Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Lancet Haematol ; 7(6): e469-e478, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32470438

ABSTRACT

BACKGROUND: Transfusion-dependent haemoglobinopathies require lifelong iron chelation therapy with one of the three iron chelators (deferiprone, deferasirox, or deferoxamine). Deferasirox and deferiprone are the only two oral chelators used in adult patients with transfusion-dependent haemoglobinopathies. To our knowledge, there are no randomised clinical trials comparing deferiprone, a less expensive iron chelator, with deferasirox in paediatric patients. We aimed to show the non-inferiority of deferiprone versus deferasirox. METHODS: DEEP-2 was a phase 3, multicentre, randomised trial in paediatric patients (aged 1 month to 18 years) with transfusion-dependent haemoglobinopathies. The study was done in 21 research hospitals and universities in Italy, Egypt, Greece, Albania, Cyprus, Tunisia, and the UK. Participants were receiving at least 150 mL/kg per year of red blood cells for the past 2 years at the time of enrolment, and were receiving deferoxamine (<100 mg/kg per day) or deferasirox (<40 mg/kg per day; deferasirox is not registered for use in children aged <2 years so only deferoxamine was being used in these patients). Any previous chelation treatment was permitted with a 7-day washout period. Patients were randomly assigned 1:1 to receive orally administered daily deferiprone (75-100 mg/kg per day) or daily deferasirox (20-40 mg/kg per day) administered as dispersible tablets, both with dose adjustment for 12 months, stratified by age (<10 years and ≥10 years) and balanced by country. The primary efficacy endpoint was based on predefined success criteria for changes in serum ferritin concentration (all patients) and cardiac MRI T2-star (T2*; patients aged >10 years) to show non-inferiority of deferiprone versus deferasirox in the per-protocol population, defined as all randomly assigned patients who received the study drugs and had available data for both variables at baseline and after 1 year of treatment, without major protocol violations. Non-inferiority was based on the two-sided 95% CI of the difference in the proportion of patients with treatment success between the two groups and was shown if the lower limit of the two-sided 95% CI was greater than -12·5%. Safety was assessed in all patients who received at least one dose of study drug. This study is registered with EudraCT, 2012-000353-31, and ClinicalTrials.gov, NCT01825512. FINDINGS: 435 patients were enrolled between March 17, 2014, and June 16, 2016, 393 of whom were randomly assigned to a treatment group (194 to the deferiprone group; 199 to the deferasirox group). 352 (90%) of 390 patients had ß-thalassaemia major, 27 (7%) had sickle cell disease, five (1%) had thalassodrepanocytosis, and six (2%) had other haemoglobinopathies. Median follow-up was 379 days (IQR 294-392) for deferiprone and 381 days (350-392) for deferasirox. Non-inferiority of deferiprone versus deferasirox was established (treatment success in 69 [55·2%] of 125 patients assigned deferiprone with primary composite efficacy endpoint data available at baseline and 1 year vs 80 [54·8%] of 146 assigned deferasirox, difference 0·4%; 95% CI -11·9 to 12·6). No significant difference between the groups was shown in the occurrence of serious and drug-related adverse events. Three (2%) cases of reversible agranulocytosis occurred in the 193 patients in the safety analysis in the deferiprone group and two (1%) cases of reversible renal and urinary disorders (one case of each) occurred in the 197 patients in the deferasirox group. Compliance was similar between treatment groups: 183 (95%) of 193 patients in the deferiprone group versus 192 (97%) of 197 patients in the deferisirox group. INTERPRETATION: In paediatric patients with transfusion-dependent haemoglobinopathies, deferiprone was effective and safe in inducing control of iron overload during 12 months of treatment. Considering the need for availability of more chelation treatments in paediatric populations, deferiprone offers a valuable treatment option for this age group. FUNDING: EU Seventh Framework Programme.


Subject(s)
Deferasirox/therapeutic use , Deferiprone/therapeutic use , Erythrocyte Transfusion/methods , Hemoglobinopathies/drug therapy , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Administration, Oral , Adolescent , Agranulocytosis/chemically induced , Agranulocytosis/epidemiology , Albania/epidemiology , Anemia, Sickle Cell/therapy , Cardiac Imaging Techniques/methods , Child , Child, Preschool , Cyprus/epidemiology , Deferasirox/administration & dosage , Deferasirox/economics , Deferiprone/administration & dosage , Deferiprone/economics , Egypt/epidemiology , Erythrocyte Transfusion/statistics & numerical data , Female , Ferritins/blood , Ferritins/drug effects , Greece/epidemiology , Hemoglobinopathies/therapy , Humans , Infant , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/economics , Iron Overload/blood , Italy/epidemiology , Magnetic Resonance Imaging , Male , Patient Compliance , Treatment Outcome , Tunisia/epidemiology , United Kingdom/epidemiology , Urologic Diseases/chemically induced , Urologic Diseases/epidemiology , beta-Thalassemia/therapy
2.
Cancer Genet ; 235-236: 93-94, 2019 06.
Article in English | MEDLINE | ID: mdl-31155481

ABSTRACT

Isodicentric chromosome 15, also called idic(15), is a rare chromosomal abnormality resulting from inverted duplication of proximal 15q. It is associated with specific clinical findings such as early central hypotonia, developmental delay, cognitive dysfunction, autism spectrum disorders, and seizure. Herein we describe a case of a girl with idic(15) syndrome who developed acute lymphoblastic leukemia (ALL) at the age of 9 years. Our case suggests a possible correlation between idic(15) and ALL, and possible functional links between these two conditions.


Subject(s)
Chromosome Disorders/genetics , Chromosome Inversion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Autism Spectrum Disorder/genetics , Child , Chromosomes, Human, Pair 15/genetics , Female , Humans
4.
Int J Cardiovasc Imaging ; 34(2): 263-271, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28770456

ABSTRACT

We aimed to evaluate the role of two-dimensional speckle tracking imaging (2DSTI) in detecting early changes of myocardial deformation in patients affected by thalassemia major (TM) and its relation with myocardial iron overload (MIO) detected by T2* cardiovascular magnetic resonance (CMR). We studied 28 TM patients (15 males, 37.4 ± 10 years). All patients underwent CMR and echocardiography in the same day. Segmental and global T2* values were measured. Values of global longitudinal strain (GLS) were derived from the three apical views, while radial and circumferential strain were obtained as average strain from the short axis views at basal, mid and apical level. Six patients (21.4%) showed significant MIO (global heart T2* < 20 ms). GLS showed a significant correlation with T2* values (R = -0.49; P = 0.001) and it was significantly lower in patients with a significant MIO than in those with no significant MIO (-18.3 ± 2 vs. -21.3 ± 2.7, P = 0.02). No significant difference was found for radial and circumferential strain in relation to the severity of MIO. Patients with impaired GLS (<-19.5%) had a significant higher risk of showing significant MIO (Odds-ratio-OR = 17; 95%). GLS is related with global T2* in TM patients. Moreover, GLS can identify TM patients with severe MIO detected by CMR.


Subject(s)
Cardiomyopathies/diagnostic imaging , Echocardiography, Doppler , Iron Overload/diagnostic imaging , Iron/metabolism , Magnetic Resonance Imaging, Cine , Multimodal Imaging/methods , Myocardial Contraction , Myocardium/metabolism , beta-Thalassemia/complications , Adult , Area Under Curve , Biomechanical Phenomena , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Chi-Square Distribution , Early Diagnosis , Female , Humans , Iron Overload/etiology , Iron Overload/metabolism , Iron Overload/physiopathology , Logistic Models , Male , Middle Aged , Observer Variation , Odds Ratio , Predictive Value of Tests , ROC Curve , Reproducibility of Results , Risk Factors , Severity of Illness Index , beta-Thalassemia/diagnosis
5.
Br J Clin Pharmacol ; 83(3): 593-602, 2017 03.
Article in English | MEDLINE | ID: mdl-27641003

ABSTRACT

AIMS: Despite long clinical experience with deferiprone, there is limited information on its pharmacokinetics in children aged <6 years. Here we assess the impact of developmental growth on the pharmacokinetics of deferiprone in this population using a population approach. Based on pharmacokinetic bridging concepts, we also evaluate whether the recommended doses yield appropriate systemic exposure in this group of patients. METHODS: Data from a study in which 18 paediatric patients were enrolled were available for the purposes of this analysis. Patients were randomised to three deferiprone dose levels (8.3, 16.7 and 33.3 mg kg-1 ). Blood samples were collected according to an optimised sampling scheme in which each patient contributed to a maximum of five samples. A population pharmacokinetic model was developed using NONMEM v.7.2. Model selection criteria were based on graphical and statistical summaries. RESULTS: A one-compartment model with first-order absorption and first-order elimination best described the pharmacokinetics of deferiprone. Drug disposition parameters were affected by body weight, with both clearance and volume increasing allometrically with size. Simulation scenarios show that comparable systemic exposure (AUC) is achieved in children and adults after similar dose levels in mg kg-1 , with median (5-95th quantiles) AUC values, respectively, of 340.6 (223.2-520.0) µmol l-1  h and 318.5 (200.4-499.0) µmol l-1  h at 75 mg kg-1 day-1 , and 453.7 (297.3-693.0) µmol l-1  h and 424.2 (266.9-664.0) µmol l-1  h at 100 mg kg-1  day-1 given as three times daily (t.i.d.) doses. CONCLUSIONS: Based on the current findings, a dosing regimen of 25 mg kg-1  t.i.d. is recommended in children aged <6 years, with the possibility of titration up to 33.3 mg kg-1  t.i.d.


Subject(s)
Pyridones/administration & dosage , Pyridones/pharmacokinetics , Child, Preschool , Computer Simulation , Deferiprone , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infant , Iron Chelating Agents/pharmacokinetics , Male , Models, Biological , Pyridones/blood , Single-Blind Method
6.
Pharm Res ; 33(2): 498-509, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26555666

ABSTRACT

PURPOSE: Here we show how a model-based approach may be used to provide further insight into the role of clinical and demographic covariates on the progression of iron overload. The therapeutic effect of deferoxamine is used to illustrate the application of disease modelling as a means to characterising treatment response in individual patients. METHODS: Serum ferritin, demographic characteristics and individual treatment data from clinical routine practice on 27 patients affected by ß-thalassaemia major were used for the purposes of this analysis. The time course of serum ferritin was described by a hierarchical nonlinear mixed effects model, in which compliance was parameterised as a covariate factor. Modelling and simulation procedures were implemented in NONMEM (7.2.0). RESULTS: A turnover model best described serum ferritin changes over time, with the effect of blood transfusions introduced on the ferritin conversion rate and the effect of deferoxamine on the elimination parameter (Kout) in a proportional manner. The results of the simulations showed that poor quality of execution is preferable over drug holidays; and that independently of the compliance pattern, the therapeutic intervention is not effective if >60% of the doses are missed. CONCLUSIONS: Modelling of ferritin response enables characterisation of the dynamics of iron overload due to chronic transfusion. The approach can be used to support decision making in clinical practice, including personalisation of the dose for existing and novel chelating agents.


Subject(s)
Chelation Therapy/methods , Deferoxamine/therapeutic use , Ferritins/blood , Iron Overload/drug therapy , Siderophores/therapeutic use , beta-Thalassemia/therapy , Adolescent , Adult , Child , Computer Simulation , Deferoxamine/blood , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Erythrocyte Transfusion/adverse effects , Humans , Iron Overload/blood , Iron Overload/etiology , Models, Biological , Siderophores/blood , Siderophores/pharmacology , Young Adult , beta-Thalassemia/blood , beta-Thalassemia/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...