scholarly journals A pilot study of subcutaneous decitabine in β-thalassemia intermedia

Blood ◽  
2011 ◽  
Vol 118 (10) ◽  
pp. 2708-2711 ◽  
Author(s):  
Nancy F. Olivieri ◽  
Yogen Saunthararajah ◽  
Vivek Thayalasuthan ◽  
Janet Kwiatkowski ◽  
Russell E. Ware ◽  
...  

Abstract Ineffective erythropoiesis, the hallmark of β-thalassemia, is a result of α/non-α globin chain imbalance.1 One strategy to redress globin-chain imbalance is to induce γ-globin gene (HBG) expression. Repression of HBG in adult erythroid cells involves DNA methylation and other epigenetic changes. Therefore, the cytosine analog decitabine, which can deplete DNA methyltransferase 1 (DNMT1), can potentially activate HBG. In 5 patients with β-thalassemia intermedia, a dose and schedule of decitabine intended to deplete DNMT1 without causing significant cytotoxicity (0.2 mg/kg subcutaneous 2 times per week for 12 weeks) increased total hemoglobin from 7.88 ± 0.88 g/dL to 9.04 ± 0.77 g/dL (P = .004) and absolute fetal hemoglobin from 3.64 ± 1.13 g/dL to 4.29 ± 1.13 g/dL (P = .003). Significant favorable changes also occurred in indices of hemolysis and red blood cell densitometry. Consistent with a noncytotoxic, differentiation altering mechanism of action, the major side effect was an asymptomatic increase in platelet counts without erythrocyte micronucleus or VDJ recombination assay evidence of genotoxicity. This study was registered at www.clinicaltrials.gov as #NCT00661726.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2078-2078
Author(s):  
Nancy F Olivieri ◽  
Yogenthiran Saunthararajah ◽  
Vivekanandan Thayalasuthan ◽  
Janet Kwiatkowski ◽  
Russell E. Ware ◽  
...  

Abstract Abstract 2078 Ineffective erythropoiesis, the hallmark of ß-thalassemia, is a result of the myriad deleterious effects of globin chain imbalance. A major translational research goal in thalassemia is to restore α/non-α globin chain balance by inducing expression of γ-globin synthesis from the intact γ-globin gene (HBG). Repression of HBG in adult erythroid cells involves DNA methylation and other epigenetic changes; one possibly useful strategy to re-induce HBG expression is to deplete DNA methyl-transferase 1 (DNMT1) in hematopoietic cells using the cytosine analogue decitabine. A dose and schedule of decitabine intended to deplete DNMT1 without causing significant cytotoxicity was examined in a pilot safety study in ß-thalassemia intermedia, a condition which, despite a lack of requirement for monthly transfusions, is often associated with significant long-term clinical complications. Six patients (≥18 years of age) with ß-thalassemia intermedia were enrolled on study to receive decitabine 0.2 mg/kg subcutaneous 2x/week on consecutive days each week for 12 weeks followed by 12 weeks of follow-up. One patient withdrew from study because of fatigue requiring transfusion after week 2. Of the five evaluable patients, two patients received 24 of the 24 planned doses of drug, one patient received 21 of 24 planned doses, and two patients received 16 of 24 planned doses. Doses were missed because of treatment-associated increases in platelet count to >1000 × 109/L which according to protocol, required interruption of therapy. The primary outcome, an increase in total hemoglobin (Hb) of ≥1.5 g/dL above that determined at baseline, was achieved in two of five evaluable patients. In the group overall, Hb increased from a baseline of (mean ± SEM) 7.88 ± 0.88 g/dL to a peak of 9.04 ± 0.77 g/dL (P = 0.004); peak values in Hb were observed from the 6th to 12th week of treatment. Absolute fetal Hb increased from a baseline of (mean ± SEM) 3.34 ± 0.97 g/dL to a peak of 4.39 ± 1.15 g/dL (P = 0.021); peak values in fetal Hb were observed in the 4th to 12th week of treatment. Reflecting decreased hemolysis and more effective erythropoiesis, indirect bilirubin declined from (mean ± SEM) 3.2 ± 1.0 mg/dL to a nadir of 2.2± 0.8 mg/dL, while reduction in serum LDH from (mean ± SEM) 479.4 ± 125.8 U/L to 362.8 ± 100.4 U/L was not significant (P =0.083). Platelet counts increased from a baseline of (mean ± SEM) 585.2 ± 90.6 (x109/L) to a peak of 940.2 ± 184.3 (x109/L) (P = 0.007); peak platelet values were observed between the 6th to 12th weeks of treatment. These increases triggered interruption of therapy in three of the five evaluable patients. In the only patient not previously splenectomized, minimal change in platelet count was observed (baseline, 233 ×109/L; peak, 296 ×109/L). No clinical events were associated with the increased platelet counts. Changes in neutrophil count, from (mean ± SEM) 6.51 ± 1.20 (x109/L) to 3.36 ± 0.63 (x109/L) were not significant (P = 0.069). The lowest neutrophil counts were observed between the 4th and 10th week of treatment. Two quantitative in vitro assays for mutagenicity, specifically enumeration of illegitimate VDJ recombination events and micronuclei within early reticulocytes, were performed at baseline, midpoint, and study exit. No significant changes were identified between baseline and 24-week values from either laboratory assay. In this first clinical study of decitabine in patients with β-thalassemia, drug dose was aimed at induction of DNMT1 depletion; the frequent but intermittent schedule of administration was intended to modify differentiation without causing prolonged cytostasis resulting in cytopenia. Consistent with a non-cytotoxic/cytostatic, differentiation-altering mechanism of action, the dose-limiting toxicity observed was not thrombocytopenia or neutropenia, but asymptomatic increases in platelet count. In conclusion, decitabine therapy was well-tolerated in thalassemia intermedia. Significant mean and individual increases in total and fetal hemoglobin concentrations observed in this study may direct the selection of patients in extended trials, to explore the potential of chromatin-relaxing therapy for β-thalassemia. Disclosures: Off Label Use: FDA approved decitabine for use in MDS, therefore, its use in thalassemia is off-label. Furthermore, the dose, schedule and route of administration used is also not on the label. Decitabine was used in a study to augment production of fetal hemoglobin in an NIH sponsored trial.


Blood ◽  
2020 ◽  
Author(s):  
Yi Gong ◽  
Xinhua Zhang ◽  
Qianqian Zhang ◽  
Yanxia Zhang ◽  
Yuhua Ye ◽  
...  

DNA methyltransferase 1 (DNMT1) is a major epigenetic regulator of the formation of large macromolecular complexes that repress human γ-globin expression by maintaining DNA methylation. However, very little is known about the association of DNMT1 variants with β-thalassemia phenotypes. We systematically investigated associations between variants in DNMT1 and phenotypes in 1,142 β-thalassemia subjects and identified a novel missense mutation (c.2633G>A, S878F) in the bromo-adjacent homology-1 (BAH1) domain of DNMT1. We functionally characterized this mutation in CD34+ cells from patientsand engineered HuDEP-2 mutant cells. Our results demonstrate that DNMT1 phosphorylation is abrogated by substitution of serine with phenylalanine at position 878, resulting in lower stability and loss of catalytic activity. S878F mutation also attenuated the interactions of DNMT1 with BCL11A, GATA1 and HDAC1/2 and reduced the recruitment of DNMT1 to the HBG promoters, leading to epigenetic de-repression of γ-globin expression. By analyzing the F-cell pattern, we demonstrated that the effect of DNMT1 mutation on increased fetal hemoglobin (HbF) is heterocellular. Furthermore, introduction of S878F mutation into erythroid cells by CRISPR-Cas9 recapitulated γ-globin reactivation. Thus, the natural S878F DNMT1 mutation is a novel modulator of HbF synthesis and represents a potential new therapeutic target for β-hemoglobinopathies.


2003 ◽  
Vol 121 (1) ◽  
pp. 28-30
Author(s):  
Sylvia Morais de Sousa ◽  
Letícia Khater ◽  
Luís Antônio Peroni ◽  
Karine Miranda ◽  
Marcelo Jun Murai ◽  
...  

CONTEXT: We verified molecular alterations in a 72-year-old Brazilian male patient with a clinical course of homozygous beta-thalassemia intermedia, who had undergone splenectomy and was surviving without regular blood transfusions. The blood cell count revealed microcytic and hypochromic anemia (hemoglobin = 6.5 g/dl, mean cell volume = 74 fl, mean cell hemoglobin = 24 pg) and hemoglobin electrophoresis showed fetal hemoglobin = 1.3%, hemoglobin A2 = 6.78% and hemoglobin A = 79.4%. OBJECTIVE: To identify mutations in a patient with the symptoms of beta-thalassemia intermedia. DESIGN: Molecular inquiry into the mutations possibly responsible for the clinical picture described. SETTING: The structural molecular biology and genetic engineering center of the Universidade Estadual de Campinas, Campinas, Brazil. PROCEDURES: DNA extraction was performed on the patient's blood samples. The polymerase chain reaction (PCR) was done using five specific primers that amplified exons and the promoter region of the beta globin gene. The samples were sequenced and then analyzed via computer programs. RESULTS: Two mutations that cause the disease were found: -101 (C > T) and codon 39 (C > T). CONCLUSIONS: This case represents the first description of 101 (C > T) mutation in a Brazilian population and it is associated with a benign clinical course.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 411-414 ◽  
Author(s):  
MC Stevens ◽  
GH Maude ◽  
M Beckford ◽  
Y Grandison ◽  
K Mason ◽  
...  

alpha Thalassemia modifies the hematologic expression of homozygous sickle cell (SS) disease, resulting in increased total hemoglobin and HbA2 and decreased HbF, mean cell volume, reticulocytes, irreversibly sickled cells, and bilirubin levels. The age at which these changes develop in children with SS disease is unknown. Ascertainment of globin gene status in a large representative sample of children with SS disease has afforded an opportunity to study the hematologic indices in nine children homozygous for alpha thalassemia 2 (two-gene group), 90 children heterozygous for alpha thalassemia 2 (three-gene group), and 167 children with a normal alpha globin gene complement (four-gene group). The two-gene group had significantly lower mean cell volumes from birth, higher red cell counts from one month, lower reticulocytes from three months, and higher HbA2 levels from one year, as compared with the four-gene group. Children with three genes had intermediate indices but resembled more closely the four-gene group. Differences in total hemoglobin or in fetal hemoglobin between the groups were not apparent by eight years of age. The most characteristic differences of the two-gene group were the raised proportional HbA2 level and low mean cell volume, the latter having some predictive value for alpha thalassemia status at birth.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 411-414 ◽  
Author(s):  
MC Stevens ◽  
GH Maude ◽  
M Beckford ◽  
Y Grandison ◽  
K Mason ◽  
...  

Abstract alpha Thalassemia modifies the hematologic expression of homozygous sickle cell (SS) disease, resulting in increased total hemoglobin and HbA2 and decreased HbF, mean cell volume, reticulocytes, irreversibly sickled cells, and bilirubin levels. The age at which these changes develop in children with SS disease is unknown. Ascertainment of globin gene status in a large representative sample of children with SS disease has afforded an opportunity to study the hematologic indices in nine children homozygous for alpha thalassemia 2 (two-gene group), 90 children heterozygous for alpha thalassemia 2 (three-gene group), and 167 children with a normal alpha globin gene complement (four-gene group). The two-gene group had significantly lower mean cell volumes from birth, higher red cell counts from one month, lower reticulocytes from three months, and higher HbA2 levels from one year, as compared with the four-gene group. Children with three genes had intermediate indices but resembled more closely the four-gene group. Differences in total hemoglobin or in fetal hemoglobin between the groups were not apparent by eight years of age. The most characteristic differences of the two-gene group were the raised proportional HbA2 level and low mean cell volume, the latter having some predictive value for alpha thalassemia status at birth.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 555-555 ◽  
Author(s):  
Hassana Fathallah ◽  
Ali Taher ◽  
Ali Bazarbachi ◽  
George F. Atweh

Abstract A number of therapeutic agents including hydroxyurea, butyrate and decitabine have shown considerable promise in the treatment of sickle cell disease (SCD). However, the same agents have shown less clinical activity in β-thalassemia. As a first step towards understanding the molecular basis of the different clinical responses to these agents, we have studied the mechanisms of induction of fetal hemoglobin (HbF) by butyrate in BFU-E derived cells from 5 patients with SCD and 9 patients with β-thalassemia intermedia. Exposure to butyrate resulted in a dose-dependent augmentation of γ-globin mRNA levels in erythroid cells from patients with SCD. In contrast, induction of γ-globin expression in erythroid cells from patients with β-thalassemia intermedia was only seen at a high concentration of butyrate. The increase in γ-globin mRNA levels in patients with SCD and β-thalassemia intermedia was associated with opening of the DNA structure as manifested by decreased DNA methylation at the γ-globin promoters. Interestingly, butyrate exposure had markedly different effects on the expression of the β- and α-globin genes in the two categories of patients. Butyrate decreased the level of β-globin mRNA in 4 out of 5 patients with SCD (P = 0.04), while in β-thalassemia the levels of β-globin mRNA did not change in 7 patients and decreased in 2 patients after butyrate exposure (P = 0.12). Thus in patients with SCD, the effects of the induction of the γ-globin gene on the γ/(β+γ) mRNA ratios were further enhanced by the butyrate-mediated decreased expression of the β-globin gene. As a result, γ/(β+γ) mRNA ratios increased in all patients with SCD, with a mean increase of 31% (P = 0.002). In contrast, butyrate increased γ/(β+γ) mRNA ratios only in 4 out of 9 patients with β-thalassemia, with a more modest mean increase of 12% (P = 0.004). Interestingly, the decreased β-globin expression in patients with SCD was associated with closing of the DNA configuration as manifested by hypermethylation of DNA at the promoter of the β-globin gene while methylation of the same promoter did not change following butyrate exposure in patients with β-thalassemia intermedia. More surprisingly, the expression of the α-globin genes increased following butyrate exposure in 4 out of 9 patients with β-thalassemia, while the levels of α-globin mRNA decreased in 4 out of 5 patients with SCD. As a result, the favorable effects of the butyrate-induced increase in γ-globin gene expression on the α: non-α mRNA imbalance in patients with β-thalassemia intermedia were partly neutralized by the corresponding increase in α-globin gene expression. These differences may explain, at least in part, the more favorable effects of inducers of HbF in SCD than in β-thalassemia. Further studies are necessary to fully understand the molecular bases of the different responses to agents that induce HbF in patients with these disorders.


Blood ◽  
2012 ◽  
Vol 119 (2) ◽  
pp. 364-367 ◽  
Author(s):  
Khaled M. Musallam ◽  
Vijay G. Sankaran ◽  
Maria Domenica Cappellini ◽  
Lorena Duca ◽  
David G. Nathan ◽  
...  

To evaluate the association between fetal hemoglobin (HbF) levels and morbidity in β-thalassemia intermedia (TI), we analyzed data from 63 untransfused patients who had also never received HbF induction therapy. Patient records were reviewed for any history of 10 predefined morbidities. Laboratory measurements for markers of ineffective erythropoiesis were also obtained. The mean age of patients was 32.1 years, 47.6% were males, and the median HbF level was 37.2%. HbF levels correlated positively with total hemoglobin, yet negatively with growth differentiation factor-15 and non–transferrin-bound iron levels. Median HbF levels were significantly lower in patients with the majority of evaluated morbidities than in those without. There was a strong negative adjusted linear correlation between the HbF level and the total number of morbidities (R2 = 0.825, P < .001). The HbF threshold of 63.7% had 95.5% sensitivity and 100% specificity for ensuring absence of morbidity. There exists a strong association between HbF levels and morbidity in the subset of untransfused patients with TI.


2020 ◽  
Vol 9 (11) ◽  
pp. 3782
Author(s):  
Martin H. Steinberg

Fetal hemoglobin (HbF) usually consists of 4 to 10% of total hemoglobin in adults of African descent with sickle cell anemia. Rarely, their HbF levels reach more than 30%. High HbF levels are sometimes a result of β-globin gene deletions or point mutations in the promoters of the HbF genes. Collectively, the phenotype caused by these mutations is called hereditary persistence of fetal hemoglobin, or HPFH. The pancellularity of HbF associated with these mutations inhibits sickle hemoglobin polymerization in most sickle erythrocytes so that these patients usually have inconsequential hemolysis and few, if any, vasoocclusive complications. Unusually high HbF can also be associated with variants of the major repressors of the HbF genes, BCL11A and MYB. Perhaps most often, we lack an explanation for very high HbF levels in sickle cell anemia.


Blood ◽  
1991 ◽  
Vol 77 (2) ◽  
pp. 371-375 ◽  
Author(s):  
A Podda ◽  
R Galanello ◽  
L Maccioni ◽  
MA Melis ◽  
C Rosatelli ◽  
...  

Abstract This report describes a patient with thalassemia intermedia-like phenotype born to normal parents in whom globin gene sequencing detected a novel abnormal hemoglobin (Hb) due to a T to A substitution at codon 60 of the beta-globin gene arising as a de novo mutation. Normal sequences were detected at the homologous beta-globin locus. This mutation results in the substitution of a polar (glutamic acid) for a nonpolar (valine) residue near the corner of the heme pocket of the beta-globin chain. The novel variant has been designated Hb Cagliari, from the place of birth of the propositus. Kinetics of globin synthesis performed following splenectomy suggest that this new Hb variant is synthesized at a near normal rate but undergoes rapid breakdown. The extreme lability of the variant explains the clinical and hematologic picture characterized by marked ineffective erythropoiesis, thalassemia-like bone changes, iron overload, high proportion of Hb F in the peripheral blood, reduced beta/alpha-globin chain synthesis ratio in peripheral blood reticulocytes, and absence of the abnormal Hb in peripheral blood at extensive protein structural analysis before splenectomy. This case indicates that a thalassemic hemoglobinopathy should be suspected in the presence of a patient with a thalassemia intermedia-like phenotype born to normal parents, even when protein structural analysis fails to detect an abnormal Hb. DNA sequencing may allow to define the mutation, thus making the proper diagnosis.


Sign in / Sign up

Export Citation Format

Share Document