A Pilot Study of Epigenetic-Differentiation Therapy with Decitabine to Treat β-Thalassemia Intermedia

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 ◽  
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.


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.


1986 ◽  
Vol 6 (9) ◽  
pp. 3278-3282
Author(s):  
D P Carlson ◽  
J Ross

A base substitution in the 5'-flanking region of a human fetal globin gene is associated with abnormal fetal hemoglobin production. It also reduces by 5- to 10-fold in vitro transcription of the gene by RNA polymerase III. We discuss potential links between polymerase III transcription and abnormal hemoglobin production.


Blood ◽  
1994 ◽  
Vol 83 (3) ◽  
pp. 822-827 ◽  
Author(s):  
AJ Dimovski ◽  
V Divoky ◽  
AD Adekile ◽  
E Baysal ◽  
JB Wilson ◽  
...  

Abstract A novel deletion of approximately 27 kb with the 5′ breakpoint 1.5 to 2.2 kb upstream of the beta-globin gene, and the 3′ breakpoint approximately 24 kb downstream of the beta-globin gene, has been found in five members of two families from Southeast Asia (Vietnam and Cambodia). Six members of another family from China, previously reported from our laboratory, have also been shown to carry this deletion. The patients presented with mild hypochromia and microcytosis, a hemoglobin (Hb) A2 level of approximately 4.0%, and a markedly increased, heterocellularly distributed, Hb F level (14.0 to 26.0%). In vitro globin-chain synthesis showed a mild imbalance with appreciable gamma-chain compensation (alpha/beta + gamma ratio of 1.46). The 3′ end of this deletion includes the 3′HS-1, and we hypothesize that removal of this region results in the loss of its gamma-globin gene-silencing effect, which causes a markedly elevated Hb F level with a modest increase in Hb A2 levels, unlike the situation in other deletional beta zero-thalassemias. The possible influence of particular sequence variations in the locus control region 5′HS-2 and the G gamma promoter, present on the chromosome with this deletion, on the overall gamma-globin gene should also be considered.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1216-1216
Author(s):  
Antonello Mai ◽  
Silvio Massa ◽  
Antonella Di Noia ◽  
Katija Jelicic ◽  
Elena Alfani ◽  
...  

Abstract Post-natal pharmacological reactivation of HbF, by restoring the unbalanced α/non-α globin chain production in red cells of patients affected by β-thalassemia or sickle cell anemia, represents a potential cure for these diseases. Many classes of compounds have been identified capable to induce Hb F synthesis in vitro by acting at different levels of the globin gene expression regulatory machinery. One of these classes is represented by inhibitors of a family of enzymes, the histone deacetylases (HDACs), involved in chromatin remodelling and gene transcription regulation. HDACs act in multi-protein complexes that remove acetyl groups from lysine residues on several proteins, including histones and are divided into three distinct structural classes, depending on whether their catalytic activity is zinc (class I/II)- or NAD+ (class III)-dependent. The effects of the HDACs inhibitors identified so far on HbF synthesis is, however, modest and often associated with high toxicity. Therefore, the potential of their clinical use is unclear. We have recently described a new family of synthetic HDACs inhibitors, the Aroyl-pyrrolyl-hydroxy-amides (APHAs), that induce differentiation, growth arrest and/or apoptosis of transformed cell in culture [Mai A et al, J Med Chem2004;47:1098]. In this study, we investigate the capability of 10 different APHA compounds to induce Hb F in two in vitro assays. One assay is based on the ability of APHA compounds to activate either the human Aγ-driven Firefly (Aγ-F) or the β-promoter drives Renilla Luciferase (β-R) reporter in GM979 cells stably transfected with a Dual Luciferase Reporter construct. The second assay is represented by the induction of γ-globin expression (by quantitative RT-PCR) in primary adult erythroblasts obtained in HEMA cultures of mononuclear cells from normal donors. The majority of the compounds tested did not significantly increased the Aγ−F (Aγ−F+β−R) reporter ratio in GM979 cells. However, the compound MC1575 increased by 3-fold (from 0.09 to 0.30) the reporter ratio in GM979 cells at a concentration of 20 μM, with modest effects of the proliferation activity of GM979 cells over the three days of the assay. When MC1575 was added at a concentration of 2–10 μM in cultures of primary adult erythroblasts induced to differentiate in serum-free media for 4 days, it induced a three fold increase of the γ/(γ+β) globin ratio (from 0.04 to 0.12), with no apparent cellular toxicity. Among the HDAC inhibitors tested in this study, MC1575 was not the most potent inhibitor of total enzyme activity. However, it was the compound that most selectively inhibited the activity of the maize homologue of mammalian class IIa HDAC enzymes [Mai et al, J Med Chem2003;46:4826]. These results are consistent with the hypothesis that each class of histone deacetylases might have a specific biological function and indicate that those of class IIa might represent the enzymes most specifically involved in globin gene regulation. We suggest that, by targeting the chemical inhibitors toward the catalytic domain of this class of enzymes, it should be possible to identify more specific, more potent and less toxic compounds for pharmacological treatment of β-thalassemia or sickle cell anemia.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4747-4747 ◽  
Author(s):  
Hans Hasselbalch

Abstract Background. Imatinib targets the ATP-binding sites of the protein tyrosine kinase domains associated with Bcr-abl, platelet-derived growth factor receptors (PDGFR) and c-kit. Most recently imatinib has been to inhibit autonomous erythropoiesis in vitro in polycythemia vera (PV)(1). Several clinical studies have indicated that imatinib may reduce phlebotomy requirements (2,3), but only a few patients have been followed for longer periods on imatinib monotherapy (2,3). Aim of the study. To evaluate the safety and efficacy of long term monotherapy with imatinib in PV. Patients.Eight patients (median age 60 years, range 39–68) have been treated. Five of the patients were enrolled in a phase II treatment protocol and three patients have been treated off protocol. None of the patients in the protocol had received cytotoxic treatment whereas the three patients treated off protocol had been treated with hydroxyurea (HU) and PEG-Intron. Imatinib was given at an initial dose of 400 mg/day in all patients. Methods. The diagnosis of PV was made according to conventional criteria, including an increased red cell blood volume and a low plasma erythropoietin. All patients were negative for the Bcr-abl transcript. A complete response (CR) was defined as phlebotomy-free within the first year of treatment, lasting at least 6 mo together with a platelet count less than 600 x 109/l and absence of splenomegaly. A partial response (PR) was defined by phlebotomy-free within the first year of treatment, lasting at least 6 mo together with a platelet count more than 600 x 109/l and a palpable spleen but less than 50 % of the original size (2). A minor response (MR) was defined as a reduction in the need of phlebotomies of at least 50 % but no change in the platelet counts. Results. Four patients have been followed for 12 mo on a dose of 400 mg daily apart from short periods, when the dose had to be decreased to 100 mg/day and 300mg/day, respectively, due to temporary side effects. Responses were seen in all evaluable patients (7/7) (CR=1; PR=6). The PR’s in two of the patients were obtained when HU (n=1) or PEG-Intron (n=1) were added. A definite decline in the Ht within the first month (< 0.45) and a reduction in the need of phlebotomies were recorded in all evaluable patients whereas the leucocyte and platelet counts displayed a highly heterogeneous response pattern with unchanged and even rising platelet counts in 3 patients. The one patient with a CR displayed a reduction of spleen size and a decrease in the degree of bone marrow fibrosis after being treated for 12 mo. Almost complete alleviation of severe pruritus was noticed in one patient a few weeks after starting imatinib. One patient had to discontinue treatment after 1 week due to severe musculoskeletal pain. Otherwise the side effects were moderate and often transient. One of the patients noticed no side effects at all. Conclusions. Imatinib in PV is followed by a decrease in the Ht but highly heterogeneous leucocyte-and platelet responses in some patients when using 400 mg/day.Combinational therapy with HU or PEG-Intron seems safe and effective.


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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2022-2022
Author(s):  
Hua Cao ◽  
Rui Gao Fei ◽  
Albert A. Bowers ◽  
Thomas J. Greshock ◽  
Tenaya Newkirt ◽  
...  

Abstract Abstract 2022 Poster Board I-1044 Previous studies have demonstrated that Histone Deacetylase (HDAC) inhibitors such as butyrate and several short chain fatty acids, can induce fetal hemoglobin in humans and animal models; however induction of Hb F is achieved in relatively high concentrations of these compounds. We have previously investigated the induction of human γ globin gene activity by the prototypical HDAC inhibitor, FK228. The results demonstrated that FK228 is a more potent γ globin gene inducer compared to other HDAC inhibitors we have tested before (Am J Hematol. 12:981). In this study, we investigated the induction of human γ globin gene function of largazole and it's thiol analogue in vitro in cultures of normal human adult BFUe and in vivo in the mice carrying a human γ globin transgene. Largazole is a HDAC inhibitor which was recently isolated from a marine vyanobacterium by Luesch and co-workers. Structural features of largazole, a macrocyclic depsopeptide, closely resemble those of FK228, FR901375 and spiruchostatin. We have reported that largazole and numerous synthetic analogues are highly potent Class I histone deacetylase inhibitors (J Am Chem Soc. 130:11219, J Am Chem Soc. 2009 Feb 4). We used flow cytometry to measure the in vitro effect of largazole and it's derivatives on the frequency of HbF-positive erythroblasts in BFUe cultures from normal individuals; real-time quantitative PCR (RT-qPCR) and high performance liquid chromatography (HPLC) were used to measure the in vivo effects of largazole on human γ globin induction in γ transgenic mice carrying a human γ globin gene.. Our results show that largazole and it's thiol derivative are potent γ hemoglobin gene inducers. In the human BFUe cultures, largazole increased the levels of fetal hemoglobin positive cells from 21.9% (control level) to 62.8% at a concentration of 0.1μM; largazole thiol increased the levels of fetal hemoglobin positive cells to 62.0% at a concentration of 1μM. Transgenic mice carrying the human μLCR Aγ construct continue to express the human γ gene in the adult stage (Blood. 77:1326). Largazole was administered through IP injection at the dosages of 0.3mg/kg/day and 0.6mg/kg/day, 5 days per week, for 2 weeks to two cohorts of transgenic mice. Largazole at the dose of 0.3mg/kg/day increased the level of human γ mRNA at the end of injection by 160.7%; at a dose of 0.6mg/kg/day human γ mRNA increased by 174.7%. At the 0.6mg/kg/day dosage the level of fetal hemoglobin in the peripheral blood of the animals increased by 3.4 and 3.2 fold at day 21 and day 28, respectively. These results provide strong in vitro and in vivo evidence that Largazole and it's thiol analogue are potent HbF inducers acting at low concentrations, and thus provide promising alternatives to compounds currently considered for induction of Hb F in patients with sickle cell disease and thalassemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4427-4427
Author(s):  
Matthew J. Olnes ◽  
Yong Tang ◽  
Susan Soto ◽  
Elaine M Sloand ◽  
Philip Scheinberg ◽  
...  

Abstract Abstract 4427 Severe aplastic anemia (SAA) is characterized by trilineage marrow hypoplasia and a paucity of hematopoietic stem cell progenitors. SAA is treated with immunosuppression or allogeneic stem cell transplantation (SCT), with a successful outcome in a majority. However, 20–40% of patients without a suitable donor for SCT do not respond to immunosuppression and may have persistent severe thrombocytopenia. Thrombopoietin (TPO) is the principal regulator of platelet production, and it exerts its effects through binding the megakaryocyte progenitor TPO receptor mpl, which stimulates production of mature megakaryocytes and platelets. Eltrombopag, a small molecule TPO mimetic that binds to mpl, increases platelet counts in healthy subjects, and in patients with chronic immune thrombocytopenic purpura. Both TPO and eltrombopag stimulate more primitive multilineage progenitors and stem cells in vitro. Patients with SAA and thrombocytopenia have very elevated TPO levels; nevertheless, we asked whether pharmacologic doses of eltrombopag could stimulate hematopoiesis in these patients without other options. We are conducting a pilot phase II study of eltrombopag in SAA patients with severe thrombocytopenia refractory to immunosuppressive therapy. Consecutive eligible adult patients were treated with oral eltrombopag at an initial dose of 50 mg daily, with escalation to a maximum dose 150 mg daily, with the goal of maintaining a platelet count of >20,000/uL above baseline. Treatment response was measured after three months and was defined as platelet count increases to 20,000/uL above baseline, or stable platelet counts with transfusion-independence for a minimum of 8 weeks. Nine patients have been enrolled and six are evaluable for response to date. Two patients did not respond to treatment. Three patients achieved platelet responses by 12 weeks of treatment, and all have sustained their responses (median follow up 10 months). Four patients exhibited improved hemoglobin levels 12 weeks after starting treatment (median hemoglobin increase of 2.1 g/dL) and two patients who were previously dependent on packed red blood cell transfusions have achieved transfusion-independence. Three neutropenic patients exhibited increased neutrophil counts after treatment with eltrombopag (median increase 0.46K cells/uL). These results provide evidence that eltrombopag can improve platelet counts in patients with severe refractory thrombocytopenia, and perhaps more surprisingly, have a clinically relevant impact on erythropoiesis and myelopoiesis. Updated data will be presented at the Society's meeting. Disclosures: Off Label Use: Eltrombopag for thrombocytopenia in refractory severe aplastic anemia patients.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 753-753 ◽  
Author(s):  
Raffaele Renella ◽  
Aleksej Perlov ◽  
Chad E Harris ◽  
Daniel E. Bauer ◽  
Jian Xu ◽  
...  

Abstract Abstract 753 Sickle cell disease (SCD) is caused by a mutation in the β-globin protein, leading to the polymerization of hemoglobin in deoxygenated conditions. The transcription factor BCL11A is a key regulator of developmental silencing of human fetal (γ-) globin, and also critical to repressing γ-globin in adult erythroid cells. A Bcl11a null mouse model carrying a transgenic YAC with a humanized β-globin locus (β-YAC) displays increased levels of fetal hemoglobin (HbF) in adult erythrocytes, and crossing these animals with a SCD murine model abolishes the SCD phenotype. BCL11A therefore constitutes a genetically validated target to induce HbF and reduce erythrocyte “sickling”, which would be predicted to ameliorate the phenotype of SCD patients. However, defective lymphoid development has been observed in Bcl11a genetic null mice, suggesting potential toxicities of BCL11A knockdown. We generated self-inactivating lentiviral vectors (LV) integrating miR-223 microRNA-based inhibitory shRNAs against BCL11A/Bcl11a. Since future clinical applications will need to balance efficacy and potential side effects, LVs were engineered to allow the comparison of effects of high level and ubiquitous versus erythroid lineage-restricted versus inducible expression of miRNA targeting BCL11A. LV backbones therefore included either a strong, viral LTR promoter/enhancer (SFFV-LV), a β-globin locus control region with the endogenous β-globin promoter (LCR-LV), or a tetracycline-inducible promoter (TET-LV). We performed assays to quantify transgenic miRNA expression and demonstrated that the BCL11A knockdown and induction of fetal globin gene output correlated with the expression of targeting miR223-based shRNA. Transduction at low MOI (=2) of murine hematopoietic stem cells (HSC) with LVs carrying the abovementioned regulatory elements leads to long-term engraftment and transgene expression in-vivo. Mice transplanted with SFFV-LV show fluorescent marking up to 70% across myeloid, lymphoid and erythroid lineages. The maximal BCL11A/Bcl11a mRNA and protein knock-down observed in primary hematopoietic cells in-vitro and in-vivo was 70%. This was confirmed in FACS-sorted bone marrow B-lymphoid (B220+) and erythroid progenitors (Terr119+/CD71+) and peripheral blood leukocytes at 4 months post-transplant. BCL11A/Bcl11a knockdown induced fetal globin gene expression depending on the vector backbone and targeting shRNA sequence employed. With SFFV-LV, we observed a 5–20 fold upregulation of fetal globin gene (γ/(ϵ+γ+β)) output in mice transplanted with HSCs containing the humanized β-YAC transgene. With TET-LV, the induction was dose-dependent and maximally caused a 150-fold increase in murine ϵγ-globin gene expression in-vitro. Human HSC transduced (MOI=2) with the LCR-LV and differentiated in-vitro resulted in a 3-fold increase of γ-globin mRNA in erythrocytes. SCD patient-derived HSC, which were transduced with LCR-LV (MOI=5) and transplanted into immunodeficient NSG mice, resulted in peripheral human erythrocytes that showed a reversal of the hemoglobin switch with a maximal induction 10% HbF as measured by flow cytometry. In a human ex-vivo B-lymphoid differentiation assay, SFFV-LV transduced (MOI=2) HSC populations with a 70% BCL11A knock-down showed no difference versus control in total cell numbers or in the sequential acquisition of CD43, CD19 and IgM (corresponding to physiological differentiation from common lymphoid progenitor to immature B-lymphocyte), thus showing no evidence for a differentiation block. Since IFN-response gene activation has been described with shRNA silencing and could potentially lead to HSC exhaustion, we quantified ISG20, ISG56 and OAS1 mRNA levels in human HSCs after miR-223-based SFFV-LV transduction (MOI=2). We observed less IFN-response gene activation in miR223-based SFFV-LV transduced HSC than in non-miRNA-based shRNA SFFV-LV transduced controls. In summary, our pre-clinical data demonstrates the potential efficacy of hematopoietic miRNA-mediated BCL11A/Bcl11a silencing to induce the expression of fetal hemoglobin in murine and human model systems, including primary cells. At the levels of BCL11A knock-down obtained, we did not observe any B-lymphoid toxicity. These results support the translation of LV-based miRNA-mediated BCL11A silencing into the clinical setting. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document