Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 386(5): 415-427, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34891223

ABSTRACT

BACKGROUND: Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent ß-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the ß-globin (ßA-T87Q) gene. METHODS: In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent ß-thalassemia and a non-ß0/ß0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS: A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS: Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-ß0/ß0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).


Subject(s)
Biological Products/therapeutic use , Genetic Therapy/methods , beta-Globins/genetics , beta-Thalassemia/therapy , Adolescent , Adult , Biological Products/adverse effects , Busulfan/therapeutic use , Child , Erythrocyte Transfusion/adverse effects , Erythropoiesis , Female , Genetic Vectors , Genotype , Hemoglobins/analysis , Humans , Iron Overload/prevention & control , Lentivirus/genetics , Male , Middle Aged , Myeloablative Agonists/therapeutic use , beta-Thalassemia/blood , beta-Thalassemia/genetics
2.
Br J Haematol ; 201(2): 334-342, 2023 04.
Article in English | MEDLINE | ID: mdl-36606625

ABSTRACT

ß-thalassemia is an haemoglobinopathy characterized by a defective synthesis of the ß-globin chain. To assess the current state of health of paediatric patients with ß-thalassemia, data from the French national registry regarding children born between 2005 and 2020 with ß-thalassemia intermedia (TI) or major (TM) were collected. A total of 237 patients (median age 7.1 years at last visit) were analysed, of whom 156 (65.8%) were born in France and 162 (68.4%) had a TM phenotype. The probability of survival for children with TM born in France was 98.3% at 15 years. Fifty-four (22.8%) children received a haematopoietic stem cell transplant with a success rate of 88.8%. Hepatic and cardiac iron overload monitoring in non-transplanted patients showed moderate overload in 15.7% (18/115) and 7.1% (7/99) of cases, respectively, while clinical complications were found in only 4 patients with TM (hepatic in 3 cases). At last visit, mean ferritinemia was 1293 ng/ml (±759). Overall, less than 10% of children underwent splenectomy. No significant impact of the disease on growth or academic achievement was observed. Deferasirox was the main first-line chelator, prescribed in 78.2% of cases, with side effects reported in 11.7% of instances.


Subject(s)
Hemoglobinopathies , Iron Overload , beta-Thalassemia , Humans , beta-Thalassemia/complications , beta-Thalassemia/epidemiology , beta-Thalassemia/therapy , Iron Overload/etiology , Iron Overload/complications , Phenotype , Hemoglobinopathies/complications , France
3.
Ann Hematol ; 101(2): 289-296, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34668980

ABSTRACT

Because of chronic anemia, hypogonadotropic hypogonadism, and iron chelation, pregnancy in homozygous and heterozygous compound beta-thalassemia patients stays a challenge. Pregnancies of transfused beta-thalassemia women registered in the French National Registry, conducted between 1995 and 2015, are described. These pregnancies were compared with pregnancies in healthy women and to data previously published in the literature. Fifty-six pregnancies of 37 women were studied. There were 5 twin pregnancies. Assisted reproductive technologies (ART) were used in 9 pregnancies. Median term at delivery was 39 amenorrhea weeks, and median weight at birth was 2780 g. Cesarean section was performed in 53.6% of the pregnancies. There were 6 thromboembolic events, 6 serious infections, 6 pregnancy-induced hypertensions (PIH), 6 intrauterine growth retardations (IUGR), 5 severe hemorrhages, 4 gestational diabetes, 3 alloimmunizations, 2 heart diseases, and 1 pre-eclampsia. There were 5 infections and 4 osteoporosis in the first year of post-partum. ART and cesarean sections were more often used in the beta-thalassemia group, compared to control subjects. Thromboembolic events, PIH, hemorrhage at delivery, and IUGR were more frequent in the beta-thalassemia group. Time to delivery was not different, but infant weight at birth was significantly smaller in the beta-thalassemia group. In the post-partum period, global maternal complications were more frequent in the beta-thalassemia group. Pregnancy in transfused beta-thalassemia women is safe with rare obstetrical and fetal complications. Cesarean section remains often chosen, and infant weight at birth remains smaller than that in the general population, despite delivery at full term.


Subject(s)
Pregnancy Complications, Hematologic/therapy , beta-Thalassemia/therapy , Adult , Cesarean Section , Cross-Sectional Studies , Erythrocyte Transfusion , Female , Fetal Growth Retardation/etiology , France/epidemiology , Humans , Infant, Newborn , Pregnancy , Pregnancy Complications, Hematologic/epidemiology , Pregnancy Outcome , Retrospective Studies , beta-Thalassemia/complications , beta-Thalassemia/epidemiology
4.
Br J Haematol ; 193(1): 188-193, 2021 04.
Article in English | MEDLINE | ID: mdl-33216975

ABSTRACT

We report here the 3-year stenosis outcome in 60 stroke-free children with sickle cell anaemia (SCA) and an abnormal transcranial Doppler history, enrolled in the DREPAGREFFE trial, which compared stem cell transplantation (SCT) with standard-care (chronic transfusion for 1-year minimum). Twenty-eight patients with matched sibling donors were transplanted, while 32 remained on standard-care. Stenosis scores were calculated after performing cerebral/cervical 3D time-of-flight magnetic resonance angiography. Fourteen patients had stenosis at enrollment, but only five SCT versus 10 standard-care patients still had stenosis at 3 years. Stenosis scores remained stable on standard-care, but significantly improved after SCT (P = 0·006). No patient developed stenosis after SCT, while two on standard-care did, indicating better stenosis prevention and improved outcome after SCT.


Subject(s)
Anemia, Sickle Cell/therapy , Blood Transfusion/statistics & numerical data , Brain/diagnostic imaging , Constriction, Pathologic/epidemiology , Hematopoietic Stem Cell Transplantation/adverse effects , Adolescent , Anemia, Sickle Cell/pathology , Blood Donors/statistics & numerical data , Blood Transfusion/standards , Brain/blood supply , Child , Child, Preschool , Constriction, Pathologic/diagnostic imaging , Constriction, Pathologic/etiology , Follow-Up Studies , Humans , Magnetic Resonance Angiography/methods , Magnetic Resonance Imaging , Outcome Assessment, Health Care , Prospective Studies , Siblings , Stroke/epidemiology , Stroke/prevention & control , Ultrasonography, Doppler, Transcranial/statistics & numerical data
5.
Haematologica ; 105(1): 91-101, 2020 01.
Article in English | MEDLINE | ID: mdl-31097628

ABSTRACT

Allogeneic stem cell transplantation remains the only curative treatment for sickle cell anemia (SCA), but the place of myeloablative conditioning in the procedure remains to be defined. The aim of the present study was to analyze long-term outcomes, including chimerism, SCA-related events and biological data (hemoglobin, reticulocytes, HbS%), and fertility in a French series of 234 SCA patients under 30 years of age who, from 1988 to 2012, received a matched-sibling-donor stem cell transplantation following standardized myeloablative conditioning [busulfan, cyclophosphamide and rabbit antithymocyte globulin (ATG)]. Since the first report of the series (1988-2004), 151 new consecutive patients with SCA have been similarly transplanted. Considering death, non-engraftment or rejection (donor cells <5%) as events, the 5-year event-free survival was 97.9% (95% confidence interval: 95.5-100%), confirming, since the year 2000, an at least 95% chance of cure. In the overall cohort (n=234, median follow up 7.9 years), event-free survival was not associated with age, but chronic-graft-versus-host disease (cGvHD) was independently associated with recipient's age >15 years (hazard ratio=4.37; P=0.002) and lower (5-15 vs 20 mg/kg) ATG dose (hazard ratio=4.55; P=0.001). At one year, 44% of patients had mixed chimerism (5-95% donor cells), but those prepared with ATG had no graft rejection. No events related to SCA occurred in patients with mixed chimerism, even those with 15-20% donor cells, but hemolytic anemia stigmata were observed with donor cells <50%. Myeloablative transplantation with matched-sibling donor currently has a higher event-free survival (98%) in patients under 30 years of age than that reported for non-myeloablative conditioning (88%). Nevertheless, the risk of cGvHD in older patients and the need to preserve fertility might be indications for a non-myeloablative conditioning.


Subject(s)
Anemia, Sickle Cell , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Aged , Anemia, Sickle Cell/therapy , Chimerism , Fertility , France/epidemiology , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Progression-Free Survival , Siblings , Transplantation Conditioning
6.
Br J Haematol ; 187(4): 530-542, 2019 11.
Article in English | MEDLINE | ID: mdl-31338833

ABSTRACT

Congenital sideroblastic anaemia (CSA) is a rare disease caused by germline mutations of genes involved in haem and iron-sulphur cluster formation, and mitochondrial protein biosynthesis. We performed a retrospective multicentre European study of a cohort of childhood-onset CSA patients to explore genotype/phenotype correlations. We studied 23 females and 20 males with symptoms of CSA. Among the patients, the most frequently mutated genes were ALAS2 (n = 10; 23·3%) and SLC25A38 (n = 8; 18·6%), causing isolated forms of microcytic anaemia of varying severity. Five patients with SLC19A2 mutations suffered from thiamine-responsive megaloblastic anaemia and three exhibited the 'anaemia, deafness and diabetes' triad. Three patients with TRNT1 mutations exhibited severe early onset microcytic anaemia associated with thrombocytosis, and two exhibited B-cell immunodeficiency, inflammatory syndrome and psychomotor delay. The prognoses of patients with TRNT1 and SLC2A38 mutations were generally dismal because of comorbidities or severe iron overload. No molecular diagnosis could be established in 14/43 cases. This study emphasizes the frequency of ALAS2 and SLC25A38 mutations and provides the largest comprehensive analysis to date of genotype/phenotype correlations in CSA. Further studies of CSA patients with data recorded in an international registry would be helpful to improve patient management and establish standardized guidelines.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Anemia, Sideroblastic/genetics , Genetic Association Studies , Genetic Diseases, X-Linked/genetics , Mitochondrial Membrane Transport Proteins/genetics , Anemia, Sideroblastic/pathology , Child , Cohort Studies , Europe , Female , Genetic Diseases, X-Linked/pathology , Humans , Male , Mutation , Nucleotidyltransferases/genetics , Retrospective Studies
7.
Haematologica ; 104(8): 1554-1564, 2019 08.
Article in English | MEDLINE | ID: mdl-30655378

ABSTRACT

We describe the clinical, hematologic and genetic characteristics of a retrospective series of 126 subjects from 64 families with hereditary xerocytosis. Twelve patients from six families carried a KCNN4 mutation, five had the recurrent p.Arg352His mutation and one had a new deletion at the exon 7-intron 7 junction. Forty-nine families carried a PIEZO1 mutation, which was a known recurrent mutation in only one-third of the cases and private sequence variation in others; 12 new probably pathogenic missense mutations were identified. The two dominant features leading to diagnosis were hemolysis that persisted after splenectomy and hyperferritinemia, with an inconstant correlation with liver iron content assessed by magnetic resonance imaging. PIEZO1-hereditary xerocytosis was characterized by compensated hemolysis in most cases, perinatal edema of heterogeneous severity in more than 20% of families and a major risk of post-splenectomy thrombotic events, including a high frequency of portal thrombosis. In KCNN4-related disease, the main symptoms were more severe anemia, hemolysis and iron overload, with no clear sign of red cell dehydration; therefore, this disorder would be better described as a 'Gardos channelopathy'. These data on the largest series to date indicate that PIEZO1-hereditary xerocytosis and Gardos channelopathy are not the same disease although they share hemolysis, a high rate of iron overload and inefficient splenectomy. They demonstrate the high variability in clinical expression as well as genetic bases of PIEZO1-hereditary xerocytosis. These results will help to improve the diagnosis of hereditary xerocytosis and to provide recommendations on the clinical management in terms of splenectomy, iron overload and pregnancy follow-up.


Subject(s)
Anemia, Hemolytic, Congenital/genetics , Channelopathies/genetics , Hydrops Fetalis/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ion Channels/genetics , Anemia, Hemolytic, Congenital/complications , Anemia, Hemolytic, Congenital/surgery , Edema/etiology , Family , Female , Hemolysis , Humans , Hydrops Fetalis/surgery , Iron Overload , Male , Mutation , Mutation, Missense , Pregnancy , Retrospective Studies , Splenectomy/adverse effects , Thrombosis
10.
JAMA ; 321(3): 266-276, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30667500

ABSTRACT

Importance: In children with sickle cell anemia (SCA), high transcranial Doppler (TCD) velocities are associated with stroke risk, which is reduced by chronic transfusion. Whether matched sibling donor hematopoietic stem cell transplantation (MSD-HSCT) can reduce velocities in patients with SCA is unknown. Objective: To determine the association of MSD-HSCT with TCD velocities as a surrogate for the occurrence of ischemic stroke in children with SCA. Design, Setting, and Participants: Nonrandomized controlled intervention study conducted at 9 French centers. Patients with SCA were enrolled between December 2010 and June 2013, with 3-year follow-up ending in January 2017. Children with SCA were eligible if younger than 15 years, required chronic transfusions for persistently elevated TCD velocities, and had at least 1 sibling without SCA from the same 2 parents. Families agreed to HLA antigen typing and transplantation if a matched sibling donor was identified or to standard care in the absence of a matched sibling donor. Exposures: MSD-HSCT (n = 32), compared with standard care (n = 35) (transfusions for ≥1 year with potential switch to hydroxyurea thereafter), using propensity score matching. Main Outcomes and Measures: The primary outcome was the highest time-averaged mean of maximum velocities in 8 cerebral arteries, measured by TCD (TCD velocity) at 1 year. Twenty-five of 29 secondary outcomes were analyzed, including the highest TCD velocity at 3 years and normalization of velocities (<170 cm/s) and ferritin levels at 1 and 3 years. Results: Sixty-seven children with SCA (median age, 7.6 years; 35 girls [52%]) were enrolled (7 with stroke history). In the matched sample, highest TCD velocities at 1 year were significantly lower on average in the transplantation group (129.6 cm/s) vs the standard care group (170.4 cm/s; difference, -40.8 cm/s [95% CI, -62.9 to -18.6]; P < .001). Of the 25 analyzed secondary end points, 4 showed significant differences, including the highest TCD velocity at 3 years (112.4 cm/s in the transplantation group vs 156.7 cm/s in the standard care group; difference, -44.3 [95% CI, -71.9 to -21.1]; P = .001); normalization rate at 1 year (80.0% in the transplantation group vs 48.0% in the standard care group; difference, 32.0% [95% CI, 0.2% to 58.6%]; P = .045); and ferritin levels at 1 year (905 ng/mL in the transplantation group vs 2529 ng/mL in the standard care group; difference, -1624 [95% CI, -2370 to -879]; P < .001) and 3 years (382 ng/mL in the transplantation group vs 2170 ng/mL in the standard care group; difference, -1788 [95% CI, -2570 to -1006]; P < .001). Conclusions and Relevance: Among children with SCA requiring chronic transfusion because of persistently elevated TCD velocities, MSD-HSCT was significantly associated with lower TCD velocities at 1 year compared with standard care. Further research is warranted to assess the effects of MSD-HSCT on clinical outcomes and over longer follow-up. Trial Registration: ClinicalTrials.gov Identifier: NCT01340404.


Subject(s)
Anemia, Sickle Cell/therapy , Cerebrovascular Circulation/physiology , Hematopoietic Stem Cell Transplantation , Siblings , Ultrasonography, Doppler, Transcranial , Allografts , Anemia, Sickle Cell/diagnostic imaging , Anemia, Sickle Cell/physiopathology , Blood Flow Velocity , Child , Female , Ferritins/blood , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Propensity Score , Quality of Life , Transplantation Conditioning
11.
Clin Immunol ; 188: 52-57, 2018 03.
Article in English | MEDLINE | ID: mdl-29330115

ABSTRACT

Evans syndrome (ES) is defined by the combination of autoimmune hemolytic anemia and immune thrombocytopenia. Clinical presentation includes manifestations of immune dysregulation, found in primary immune deficiencies, autoimmune lymphoproliferative syndrome with FAS (ALPS-FAS), Cytotoxic T Lymphocyte Antigen-4 (CTLA-4) and Lipopolysaccharide-Responsive vesicle trafficking Beige-like and Anchor protein (LRBA) defects. We report the clinical history and genetic results of 18 children with ES after excluding ALPS-FAS. Thirteen had organomegaly, five lymphocytic infiltration of non-lymphoid organs, nine hypogammaglobulinemia and fifteen anomalies in lymphocyte phenotyping. Seven patients had genetic defects: three CTLA4 mutations (c.151C>T; c.109+1092_568-512del; c.110-2A>G) identified by Sanger sequencing and four revealed by Next Generation Sequencing: LRBA (c.2450+1C>T), STAT3 gain-of-function (c.2147C>T; c.2144C>T) and KRAS (c.37G>T). No feature emerged to distinguish patients with or without genetic diagnosis. Our data on pediatric-onset ES should prompt physicians to perform extensive screening for mutations in the growing pool of genes involved in primary immune deficiencies with autoimmunity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Anemia, Hemolytic, Autoimmune/genetics , CTLA-4 Antigen/genetics , Mutation , Thrombocytopenia/genetics , Anemia, Hemolytic, Autoimmune/diagnosis , Anemia, Hemolytic, Autoimmune/pathology , Child , Child, Preschool , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/pathology , Thrombocytopenia/diagnosis
12.
Haematologica ; 103(7): 1143-1149, 2018 07.
Article in English | MEDLINE | ID: mdl-29599204

ABSTRACT

In this retrospective study, we evaluate long-term complications in nearly all ß-thalassemia-major patients who successfully received allogeneic hematopoietic stem cell transplantation in France. Ninety-nine patients were analyzed with a median age of 5.9 years at transplantation. The median duration of clinical follow up was 12 years. All conditioning regimens were myeloablative, most were based on busulfan combined with cyclophosphamide, and more than 90% of patients underwent a transplant from a matched sibling donor. After transplantation, 11% of patients developed thyroid dysfunction, 5% diabetes, and 2% heart failure. Hypogonadism was present in 56% of females and 14% of males. Female patients who went on to normal puberty after transplant were significantly younger at transplantation than those who experienced delayed puberty (median age 2.5 vs 8.7 years). Fertility was preserved in 9 of 27 females aged 20 years or older and 2 other patients became pregnant following oocyte donation. In addition to patient's age and higher serum ferritin levels at transplantation, time elapsed since transplant was significantly associated with decreased height growth in multivariate analysis. Weight growth increased after transplantation particularly in females, 36% of adults being overweight at last evaluation. A comprehensive long-term monitoring, especially of endocrine late effects, is required after hematopoietic stem cell transplantation for thalassemia.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , beta-Thalassemia/complications , beta-Thalassemia/epidemiology , Adolescent , Adult , Child , Child, Preschool , Female , France/epidemiology , Graft vs Host Disease/epidemiology , Graft vs Host Disease/etiology , Health Surveys , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Postoperative Period , Prognosis , Retrospective Studies , Transplantation, Homologous/adverse effects , Treatment Outcome , Young Adult , beta-Thalassemia/therapy
13.
Blood ; 126(11): 1273-80, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26148990

ABSTRACT

The Gardos channel is a Ca(2+)-sensitive, intermediate conductance, potassium selective channel expressed in several tissues including erythrocytes and pancreas. In normal erythrocytes, it is involved in cell volume modification. Here, we report the identification of a dominantly inherited mutation in the Gardos channel in 2 unrelated families and its association with chronic hemolysis and dehydrated cells, also referred to as hereditary xerocytosis (HX). The affected individuals present chronic anemia that varies in severity. Their red cells exhibit a panel of various shape abnormalities such as elliptocytes, hemighosts, schizocytes, and very rare stomatocytic cells. The missense mutation concerns a highly conserved residue among species, located in the region interacting with Calmodulin and responsible for the channel opening and the K(+) efflux. Using 2-microelectrode experiments on Xenopus oocytes and patch-clamp electrophysiology on HEK293 cells, we demonstrated that the mutated channel exhibits a higher activity and a higher Ca(2+) sensitivity compared with the wild-type (WT) channel. The mutated channel remains sensitive to inhibition suggesting that treatment of this type of HX by a specific inhibitor of the Gardos channel could be considered. The identification of a KCNN4 mutation associated with chronic hemolysis constitutes the first report of a human disease caused by a defect of the Gardos channel.


Subject(s)
Anemia, Hemolytic, Congenital/genetics , Hydrops Fetalis/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Mutant Proteins/genetics , Mutation, Missense , Adult , Amino Acid Sequence , Anemia, Hemolytic, Congenital/blood , Animals , Child, Preschool , Erythrocytes, Abnormal/metabolism , Female , Genes, Dominant , HEK293 Cells , Humans , Hydrops Fetalis/blood , In Vitro Techniques , Infant , Infant, Newborn , Intermediate-Conductance Calcium-Activated Potassium Channels/blood , Intermediate-Conductance Calcium-Activated Potassium Channels/chemistry , Male , Models, Molecular , Molecular Sequence Data , Mutant Proteins/blood , Mutant Proteins/chemistry , Oocytes/metabolism , Osmotic Fragility , Patch-Clamp Techniques , Pedigree , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus laevis
15.
Adv Exp Med Biol ; 1013: 89-122, 2017.
Article in English | MEDLINE | ID: mdl-29127678

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) can cure single gene disorders such as thalassemia and sickle cell anemia (SCA). These non-malignant diseases have in common severe hemolytic anemia and high proliferative bone marrow, requiring frequent transfusions. The risk of rejection is high and graft-vs-host disease is not desirable. Important progress has been made in the management of these diseases, including leukocyte depletion of blood products, and chelation therapy, for both diseases, and erythrocytapheresis and hydroxycarbamide for SCA. However, morbidity and quality of life are still of concern. Results have also significantly improved for HSCT, with the reduction of rejection by using anti-thymocyte globulin (ATG), which also decreases the risk of chronic graft-vs-host disease. Current data show a more than 90% chance of cure with myeloablative conditioning in children with hemoglobinopathy and a geno-identical donor. Results are similar whether the cell source is cord blood or bone marrow. Because of the risk of conditioning-related infertility, ovarian and/or testis cryopreservation should be discussed. Non-myeloablative conditioning regimens have also been successfully developed in adults with SCA and organ dysfunction, making cure possible. These encouraging results should incite to perform HLA typing early in families with hemoglobinopathies, and to systematically propose sibling cord blood cryopreservation for those without geno-identical donor.


Subject(s)
Anemia, Sickle Cell/therapy , Hematopoietic Stem Cell Transplantation/methods , beta-Thalassemia/therapy , Adult , Antilymphocyte Serum/administration & dosage , Child , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility Testing , Humans , Immunosuppressive Agents/administration & dosage , Quality of Life , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome
16.
Blood ; 124(18): 2867-71, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25193871

ABSTRACT

Mutations in genes encoding proteins that are involved in mitochondrial heme synthesis, iron-sulfur cluster biogenesis, and mitochondrial protein synthesis have previously been implicated in the pathogenesis of the congenital sideroblastic anemias (CSAs). We recently described a syndromic form of CSA associated with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD). Here we demonstrate that SIFD is caused by biallelic mutations in TRNT1, the gene encoding the CCA-adding enzyme essential for maturation of both nuclear and mitochondrial transfer RNAs. Using budding yeast lacking the TRNT1 homolog, CCA1, we confirm that the patient-associated TRNT1 mutations result in partial loss of function of TRNT1 and lead to metabolic defects in both the mitochondria and cytosol, which can account for the phenotypic pleiotropy.


Subject(s)
Anemia, Sideroblastic/congenital , Anemia, Sideroblastic/genetics , Developmental Disabilities/complications , Fever/complications , Genetic Diseases, X-Linked/genetics , Immunologic Deficiency Syndromes/complications , Mutation/genetics , RNA Nucleotidyltransferases/genetics , Alleles , Anemia, Sideroblastic/complications , Anemia, Sideroblastic/enzymology , Developmental Disabilities/genetics , Fever/genetics , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/enzymology , HEK293 Cells , Humans , Immunologic Deficiency Syndromes/genetics
17.
Br J Haematol ; 171(4): 615-24, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26205481

ABSTRACT

The level of circulating platelet-, erythrocyte-, leucocyte- and endothelial-derived microparticles detected by high-sensitivity flow cytometry was investigated in 37 ß-thalassaemia major patients receiving a regular transfusion regimen. The phospholipid procoagulant potential of the circulating microparticles and the microparticle-dependent tissue factor activity were evaluated. A high level of circulating erythrocyte- and platelet-microparticles was found. In contrast, the number of endothelial microparticles was within the normal range. Platelet microparticles were significantly higher in splenectomized than in non-splenectomized patients, independent of platelet count (P < 0·001). Multivariate analysis indicated that phospholipid-dependent procoagulant activity was influenced by both splenectomy (P = 0·001) and platelet microparticle level (P < 0·001). Erythrocyte microparticles were not related to splenectomy, appear to be devoid of proper procoagulant activity and no relationship between their production and haemolysis, dyserythropoiesis or oxidative stress markers could be established. Intra-microparticle labelling with anti-HbF antibodies showed that they originate only partially (median of 28%) from thalassaemic erythropoiesis. In conclusion, when ß-thalassaemia major patients are intensively transfused, the procoagulant activity associated with thalassaemic erythrocyte microparticles is probably diluted by transfusions. In contrast, platelet microparticles, being both more elevated and more procoagulant, especially after splenectomy, may contribute to the residual thrombotic risk reported in splenectomized multi-transfused ß-thalassaemia major patients.


Subject(s)
Blood Platelets/physiology , Blood Transfusion , Cell-Derived Microparticles/physiology , Thrombophilia/blood , beta-Thalassemia/blood , Adolescent , Adult , Autoantibodies/blood , Autoantibodies/immunology , Blood Platelets/ultrastructure , Cell-Derived Microparticles/classification , Combined Modality Therapy , Diabetes Mellitus/etiology , Erythrocyte Membrane/ultrastructure , Female , Fetal Hemoglobin/immunology , Flow Cytometry , Humans , Hypogonadism/etiology , Iron/blood , Iron Overload/blood , Iron Overload/etiology , Male , Membrane Lipids/blood , Middle Aged , Oxidative Stress , Phosphatidylserines/blood , Risk , Splenectomy , Thrombophilia/etiology , Transfusion Reaction , Young Adult , beta-Thalassemia/complications , beta-Thalassemia/surgery , beta-Thalassemia/therapy
18.
Blood ; 122(1): 112-23, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23553769

ABSTRACT

Congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited disorders identified by pathological erythroid precursors with perinuclear mitochondrial iron deposition in bone marrow. An international collaborative group of physicians and laboratory scientists collated clinical information on cases of CSA lacking known causative mutations, identifying a clinical subgroup of CSA associated with B immunodeficiency, periodic fevers, and development delay. Twelve cases from 10 families were identified. Median age at presentation was 2 months. Anemia at diagnosis was sideroblastic, typically severe (median hemoglobin, 7.1 g/dL) and markedly microcytic (median mean corpuscular volume, 62.0 fL). Clinical course involved recurrent febrile illness and gastrointestinal disturbance, lacking an infective cause. Investigation revealed B-cell lymphopenia (CD19⁺ range, 0.016-0.22 × 109/L) and panhypogammaglobulinemia in most cases. Children displayed developmental delay alongside variable neurodegeneration, seizures, cerebellar abnormalities, sensorineural deafness, and other multisystem features. Most required regular blood transfusion, iron chelation, and intravenous immunoglobulin replacement. Median survival was 48 months, with 7 deaths caused by cardiac or multiorgan failure. One child underwent bone marrow transplantation aged 9 months, with apparent cure of the hematologic and immunologic manifestations. We describe and define a novel CSA and B-cell immunodeficiency syndrome with additional features resembling a mitochondrial cytopathy. The molecular etiology is under investigation.


Subject(s)
Anemia, Sideroblastic/diagnosis , B-Lymphocytes/immunology , Developmental Disabilities/diagnosis , Familial Mediterranean Fever/diagnosis , Immunologic Deficiency Syndromes/diagnosis , Anemia, Sideroblastic/blood , Anemia, Sideroblastic/genetics , Developmental Disabilities/blood , Developmental Disabilities/genetics , Familial Mediterranean Fever/blood , Familial Mediterranean Fever/genetics , Female , Hearing Loss, Sensorineural/blood , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Humans , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Infant , Infant, Newborn , Male , Nervous System Diseases/blood , Nervous System Diseases/diagnosis , Nervous System Diseases/genetics , Pedigree , Phenotype , Syndrome
20.
Haematologica ; 100(4): 452-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25480500

ABSTRACT

Clinical and hematologic characteristics of beta(ß)-thalassemia are determined by several factors resulting in a wide spectrum of severity. Phenotype modulators are: HBB mutations, HBA defects and fetal hemoglobin production modulators (HBG2:g.-158C>T polymorphism, HBS1L-MYB intergenic region and the BCL11A). We characterized 54 genetic variants at these five loci robustly associated with the amelioration of beta-thalassemia phenotype, to build a predictive score of severity using a representative cohort of 890 ß-thalassemic patients. Using Cox proportional hazard analysis on a training set, we assessed the effect of these loci on the age at which patient started regular transfusions, built a Thalassemia Severity Score, and validated it on a testing set. Discriminatory power of the model was high (C-index=0.705; R(2)=0.343) and the validation conducted on the testing set confirmed its predictive accuracy with transfusion-free survival probability (P<0.001) and with transfusion dependency status (Area Under the Receiver Operating Characteristic Curve=0.774; P<0.001). Finally, an automatized on-line calculation of the score was made available at http://tss.unica.it. Besides the accurate assessment of genetic predictors effect, the present results could be helpful in the management of patients, both as a predictive score for screening and a standardized scale of severity to overcome the major-intermedia dichotomy and support clinical decisions.


Subject(s)
Genetic Variation , beta-Globins/genetics , beta-Thalassemia/diagnosis , beta-Thalassemia/genetics , Blood Transfusion , DNA, Intergenic , Female , Genetic Association Studies , Genetic Loci , Humans , Kaplan-Meier Estimate , Male , Mutation , Phenotype , Polymorphism, Single Nucleotide , Prognosis , Retrospective Studies , Severity of Illness Index , beta-Thalassemia/mortality , beta-Thalassemia/therapy
SELECTION OF CITATIONS
SEARCH DETAIL