J Hematol ; 89(11): E193-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25042343

ABSTRACT

It is controversial whether acute myeloid leukemia (AML) patients with 20-29% bone marrow (BM) blasts, formerly referred to as refractory anemia with excess blasts in transformation (RAEBT), should be considered AML or myelodysplastic syndrome (MDS) for the purposes of treatment and prognostication. We retrospectively studied 571 de novo AML in patients aged >50 years, including 142 RAEBT and 429 with ≥30% blasts (AML30), as well as 151 patients with 10-19% BM blasts (RAEB2). RAEBT patients were older and had lower white blood count, but higher hemoglobin, platelet count, and karyotype risk scores compared to AML30, while these features were similar to RAEB2. FLT3 and NPM1 mutations and monocytic morphology occurred more commonly in AML30 than in RAEBT. RAEBT patients were treated less often with induction therapy than AML30, whereas allogeneic stem cell transplant frequency was similar. The median and 4-year OS of RAEBT patients were longer than those of AML30 patients (20.5 vs 12.0 months and 28.6% vs 20.4%, respectively, P = 0.003); this difference in OS was manifested in patients in the intermediate UKMRC karyotype risk group, whereas OS of RAEBT patients and AML30 patients in the adverse karyotype risk group were not significantly different. Multivariable analysis showed that RAEBT (P < 0.0001), hemoglobin (P = 0.005), UKMRC karyotype risk group (P = 0.002), normal BM karyotype (P = 0.004), treatment with induction therapy (P < 0.0001), and stem cell transplant (P < 0.0001) were associated with longer OS. Our findings favor considering de novo RAEBT as a favorable prognostic subgroup of AML.


Subject(s)
Anemia, Refractory, with Excess of Blasts/pathology , Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Age Factors , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Refractory, with Excess of Blasts/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Count , Chromosome Aberrations , DNA Methylation/drug effects , Female , Hematopoietic Stem Cell Transplantation , Humans , Kaplan-Meier Estimate , Karyotype , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Remission Induction , Retrospective Studies , Treatment Outcome , fms-Like Tyrosine Kinase 3/genetics
13.
Acta Haematol ; 132(2): 220-5, 2014.
Article in English | MEDLINE | ID: mdl-24714374

ABSTRACT

BACKGROUND: Sweet's syndrome (SS) is an acute febrile neutrophilic dermatosis. It can occur as an idiopathic, drug-induced or malignancy-associated entity. SS is also seen in patients with myelodysplastic syndrome (MDS) where it may present atypically, both clinically and histologically. In a few rare cases of MDS, lymphocytic infiltrates are the presenting feature of SS. METHODS: MEDLINE and Scopus were the data sources for our review. RESULTS: A clinicopathological subsetemerged of 12 male SS patients with MDS and a mean age of 67.3 years in which the initial SS lesions were lymphocytic infiltrates. However, from 0.5 to 8 years later, sequential biopsies revealed neutrophilic dermal infiltration typical of SS. CONCLUSION: Initially lymphocytic infiltrates in this subset could be attributed either to an early timing of the biopsy concerning the age of the lesion or to the dysgranulopoiesis syndrome. A possible relationship between the dysfunction of the receptor of the granulocyte-macrophage colony stimulating factor, the gene of which is located on the pseudoautosomal X-Y region, may exist in MDS patients with initially lymphocytic SS. This could explain the male gender of this subset and might establish initially lymphocytic SS as a distinguished clinicopathological entity for predicting the occurrence and even the prognosis of MDS.


Subject(s)
Myelodysplastic Syndromes/complications , Sweet Syndrome/etiology , Aged , Anemia, Refractory, with Excess of Blasts/complications , Anemia, Refractory, with Excess of Blasts/pathology , Biopsy , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Disease Progression , Humans , Lymphocytes/pathology , Male , Middle Aged , Models, Immunological , Myelodysplastic Syndromes/pathology , Neutrophil Infiltration , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Sex Factors , Skin/pathology , Sweet Syndrome/genetics , Sweet Syndrome/immunology , Sweet Syndrome/pathology
14.
Am J Hematol ; 89(8): E109-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24723457

ABSTRACT

The SF3B1 mutation can be detected in patients with myelodysplastic syndrome (MDS), but the report regarding the association of this mutation with other genetic alterations and its stability during disease progression is limited. In this study, SF3B1 mutations were identified in 10% of total cohort of 479 MDS patients and 61.8% of 34 patients with refractory anemia with ring sideroblasts (RARS). SF3B1 mutations were closely associated with older age, higher platelet counts, lower lactate dehydrogenase levels, good-risk cytogenetics, and mutations of DNMT3A, but inversely related to ASXL1 mutations. Most SF3B1-mutated patients had concurrent other genetic alterations, including DNMT3A and RUNX1 mutations. There was no prognostic difference between patients with SF3B1 mutations and those without. Sequential studies in 417 samples from 142 patients demonstrated that all SF3B1-mutated patients retained the same mutations during disease evolution with the exception of two patients who lost the mutation after allogeneic hematopoietic stem cell transplantation, whereas none of the SF3B1-wild patients acquired a novel mutation during clinical follow-ups. In conclusion, the patients with SF3B1 mutations had distinct clinic-biologic features. SF3B1 mutations, accompanied with other genetic alterations, especially DNMT3A mutations, may play a role in the development of MDS, but have little role in disease progression.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Mutation Rate , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/metabolism , Anemia, Refractory, with Excess of Blasts/pathology , Blood Platelets/metabolism , Blood Platelets/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Disease Progression , Female , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Phosphoproteins/metabolism , RNA Splicing Factors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism
15.
Acta Haematol ; 132(2): 134-9, 2014.
Article in English | MEDLINE | ID: mdl-24556628

ABSTRACT

We report the cases of 3 patients with hematological malignancies and complex karyotypes involving der(5; 17) (p10;q10), which results in the loss of 5q and 17p. Although deletions of 5q and 17p are recurrent abnormalities in hematological disease, only about 20 cases harboring der(5; 17) (p10;q10) have been reported. We address the tumorigenesis and morphological characteristics of hematological malignancies involving der(5; 17)(p10;q10), along with a review of the literature.


Subject(s)
Chromosomes, Human, Pair 17/ultrastructure , Chromosomes, Human, Pair 5/ultrastructure , Hematologic Neoplasms/genetics , Translocation, Genetic , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/drug therapy , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/pathology , Aneuploidy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Cells/ultrastructure , Cell Transformation, Neoplastic/genetics , Chromosome Aberrations , Chromosome Banding , Contraindications , Fatal Outcome , Female , Hematologic Neoplasms/pathology , Humans , Karyotype , Lenalidomide , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Lymphoma, T-Cell, Peripheral/drug therapy , Male , Megakaryocytes/ultrastructure , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Recurrence , Remission Induction , Thalidomide/analogs & derivatives
16.
Cytometry B Clin Cytom ; 86(6): 383-96, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24106013

ABSTRACT

BACKGROUND: Single Cell Network Profiling (SCNP) is a multiparametric flow cytometry-based assay that quantifiably and simultaneously measures changes in intracellular signaling proteins in response to in vitro extracellular modulators at the single cell level. Myelodysplastic syndrome (MDS) is a heterogeneous clonal disorder of hematopoietic stem cells that occurs in elderly subjects and is characterized by dysplasia and ineffective hematopoiesis. The functional responsiveness of MDS bone marrow (BM) hematopoietic cells, including functionally distinct myeloid and erythroid precursor subsets, to hematopoietic growth factors (HGF) and the relationship of modulated signaling to disease characteristics is poorly understood. METHODS: SCNP was used first to examine the effects of age on erythropoietin (EPO) and granulocyte colony stimulating factor (GCSF)-induced signaling in myeloid, nucleated red blood cells (nRBC), and CD34 expressing cell subsets in healthy BM (n = 15). SCNP was then used to map functional signaling profiles in low risk (LR) MDS (n = 7) for comparison to signaling in samples from healthy donors and to probe signaling associations within clinically defined subgroups. RESULTS: In healthy BM samples, signaling responses to HGF were quite homogeneous (i.e., tightly regulated) with age-dependent effects observed in response to EPO but not to GCSF. Despite the relatively small number of samples assayed in the study, LR MDS could be classified into distinct subgroups based on both cell subset frequency and signaling profiles. CONCLUSIONS: As a correlate of underlying genetic abnormalities, signal transduction analyses may provide a functional and potentially clinically relevant classification of MDS. Further evaluation in a larger cohort is warranted.


Subject(s)
Bone Marrow Cells/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Myelodysplastic Syndromes/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/metabolism , Anemia, Refractory, with Excess of Blasts/pathology , Bone Marrow Cells/pathology , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoietin/pharmacology , Flow Cytometry/methods , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Middle Aged , Myelodysplastic Syndromes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Signal Transduction , Young Adult
17.
An. pediatr. (2003, Ed. impr.) ; 78(6): 393-397, jun. 2013. tab
Article in Spanish | IBECS | ID: ibc-112821

ABSTRACT

Los síndromes mielodisplásicos (SMD) son desórdenes clonales de las células hematopoyéticas, con riesgo variable de transformación a leucemia mieloblástica aguda. La evolución a leucemia linfoblástica aguda (LLA) es extremadamente rara, con muy pocos casos pediátricos. Este artículo describe 2 nuevos casos de SMD que progresaron a LLA, junto a una revisión de la literatura, observando un pronóstico similar a los adultos (AU)


Myelodysplastic syndromes (MDS) are clonal disorders of hematopoietic stem cells, with a variable risk of transformation to acute myeloid leukemia. Progression into acute lymphoblastic leukemia (ALL) is an extremely rare event, with very few cases published in children. In this report, we describe two cases of myelodysplastic syndromes that progressed to ALL. Moreover, we review previously reported cases of MDS transformation to acute lymphoblastic leukemia in the pediatric population whose prognosis seems to be similar to that for adults (AU)


Subject(s)
Humans , Male , Female , Infant , Child, Preschool , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Myelodysplastic Syndromes/pathology , Anemia, Refractory, with Excess of Blasts/pathology , Fever/etiology , Risk Factors
18.
Ann Hematol ; 92(1): 19-24, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22948274

ABSTRACT

Morphology is the basis of the diagnosis of myelodysplastic syndromes (MDS). The WHO classification offers prognostic information and helps with the treatment decisions. However, morphological changes are subject to potential inter-observer variance. The aim of our study was to explore the reliability of the 2008 WHO classification of MDS, reviewing 100 samples previously diagnosed with MDS using the 2001 WHO criteria. Specimens were collected from 10 hospitals and were evaluated by 10 morphologists, working in five pairs. Each observer evaluated 20 samples, and each sample was analyzed independently by two morphologists. The second observer was blinded to the clinical and laboratory data, except for the peripheral blood (PB) counts. Nineteen cases were considered as unclassified MDS (MDS-U) by the 2001 WHO classification, but only three remained as MDS-U by the 2008 WHO proposal. Discordance was observed in 26 of the 95 samples considered suitable (27 %). Although there were a high number of observers taking part, the rate of discordance was quite similar among the five pairs. The inter-observer concordance was very good regarding refractory anemia with excess blasts type 1 (RAEB-1) (10 of 12 cases, 84 %), RAEB-2 (nine of 10 cases, 90 %), and also good regarding refractory cytopenia with multilineage dysplasia (37 of 50 cases, 74 %). However, the categories with unilineage dysplasia were not reproducible in most of the cases. The rate of concordance with refractory cytopenia with unilineage dysplasia was 40 % (two of five cases) and 25 % with RA with ring sideroblasts (two of eight). Our results show that the 2008 WHO classification gives a more accurate stratification of MDS but also illustrates the difficulty in diagnosing MDS with unilineage dysplasia.


Subject(s)
Bone Marrow Examination , Bone Marrow/pathology , Myelodysplastic Syndromes/diagnosis , Observer Variation , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/pathology , Biopsy , Cell Lineage , Chromosome Aberrations , Cytogenetic Analysis , Hematology , Humans , Laboratories, Hospital , Laboratory Proficiency Testing , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/pathology , Reproducibility of Results , Single-Blind Method , Spain , World Health Organization
19.
Exp Hematol ; 41(2): 172-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085464

ABSTRACT

Matrix metalloprotease-9 (MMP9) plays a critical role in acute myeloid leukemia (AML) by increasing the invasive properties of malignant myeloblasts. The role of this enzyme in high-risk myelodysplastic diseases (MDS) and the effect of azacitidine on its expression in MDS and AML have not been studied in detail. In this work, we have analyzed the effect of different concentrations of azacitidine in two well-established, MDS-derived, acute myeloid leukemic cell lines: MOLM-13 and SKM-1. We have demonstrated that 1 µmol/L azacitidine decreases MMP9 DNA methylation levels and that this is correlated with a significant increase in messenger RNA expression in both cell lines. Surprisingly, changes in protein levels were minor. This paradoxic effect is explained by the drug-dependent induction of apoptosis that reduces the amount of active secreting cells. A balance between induced expression and apoptosis was established at an azacitidine concentration of 0.2 µmol/L in MOLM-13 cells. This dose significantly increased the invasive capacity of viable cells, as measured in the Matrigel assay. To evaluate the clinical relevance of this observation, we have examined the effect of azacitidine on MMP9 expression in bone marrow from five patients with MDS, with the finding that this drug significantly increased MMP9 protein levels in all analyzed patients after six cycles of treatment. Based on these results, we conclude that azacitidine increases MMP9 expression and may enhance invasiveness in vitro. Because all five patients relapsed, these findings might explain, at least partially, the clinical failure of the drug and the progression to a more aggressive disease.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites/therapeutic use , Azacitidine/pharmacology , Leukemia, Monocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/pathology , Matrix Metalloproteinase 9/biosynthesis , Neoplasm Proteins/biosynthesis , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/enzymology , Anemia, Refractory, with Excess of Blasts/pathology , Antimetabolites/adverse effects , Apoptosis/drug effects , Azacitidine/adverse effects , Azacitidine/therapeutic use , Catalysis/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , DNA Methylation/drug effects , Disease Progression , Drug Resistance, Neoplasm , Enzyme Induction/drug effects , Female , Humans , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/enzymology , Neoplasms, Second Primary/pathology , Promoter Regions, Genetic/drug effects , Risk
SELECTION OF CITATIONS
SEARCH DETAIL