Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958467

ABSTRACT

Myelodysplastic syndromes/neoplasms (MDS) are clonal hematologic disorders characterized by morphologic abnormalities of myeloid cells and peripheral cytopenias. While genetic abnormalities underlie the pathogenesis of these disorders and their heterogeneity, current classifications of MDS rely predominantly on morphology. We performed genomic profiling of 3,233 patients with MDS or related disorders to delineate molecular subtypes and define their clinical implications. Gene mutations, copy-number alterations (CNAs), and copy-neutral loss of heterozygosity (cnLOH) were derived from targeted sequencing of a 152-gene panel, with abnormalities identified in 91, 43, and 11% of patients, respectively. We characterized 16 molecular groups, encompassing 86% of patients, using information from 21 genes, 6 cytogenetic events, and LOH at the TP53 and TET2 loci. Two residual groups defined by negative findings (molecularly not-otherwise specified, absence of recurrent drivers) comprised 14% of patients. The groups varied in size from 0.5% to 14% of patients and were associated with distinct clinical phenotypes and outcomes. The median bone marrow blast percentage across groups ranged from 1.5 to 10%, and the median overall survival from 0.9 to 8.2 years. We validated 5 well-characterized entities, added further evidence to support 3 previously reported subsets, and described 8 novel groups. The prognostic influence of bone marrow blasts depended on the genetic subtypes. Within genetic subgroups, therapy-related MDS and myelodysplastic/myeloproliferative neoplasms (MDS/MPN) had comparable clinical and outcome profiles to primary MDS. In conclusion, genetically-derived subgroups of MDS are clinically relevant and may inform future classification schemas and translational therapeutic research.

2.
Blood ; 2024 04 30.
Article in English | MEDLINE | ID: mdl-38687605

ABSTRACT

Mutations in UBA1, which are disease-defining for VEXAS syndrome, have been reported in patients diagnosed with myelodysplastic syndromes (MDS). Here, we define the prevalence and clinical associations of UBA1 mutations in a representative cohort of patients with MDS. Digital droplet PCR profiling of a selected cohort of 375 male patients lacking MDS disease-defining mutations or established WHO disease classification identified 28 patients (7%) with UBA1 p.M41T/V/L mutations. Using targeted sequencing of UBA1 in a representative MDS cohort (n=2,027), we identified an additional 27 variants in 26 patients (1%), which we classified as likely/pathogenic (n=12) and unknown significance (n=15). Among the total 40 patients with likely/pathogenic variants (2%), all were male and 63% were classified by WHO2016 as MDS-MLD/SLD. Patients had a median of one additional myeloid gene mutation, often in TET2 (n=12), DNMT3A (n=10), ASXL1 (n=3), or SF3B1 (n=3). Retrospective clinical review where possible showed that 83% (28/34) UBA1-mutant cases had VEXAS-associated diagnoses or inflammatory clinical presentation. The prevalence of UBA1-mutations in MDS patients argues for systematic screening for UBA1 in the management of MDS.

3.
N Engl J Med ; 386(21): 1998-2010, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35613022

ABSTRACT

BACKGROUND: Although hypomethylating agents are currently used to treat patients with cancer, whether they can also reactivate and up-regulate oncogenes is not well elucidated. METHODS: We examined the effect of hypomethylating agents on SALL4, a known oncogene that plays an important role in myelodysplastic syndrome and other cancers. Paired bone marrow samples that were obtained from two cohorts of patients with myelodysplastic syndrome before and after treatment with a hypomethylating agent were used to explore the relationships among changes in SALL4 expression, treatment response, and clinical outcome. Leukemic cell lines with low or undetectable SALL4 expression were used to study the relationship between SALL4 methylation and expression. A locus-specific demethylation technology, CRISPR-DNMT1-interacting RNA (CRISPR-DiR), was used to identify the CpG island that is critical for SALL4 expression. RESULTS: SALL4 up-regulation after treatment with hypomethylating agents was observed in 10 of 25 patients (40%) in cohort 1 and in 13 of 43 patients (30%) in cohort 2 and was associated with a worse outcome. Using CRISPR-DiR, we discovered that demethylation of a CpG island within the 5' untranslated region was critical for SALL4 expression. In cell lines and patients, we confirmed that treatment with a hypomethylating agent led to demethylation of the same CpG region and up-regulation of SALL4 expression. CONCLUSIONS: By combining analysis of patient samples with CRISPR-DiR technology, we found that demethylation and up-regulation of an oncogene after treatment with a hypomethylating agent can indeed occur and should be further studied. (Funded by Associazione Italiana per la Ricerca sul Cancro and others.).


Subject(s)
Antineoplastic Agents , Demethylation , Myelodysplastic Syndromes , Oncogenes , Up-Regulation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clustered Regularly Interspaced Short Palindromic Repeats , Demethylation/drug effects , Humans , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Oncogenes/drug effects , Oncogenes/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
4.
Br J Haematol ; 204(5): 2049-2056, 2024 May.
Article in English | MEDLINE | ID: mdl-38343073

ABSTRACT

Iron overload from repeated transfusions has a negative impact on cardiac function, and iron chelation therapy may help prevent cardiac dysfunction in transfusion-dependent patients with myelodysplastic syndromes (MDS). TELESTO (NCT00940602) was a prospective, placebo-controlled, randomised study to evaluate the iron chelator deferasirox in patients with low- or intermediate-1-risk MDS and iron overload. Echocardiographic parameters were collected at screening and during treatment. Patients receiving deferasirox experienced a significant decrease in the composite risk of hospitalisation for congestive heart failure (CHF) or worsening of cardiac function (HR = 0.23; 95% CI: 0.05, 0.99; nominal p = 0.0322) versus placebo. No significant differences between the arms were found in left ventricular ejection fraction, ventricular diameter and mass or pulmonary artery pressure. The absolute number of events was low, but the enrolled patients were younger than average for patients with MDS, with no serious cardiac comorbidities and a modest cardiovascular risk profile. These results support the effectiveness of deferasirox in preventing cardiac damage caused by iron overload in this patient population. Identification of patients developing CHF is challenging due to the lack of distinctive echocardiographic features. The treatment of iron overload may be important to prevent cardiac dysfunction in these patients, even those with moderate CHF risk.


Subject(s)
Deferasirox , Iron Chelating Agents , Iron Overload , Myelodysplastic Syndromes , Humans , Deferasirox/therapeutic use , Myelodysplastic Syndromes/therapy , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/complications , Male , Female , Iron Chelating Agents/therapeutic use , Middle Aged , Aged , Iron Overload/etiology , Iron Overload/drug therapy , Prospective Studies , Benzoates/therapeutic use , Benzoates/adverse effects , Heart Failure/etiology , Transfusion Reaction/etiology , Echocardiography , Adult , Aged, 80 and over , Triazoles/therapeutic use , Triazoles/adverse effects , Blood Transfusion
5.
Br J Haematol ; 204(5): 1838-1843, 2024 May.
Article in English | MEDLINE | ID: mdl-38471524

ABSTRACT

Real-world data have revealed that a substantial portion of patients with myelodysplastic syndromes (MDS) does not respond to epigenetic therapy with hypomethylating agents (HMAs). The cellular and molecular reasons for this resistance to the demethylating agent and biomarkers that would be able to predict the treatment refractoriness are largely unknown. In this study, we shed light on this enigma by characterizing the epigenomic profiles of patients with MDS treated with azacitidine. Our approach provides a comprehensive view of the evolving DNA methylation architecture of the disease and holds great potential for advancing our understanding of MDS treatment responses to HMAs.


Subject(s)
Azacitidine , DNA Methylation , Myelodysplastic Syndromes , Humans , Azacitidine/therapeutic use , Azacitidine/pharmacology , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Retrospective Studies , Male , Female , Aged , Middle Aged , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/pharmacology , Aged, 80 and over , Epigenesis, Genetic/drug effects , Treatment Outcome
6.
N Engl J Med ; 382(2): 140-151, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31914241

ABSTRACT

BACKGROUND: Patients with anemia and lower-risk myelodysplastic syndromes in whom erythropoiesis-stimulating agent therapy is not effective generally become dependent on red-cell transfusions. Luspatercept, a recombinant fusion protein that binds transforming growth factor ß superfamily ligands to reduce SMAD2 and SMAD3 signaling, showed promising results in a phase 2 study. METHODS: In a double-blind, placebo-controlled, phase 3 trial, we randomly assigned patients with very-low-risk, low-risk, or intermediate-risk myelodysplastic syndromes (defined according to the Revised International Prognostic Scoring System) with ring sideroblasts who had been receiving regular red-cell transfusions to receive either luspatercept (at a dose of 1.0 up to 1.75 mg per kilogram of body weight) or placebo, administered subcutaneously every 3 weeks. The primary end point was transfusion independence for 8 weeks or longer during weeks 1 through 24, and the key secondary end point was transfusion independence for 12 weeks or longer, assessed during both weeks 1 through 24 and weeks 1 through 48. RESULTS: Of the 229 patients enrolled, 153 were randomly assigned to receive luspatercept and 76 to receive placebo; the baseline characteristics of the patients were balanced. Transfusion independence for 8 weeks or longer was observed in 38% of the patients in the luspatercept group, as compared with 13% of those in the placebo group (P<0.001). A higher percentage of patients in the luspatercept group than in the placebo group met the key secondary end point (28% vs. 8% for weeks 1 through 24, and 33% vs. 12% for weeks 1 through 48; P<0.001 for both comparisons). The most common luspatercept-associated adverse events (of any grade) included fatigue, diarrhea, asthenia, nausea, and dizziness. The incidence of adverse events decreased over time. CONCLUSIONS: Luspatercept reduced the severity of anemia in patients with lower-risk myelodysplastic syndromes with ring sideroblasts who had been receiving regular red-cell transfusions and who had disease that was refractory to or unlikely to respond to erythropoiesis-stimulating agents or who had discontinued such agents owing to an adverse event. (Funded by Celgene and Acceleron Pharma; MEDALIST ClinicalTrials.gov number, NCT02631070; EudraCT number, 2015-003454-41.).


Subject(s)
Activin Receptors, Type II/therapeutic use , Anemia, Sideroblastic/drug therapy , Erythrocyte Transfusion , Hematinics/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Myelodysplastic Syndromes/drug therapy , Recombinant Fusion Proteins/therapeutic use , Activin Receptors, Type II/adverse effects , Adult , Aged , Aged, 80 and over , Anemia, Sideroblastic/therapy , Double-Blind Method , Female , Hematinics/adverse effects , Hemoglobins/analysis , Humans , Immunoglobulin Fc Fragments/adverse effects , Infusions, Subcutaneous , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Recombinant Fusion Proteins/adverse effects
7.
Cancer ; 127(12): 2015-2024, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33739457

ABSTRACT

BACKGROUND: Azacitidine (AZA) is the standard treatment for myelodysplastic syndromes (MDS); however, many patients prematurely stop therapy and have a dismal outcome. METHODS: The authors analyzed outcomes after AZA treatment for 402 MDS patients consecutively enrolled in the Italian MDS Registry of the Fondazione Italiana Sindromi Mielodisplastiche, and they evaluated the North American MDS Consortium scoring system in a clinical practice setting. RESULTS: At treatment discontinuation, 20.3% of the patients were still responding to AZA, 35.4% of the cases had primary resistance, and 44.3% developed adaptive resistance. Overall survival (OS) was better for patients who discontinued treatment while in response because of planned allogeneic hematopoietic stem cell transplantation (HSCT; median OS, not reached) in comparison with patients with primary resistance (median OS, 4 months) or adaptive resistance (median OS, 5 months) or patients responsive but noncompliant/intolerant to AZA (median OS, 4 months; P = .004). After AZA discontinuation, 309 patients (77%) received best supportive care (BSC), 60 (15%) received active treatments, and 33 (8%) received HSCT. HSCT was associated with a significant survival advantage, regardless of the response to AZA. The North American MDS Consortium scoring system was evaluable in 278 of the 402 cases: patients at high risk had worse OS than patients at low risk (3 and 7 months, respectively; P < .001). The score was predictive of survival both in patients receiving BSC (median OS, 2 months for high-risk patients vs 5 months for low-risk patients) and in patients being actively treated (median OS, 8 months for high-risk patients vs 16 months for low-risk patients; P < .001), including transplant patients. CONCLUSIONS: Real-life data confirm that this prognostic scoring system for MDS patients failing a hypomethylating agent seems to be a useful tool for optimal prognostic stratification and for choosing a second-line treatment after AZA discontinuation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes , Antimetabolites, Antineoplastic , Azacitidine , Humans , Myelodysplastic Syndromes/therapy , North America , Treatment Outcome
8.
FASEB J ; 34(11): 15400-15416, 2020 11.
Article in English | MEDLINE | ID: mdl-32959428

ABSTRACT

MDS are characterized by anemia and transfusion requirements. Transfused patients frequently show iron overload that negatively affects hematopoiesis. Iron chelation therapy can be effective in these MDS cases, but the molecular consequences of this treatment need to be further investigated. That is why we studied the molecular features of iron effect and Deferasirox therapy on PI-PLCbeta1 inositide signaling, using hematopoietic cells and MDS samples. At baseline, MDS patients showing a positive response after iron chelation therapy displayed higher levels of PI-PLCbeta1/Cyclin D3/PKCalpha expression. During treatment, these responder patients, as well as hematopoietic cells treated with FeCl3 and Deferasirox, showed a specific reduction of PI-PLCbeta1/Cyclin D3/PKCalpha expression, indicating that this signaling pathway is targeted by Deferasirox. The treatment was also able to specifically decrease the production of ROS. This effect correlated with a reduction of IL-1A and IL-2, as well as Akt/mTOR phosphorylation. In contrast, cells exposed only to FeCl3 and cells from MDS patients refractory to Deferasirox showed a specific increase of ROS and PI-PLCbeta1/Cyclin D3/PKCalpha expression. All in all, our data show that PI-PLCbeta1 signaling is a target for iron-induced oxidative stress and suggest that baseline PI-PLCbeta1 quantification could predict iron chelation therapy response in MDS.


Subject(s)
Cyclin D3/metabolism , Iron Overload/complications , Iron/adverse effects , Myelodysplastic Syndromes/therapy , Oxidative Stress/drug effects , Phospholipase C beta/metabolism , Protein Kinase C-alpha/metabolism , Aged , Blood Transfusion/statistics & numerical data , Cyclin D3/genetics , Deferasirox/pharmacology , Female , Gene Expression Regulation , Humans , Iron Chelating Agents/pharmacology , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Phospholipase C beta/genetics , Phosphorylation , Protein Kinase C-alpha/genetics , Signal Transduction
9.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467674

ABSTRACT

Erythropoiesis regulation is essential in normal physiology and pathology, particularly in myelodysplastic syndromes (MDS) and ß-thalassemia. Several signaling transduction processes, including those regulated by inositides, are implicated in erythropoiesis, and the latest MDS or ß-thalassemia preclinical and clinical studies are now based on their regulation. Among others, the main pathways involved are those regulated by transforming growth factor (TGF)-ß, which negatively regulates erythrocyte differentiation and maturation, and erythropoietin (EPO), which acts on the early-stage erythropoiesis. Also small mother against decapentaplegic (SMAD) signaling molecules play a role in pathology, and activin receptor ligand traps are being investigated for future clinical applications. Even inositide-dependent signaling, which is important in the regulation of cell proliferation and differentiation, is specifically associated with erythropoiesis, with phospholipase C (PLC) and phosphatidylinositol 3-kinase (PI3K) as key players that are becoming increasingly important as new promising therapeutic targets. Additionally, Roxadustat, a new erythropoiesis stimulating agent targeting hypoxia inducible factor (HIF), is under clinical development. Here, we review the role and function of the above-mentioned signaling pathways, and we describe the state of the art and new perspectives of erythropoiesis regulation in MDS and ß-thalassemia.


Subject(s)
Erythropoiesis , Myelodysplastic Syndromes/metabolism , Signal Transduction , beta-Thalassemia/metabolism , Animals , Cell Differentiation , Cell Proliferation , Clinical Trials as Topic , Erythropoietin/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Hematinics/therapeutic use , Humans , Hypoxia-Inducible Factor 1/metabolism , Isoquinolines/pharmacology , Ligands , Mice , Phosphatidylinositol 3-Kinases/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Type C Phospholipases/metabolism
11.
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
12.
Br J Haematol ; 177(5): 741-750, 2017 06.
Article in English | MEDLINE | ID: mdl-28419408

ABSTRACT

Iron chelation is controversial in higher risk myelodysplastic syndromes (HR-MDS), outside the allogeneic transplant setting. We conducted a retrospective, multicentre study in 51 patients with transfusion-dependent, intermediate-to-very high risk MDS, according to the revised international prognostic scoring system, treated with the oral iron chelating agent deferasirox (DFX). Thirty-six patients (71%) received azacitidine concomitantly. DFX was given at a median dose of 1000 mg/day (range 375-2500 mg) for a median of 11 months (range 0·4-75). Eight patients (16%) showed grade 2-3 toxicities (renal or gastrointestinal), 4 of whom (8%) required drug interruption. Median ferritin levels decreased from 1709 µg/l at baseline to 1100 µg/l after 12 months of treatment (P = 0·02). Seventeen patients showed abnormal transaminase levels at baseline, which improved or normalized under DFX treatment in eight cases. One patient showed a remarkable haematological improvement. At a median follow up of 35·3 months, median overall survival was 37·5 months. The results of this first survey of DFX in HR-MDS are comparable, in terms of safety and efficacy, with those observed in lower-risk MDS. Though larger, prospective studies are required to demonstrate real clinical benefits, our data suggest that DFX is feasible and might be considered in a selected cohort of HR-MDS patients.


Subject(s)
Benzoates/therapeutic use , Chelation Therapy/methods , Iron Chelating Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Triazoles/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Deferasirox , Erythrocyte Transfusion/statistics & numerical data , Female , Ferritins/metabolism , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome
13.
Am J Hematol ; 92(10): 1037-1046, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28675513

ABSTRACT

BACKGROUND: In 'real-life', the Nordic score guides Erythropoietic stimulating agent (ESA) use in lower-risk myelodysplastic syndrome (MDS) with predicted response rates of 25% or 74%. As new treatments emerge, a more discriminating score is needed. OBJECTIVES: To validate existing ESA predictive scores and develop a new score that identifies non-responders. METHODS: ESA-treated patients were identified in 3 MDS registries in Italy and Canada (FISM 555, GROM 233, and MDS-CAN 208). Clinical and disease-related variables were captured. Nordic, MDS-CAN, and IPSS-R-based ESA scores were calculated and documented ESA responses compared. RESULTS: 996 ESA-treated patients were identified. Overall response rate (ORR) was 59%. The database was randomly divided into balanced derivation (n = 463) and validation (n = 462) cohorts. By multivariate analysis, transfusion independence, erythropoietin (EPO) level <100 IU/L, and IPSS low-risk were independently predictive of response. Assigning a score of 1 to each resulted in a scoring system of 0-3 with response rates of 23%, 43%, 67%, and 85%. ORR was concordant in the validation cohort. The 'ITACA' score had the highest discriminating power of response. CONCLUSION: ITACA is an internally-validated predictive SS of ESA response in real-life 'good risk' MDS patients derived from a large international dataset that surpasses others. The incorporation of biologic markers to better identify non-responders is still needed.


Subject(s)
Hematinics/therapeutic use , Myelodysplastic Syndromes , Aged , Aged, 80 and over , Canada/epidemiology , Databases, Factual , Female , Humans , International Cooperation , Italy/epidemiology , Logistic Models , Male , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/mortality , Predictive Value of Tests , Prognosis , Prospective Studies , Registries , Survival Rate
17.
Eur J Haematol ; 92(6): 527-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24580147

ABSTRACT

BACKGROUND: In the absence of randomized, controlled trial data to support iron chelation therapy in transfusion-dependent patients with myelodysplastic syndromes (MDS), continued evidence from large prospective clinical trials evaluating the efficacy and safety of iron chelation therapy in this patient population is warranted. METHODS: The safety and efficacy of deferasirox was examined in a prospective, open-label, single-arm, multicenter trial of transfusion-dependent patients with International Prognostic Scoring System low- or intermediate-1-risk MDS and evidence of transfusion-related iron overload. The effects of deferasirox therapy on hematological response and disease progression were also examined. RESULTS: Of 159 participants enrolled from 37 Italian centers, 152 received ≥1 dose of deferasirox (initiated at 10-20 mg/kg/day and titrated as appropriate), and 68 completed the study. Of 84 patients who discontinued deferasirox therapy, 22 died during the trial, and 28 withdrew due to an adverse event (AE). Fourteen treatment-related grade 3 AEs occurred in 11 patients, whereas no grade 4 or 5 drug-related AEs were reported. Significant risks for dropout were a higher serum ferritin level at baseline, a higher MDS-Specific Comorbidity Index, and a shorter diagnosis-enrollment interval. Median serum ferritin level fell from 1966 ng/mL to 1475 ng/mL (P < 0.0001). The cumulative incidence of transfusion independence, adjusted for death and disease progression, was 2.6%, 12.3%, and 15.5% after 6, 9, and 12 months, respectively. CONCLUSIONS: Deferasirox therapy in transfusion-dependent patients with MDS was moderately well tolerated and effectively lowered serum ferritin levels. Positive hematological responses were observed, and a subset of patients achieved transfusion independence.


Subject(s)
Benzoates/therapeutic use , Blood Transfusion , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Iron Overload/etiology , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/therapy , Triazoles/therapeutic use , Adult , Aged , Aged, 80 and over , Benzoates/adverse effects , Deferasirox , Female , Ferritins/blood , Humans , Iron Chelating Agents/adverse effects , Male , Middle Aged , Risk Factors , Transfusion Reaction , Treatment Outcome , Triazoles/adverse effects , Young Adult
18.
Recenti Prog Med ; 105(3): 123-6, 2014 Mar.
Article in Italian | MEDLINE | ID: mdl-24675455

ABSTRACT

Patients affected by myelodysplastic syndromes (MDS) with transfusion-dependent anemia are destined to develop iron overload. The main diagnostic tools for the diagnosis of transfusional iron overload are serum ferritin and transfusion history. In MDS several studies showed that iron overload is an independent negative prognostic factor. Deferasirox, an oral iron chelator, has shown efficacy and acceptable tolerability in MDS setting, and has also been shown to improve peripheral cytopenia in 10-20% of patients. Iron chelation therapy is recommended, after the transfusion of 20 red cell units, in low-risk MDS patients, and also in high-risk patients responding to treatment of the disease and/or candidates to receive allogeneic hematopoietic stem cell transplantation.


Subject(s)
Blood Transfusion/methods , Iron Overload/drug therapy , Iron/metabolism , Myelodysplastic Syndromes/therapy , Anemia/etiology , Anemia/therapy , Benzoates/therapeutic use , Chelation Therapy/methods , Deferasirox , Ferritins/blood , Hematopoietic Stem Cell Transplantation/methods , Humans , Iron Chelating Agents/therapeutic use , Iron Overload/diagnosis , Iron Overload/etiology , Risk , Transfusion Reaction , Triazoles/therapeutic use
19.
Recenti Prog Med ; 105(3): 118-22, 2014 Mar.
Article in Italian | MEDLINE | ID: mdl-24675454

ABSTRACT

Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic disorders of the myeloid lineage characterized by peripheral cytopenias and frequent leukemic evolution. MDS differ for clinical presentation, disease behavior and progression and this is the reflection of remarkable variability at molecular level. To this moment disease diagnosis is still dependent on bone marrow morphology that, although high concordance rates among experts are reported, remains subjective. Karyotype analysis is mandatory but diagnosis may be difficult in presence of normal karyotype or non-informative cytogenetics. Standardized molecular markers are needed to better define diagnosis, prediction of disease progression and prognosis. Furthermore, a molecular biology analysis could provide an important therapeutic tool towards tailored therapy and new insights in the disease's biology.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Molecular Biology/methods , Myelodysplastic Syndromes/diagnosis , Biomarkers/metabolism , Bone Marrow/pathology , Cytogenetic Analysis , Disease Progression , Humans , Karyotyping/methods , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/pathology , Prognosis
20.
Methods Mol Biol ; 2777: 163-176, 2024.
Article in English | MEDLINE | ID: mdl-38478343

ABSTRACT

The hierarchical organization of the leukemic stem cells (LSCs) is identical to that of healthy counterpart cells. It may be split into roughly three stages: a small number of pluripotent stem cells at the top, few lineage-restricted cells in the middle, and several terminally differentiated blood cells at the bottom. Although LSCs can differentiate into the hematopoietic lineage, they can also accumulate as immature progenitor cells, also known as blast cells. Since blast cells are uncommon in healthy bloodstreams, their presence might be a sign of cancer. For instance, a 20% blast cutoff in peripheral blood or bone marrow is formally used to distinguish acute myeloid leukemia from myelodysplastic neoplasms, which is essential to plan the patients' management. Many techniques may be useful for blast enumeration: one of them is flow cytometry, which can perform analyses on many cells by detecting the expression of cell surface markers. Leukemic and non-leukemic blast cells might indeed be characterized by the same surface markers, but these markers are usually differently expressed. Here we propose to use CD45, in combination with CD34 and other cell surface markers, to identify and immunophenotype blast cells in patient-derived samples.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/genetics , Bone Marrow/metabolism , Antigens, CD34/metabolism , Flow Cytometry/methods , Neoplastic Stem Cells/metabolism , Immunophenotyping
SELECTION OF CITATIONS
SEARCH DETAIL