scholarly journals Genome Editing Strategies to Treat Beta-Hemoglobinopathies

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-16-SCI-16
Author(s):  
Mitchell J Weiss

Genetic forms of anemia caused by HBB gene mutations that impair beta globin production are extremely common worldwide. The resultant disorders, mainly sickle cell disease (SCD) and beta-thalassemia, cause substantial morbidity and early mortality. Treatments for these diseases include medical therapies and bone marrow transplantation (BMT), which can be curative. However, medical therapies are suboptimal and BMT is associated with serious toxicities, particularly because HLA-matched allogeneic sibling donors are not available for most patients. Thus, new therapies are urgently needed for millions of affected individuals. Gene therapy offers great promise to cure SCD and beta thalassemia and emerging genome editing technologies represent a new form of gene therapy. Approaches to cure SCD and beta-thalassemia via genome editing include: 1) Correction of HBB mutations by homology directed repair (HDR); 2) use of non-homologous end joining (NHEJ) to activate gamma globin production and raise fetal hemoglobin (HbF) levels; 3) NHEJ to disrupt alpha-globin genes (HBA1 or HBA2) and thereby alleviate globin chain imbalance in intermediately severe forms of beta thalassemia. Challenges for these approaches include selection of the most effective genome editing tools, optimizing their delivery to hematopoietic stem cells (HSCs), improving specificity and better understanding potential off target effects, particularly those that are biologically relevant. Technologies for genome editing are advancing rapidly and being tested in preclinical models for HBB-mutated disorders. Ultimately, however, the best strategies can only be identified in clinical trials. This will require close collaborations between basic/translational researchers who study genome editing, clinical hematologists and collaboration between experts in academia and the bio-pharmaceutical industry. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-23-SCI-23
Author(s):  
Giuliana Ferrari

Beta-thalassemia and sickle cell disease (SCD) are congenital anemias caused by mutations in the beta-globin gene, resulting in either reduced/absent production of globin chains or abnormal hemoglobin structure. At present, the definitive cure is represented by allogeneic hematopoietic stem cell transplantation, with a probability to find a well-matched donor of <25%. Experimental gene therapy for hemoglobinopathies is based on transplantation of autologous hematopoietic stem cells genetically modified to express therapeutic hemoglobin levels. Approaches to genetically modify HSCs for treatment of hemoglobinopathies include: 1) the addition of globin genes by lentiviral vectors and 2) gene editing by nucleases to reactivate fetal hemoglobin either through inhibition of repressors or by reproducing mutations associated with high fetal hemoglobin levels. The outcomes of early clinical trials are showing the safety and potential efficacy, as well as the hurdles still limiting a general application.Current challenges and improved strategies will be presented and discussed. Disclosures No relevant conflicts of interest to declare. OffLabel Disclosure: Plerixafor


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3993-3993
Author(s):  
Linda Yingqi Lin ◽  
Samuele Cancellieri ◽  
Jing Zeng ◽  
Francesco Masillo ◽  
My Anh Nguyen ◽  
...  

Abstract CRISPR gene editing holds great promise to modify somatic genomes to ameliorate disease. In silico prediction of homologous sites coupled with biochemical evaluation of possible genomic off-targets may predict genotoxicity risk of individual gene editing reagents. However, standard computational and biochemical methods focus on reference genomes and do not consider the impact of genetic diversity on off-target potential. Here we developed a web application called CRISPRme that explicitly and efficiently integrates human genetic variant datasets with orthogonal genomic annotations to predict and prioritize off-target sites at scale. The method considers both single-nucleotide variants (SNVs) and indels, accounts for bona fide haplotypes, accepts spacer:protospacer mismatches and bulges, and is suitable for personal genome analyses. We tested the tool with a guide RNA (gRNA) targeting the BCL11A erythroid enhancer that has shown therapeutic promise in clinical trials for sickle cell disease (SCD) and β-thalassemia (Frangoul et al. NEJM 2021). We find that the top predicted off-target site is produced by a non-reference allele common in African-ancestry populations (rs114518452, minor allele frequency (MAF) = 4.5%) that introduces a protospacer adjacent motif (PAM) for SpCas9. We validate that SpCas9 generates indels (~9.6% frequency) and chr2 pericentric inversions in a strictly allele-specific manner in edited CD34+ hematopoietic stem/progenitor cells (HSPCs), although a high-fidelity Cas9 variant mitigates this off-target. This report illustrates how population and private genetic variants should be considered as modifiers of genome editing outcomes. We expect that variant-aware off-target assessment will be required for therapeutic genome editing efforts going forward, including both ongoing and future clinical trials, and we provide a powerful approach for comprehensive off-target prediction. CRISPRme is available at crisprme.di.univr.it. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 2 ◽  
Author(s):  
Gloria Barbarani ◽  
Agata Łabedz ◽  
Antonella Ellena Ronchi

Hemoglobin is a tetrameric protein composed of two α and two β chains, each containing a heme group that reversibly binds oxygen. The composition of hemoglobin changes during development in order to fulfill the need of the growing organism, stably maintaining a balanced production of α-like and β-like chains in a 1:1 ratio. Adult hemoglobin (HbA) is composed of two α and two β subunits (α2β2 tetramer), whereas fetal hemoglobin (HbF) is composed of two γ and two α subunits (α2γ2 tetramer). Qualitative or quantitative defects in β-globin production cause two of the most common monogenic-inherited disorders: β-thalassemia and sickle cell disease. The high frequency of these diseases and the relative accessibility of hematopoietic stem cells make them an ideal candidate for therapeutic interventions based on genome editing. These strategies move in two directions: the correction of the disease-causing mutation and the reactivation of the expression of HbF in adult cells, in the attempt to recreate the effect of hereditary persistence of fetal hemoglobin (HPFH) natural mutations, which mitigate the severity of β-hemoglobinopathies. Both lines of research rely on the knowledge gained so far on the regulatory mechanisms controlling the differential expression of globin genes during development.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2150-2150
Author(s):  
Evangelia Yannaki ◽  
Thalia Papayannopoulou ◽  
Erica Jonlin ◽  
Ioannis Batsis ◽  
Pamela S. Becker ◽  
...  

Abstract Abstract 2150 Poster Board II-127 For gene therapy (GT) of thalassemia (TH), high numbers of genetically-modified hematopoietic stem cells (HSCs) are required to effectively compete for niche space in the hypercellular thalassemic bone marrow (bm). Mobilized peripheral blood is the preferable source of HSCs for thalassemia GT due to higher yields of CD34+cells compared to bm harvest. There is limited information on the mobilization efficacy of adult patients with major β-TH as well as on the safety of the procedure in a condition of splenomegaly and extramedullary hemopoiesis. Rare events of splenic rupture or thrombosis with G-CSF in normal donors raise safety concerns for its use in TH where chronic splenomegaly and hypercoagulability exist. Pretreatment of patients with hydroxyurea (HU) could reduce the risk of splenic rupture or thrombosis by decreasing the splenic hemopoiesis and thereby the spleen size in the non-splenectomized (non-SPL), and the circulating cells in the splenectomized (SPL) patients before G-CSF. In an on going mobilization study, we aim to assess the safety and efficacy of G-CSF mobilization with or without HU pretreatment in adult patients with β-TH major. Sufficient mobilization is considered to be the yield of ≥2×106CD34+cells/kg/2aphereses. Sixteen patients have been enrolled so far, 9 SPL and 7 non-SPL. One non-SPL patient withdrew during the study. Six SPL and 4 non-SPL patients received HU pretreatment (20mg/kg/d the non-SPL, 25-30mg/kg/d the SPL) for 1 month before G-CSF. There was a 1-2 weeks' interval between HU cessation and G-CSF initiation. No severe adverse events were observed. In non-SPL patients, HU decreased the spleen volume over baseline (306cm3 vs 536cm3, p=0,03) resulting in 9% max increase during mobilization compared to 45% size increase in patients w/o HU pretreatment. In non-SPL patients, HU negatively affected the CD34+yield when the ‘wash-out' period before G-CSF was 8 days (mean CD34+cells 1,86±0,76×106/kg/2aphereses, n=2). However, when the interval period from the HU stop to the G-CSF initiation increased up to 18 days allowing for bm recovery after the myelosuppressive effect of HU, mobilization was successful (P12:CD34+cells 6,5×106/kg/2aph). Non-SPL patients w/o HU pretreatment (n=3) yielded adequate numbers of HSCs (CD34+cells:5,8±3,89×106/kg/2aph). Surprisingly, CD34+cell yields were very low in the first 2 non-HU pretreated SPL patients (CD34+cells:0,98±0,14×106/kg/2aph). This was due to the development of early excessive leukocytosis (mean max WBCs 81×109/l, day 3) with the regular 10mcg/kg/d G-CSF dose, which necessitated dose hold or therapeutic leukapheresis and resulted in loss of the CD34+cell peak in blood. However, when mobilization started with lower (2,5mcg/kg/d) and adjusted to the WBCs doses of G-CSF (mean daily dose 3,21mcg/kg) and aphereses were initiated later (day6), CD34+cell yield markedly improved (P15:4,5×106/kg/2aph) without inducing early excessive leukocytosis (max WBCs 67×109/l, day 5). In SPL patients, HU was shown to decrease the high PLT and WBC numbers before G-CSF (PLTs:from 640±132×109/l at baseline to 240±53×109/l, p=0,0002 / WBCs:from 20,23±15,8×109/l at baseline to 11,47±6,11×109/l, p=0,23) potentially reducing the risk of thrombosis and partially preventing excessive leukocytosis during mobilization (max WBCs SPL-HU:51,68±29,37×109/l vs SPL-no HU:74,77±8,78×109/l, p=0,23). HU pretreatment negatively affected the yield in SPL patients when the ‘wash-out' period before G-CSF was 7-10 days (mean CD34+yield 0.62±0,41×106/kg/2aph, n=5). However, when the interval period from HU stop to G-CSF initiation increased up to 12 days, mobilization was successful (P16:CD34+cells 3,8×106/kg/2aphereses). G-CSF dose adjustment was also needed in HU-pretreated SPL patients with WBCs≥14,6×109/l before G-CSF (P9,P16). Overall, it seems that mobilization of SPL thalassemic patients is challenging. Mobilization is not inherently inefficient in SPL patients but it results from mandatory G-CSF-dose modifications to avoid hyperleukocytosis. Patient-tailored schemes of G-CSF mobilization or alternative ways of mobilization (ie AMD 3100) will be required in order to obtain high numbers of HSCs from SPL patients. HU seems to play a safety role as pretreatment before mobilization, especially in the SPL patients, however the time to G-CSF initiation after HU cessation is critical for a sufficient CD34+cell yield. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2901-2901
Author(s):  
Maria Rosa Lidonnici ◽  
Annamaria Aprile ◽  
Marta Claudia Frittoli ◽  
Giacomo Mandelli ◽  
Bernhard Gentner ◽  
...  

Abstract Successful gene therapy of inherited blood diseases relies on transplantation and engraftment of autologous genetically engineered hematopoietic stem/progenitor cells (HSPCs) in myeloablated patients. Hematopoietic reconstitution and clinical benefit are related to cell dose, although single disease features might play a role favoring selection of relevant progenitor populations. Gene therapy trials in young pediatric patients are performed isolating CD34+ cells from bone marrow (BM), while in adults mobilized peripheral blood stem cells (PBSC) should represent the favorite target. In the context of gene therapy for thalassemia, the choice of HSPC source is crucial since intrinsic characteristics of patients (splenomegaly and thrombophilia) dictate caution in the use of G-CSF as mobilization agent and prompt investigation of new agents. Moreover, adult thalassemic patients may possibly have a decreased BM stem cell reservoir, due to the BM suppression in response to multiple transfusions. A phase II clinical protocol exploring the use of Plerixafor as a single mobilizing agent in adult patients affected by transfusion dependent beta-thalassemia (EudraCT 2011-000973-30) started in 2012 at our hospital. Plerixafor selectively and reversibly antagonizes the binding of SDF-1 to its receptor CXCR4 with subsequent egress of HSCs to the peripheral blood. The availability of a new source of HSPCs, potentially superior in terms of CD34+ cell yield, transduction efficiency and biological features to steady-state BM, would have a significant impact on the feasibility and efficacy of gene therapy. Four subjects were enrolled and treated by subcutaneously administration of Plerixafor at the single dose of 0.24 mg/kg followed by leukoapheresis. Mobilization of CD34+ cells occurred very rapidly with a peak between 7 to 9 hrs. Three out of four patients achieved the minimal target cell dose (2 x 106 cells/kg) and no severe adverse event occurred. To the aim of engineering Plerixafor-mobilized CD34+ cells for gene therapy, we performed a comprehensive characterization of their biological, molecular and functional properties. In vivo reconstitution potential and lympho-myeloid differentiation were tested following transplantation in NSG mice and compared to those of PBSCs mobilized by G-CSF. Percentages of engrafted human cells in NSG mice transplanted with Plerixafor -PBSCs were about 2- to 5-fold higher than those found in mice transplanted with G-CSF PBSCs. On the same line, the SRC frequency, obtained by pooled engraftment data, was significantly higher (1 SRC out of 47.875 CD34+ cells vs.1 SRC out of 141.203 CD34+ cells). The phenotypic analysis of the frequency of primitive hematopoietic sub-populations revealed that Plerixafor mobilizes preferentially HSPCs and LT-HSPCs, with a percentage of CD34+ CD38-/low CD90+ CD45RA- CD49f+ cells higher than that found in G-CSF PBSCs. This result mirrors the enhanced number of SRCs found in the CD34+ cell population mobilized by Plerixafor. In order to further define the molecular features of HSPCs from different sources, we are studying signalling networks in response to specific cytokines by phospho-proteins analysis and gene expression by microarrays analysis. Our studies are focused on self-renewal, homing, engraftment and multilineage differentiation processes and bioinformatic analysis will reveal the molecular machinery underlying 'stemness' properties of Plerixafor mobilized cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3549-3549
Author(s):  
Sukanya Chumchuen ◽  
Tanapat Pornsukjantra ◽  
Pinyaphat Khamphikham ◽  
Usanarat Anurathapan ◽  
Orapan Sripichai ◽  
...  

LRF/ZBTB7A is a transcription factor that has been recently identified as a new key regulator of fetal hemoglobin (HbF; α2γ2) production in erythroid cells. Reduction of LRF/ZBTB7A expression led to increases in levels of HbF in human CD34+ hematopoietic stem and progenitor cell (HSPC)-derived erythroblast and in human immortalized erythroid line (HUDEP-2). Since reactivation of γ-globin gene is associated with the improvement of clinical manifestations of β-hemoglobinopathy patients, decrement in LRF/ZBTB7A expression might be a substantial interest as a novel target for gene therapy in β-thalassemia. In this study, we investigated the effects of LRF/ZBTB7A downregulation in erythroid cells derived from β-thalassemia/HbE patients in order to evaluate its therapeutic potential. The hematopoietic CD34+ progenitor cells were collected from 3 patients and 3 healthy normal individuals' peripheral blood and subjected for in vitro erythroblast culture. The cells were transduced with lentivirus carrying LRF/ZBTB7A specific shRNA, and used untransduced cells and non-targeted control shRNA (shNTC) as experimental controls. The LRF/ZBTB7A shRNA reduced LRF/ZBTB7A transcript and protein to nearly undetectable levels. Interestingly, downregulation of LRF/ZBTB7A increased expression of γ-globin, ε-globin and ζ-globin in both adult normal and β-thalassemia/HbE derived cells, whereas α-globin, β-globin and δ-globin expression were decreased. As previously reported, we found that the LRF/ZBTB7A knockdown produced a robust increase in HbF levels in both normal (43.3±9.0% vs. 5.9±2.1% in shNTC) and β-thalassemia/HbE erythroblasts (78.1±3.5% vs. 26.3±3.9% in shNTC). Noteworthy, the delay of erythroid differentiation was observed in the LRF/ZBTB7A knockdown cells of both derived from β-thalassemia/HbE patients and normal control, suggesting an additional role of LRF/ZBTB7A in regulating erythroid maturation. These data support the manipulation of LRF/ZBTB7A as one of the most interesting gene therapy candidates for treating the β-thalassemia, but the effect on erythroid cell maturation is needed to be concerned and required further investigation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1988 ◽  
Vol 72 (5) ◽  
pp. 1771-1776
Author(s):  
S Shiokawa ◽  
H Yamada ◽  
Y Takihara ◽  
E Matsunaga ◽  
Y Ohba ◽  
...  

A DNA fragment containing the deletion junction region from a Japanese individual with homozygous delta beta-thalassemia has been cloned. A clone containing the normal DNA surrounding the 3′ breakpoint of this deletion and a clone carrying the G gamma- and A gamma-globin genes of this patient were also isolated. Sequences of the deletion junction and both gamma-globin genes were determined. A comparison of these sequences with previously determined sequences of the normal counterparts revealed that the 5′ breakpoint is located between 2,134 and 2,137 base pairs (bp) 3′ to the polyA site of the A gamma-globin gene, the 5′ breakpoint is located just downstream of the 3′ border of the fetal gamma-globin duplication unit, and no molecular defects are evident within the gamma-globin gene region. A comparison between the sequences of the normal DNA surrounding the 3′ breakpoint and the normal DNA surrounding the 5′ breakpoint shows that deletion is the result of a nonhomologous recombination event. There are A+T-rich stretches near the 5′ and 3′ breakpoints in the normal DNA, and a portion of an Aly repeat is located in the region 3′ to the 3′ breakpoint. Southern blot analysis using probes 3′ to the beta-globin gene showed that the deletion extends in the 3′ direction further than any other deletions associated with delta beta-thalassemia and hereditary persistence of fetal hemoglobin (HPFH) heretofore reported. These results are discussed in terms of the mechanism generating large deletions in mammalian cells and three models for the regulation of gamma-globin and beta-globin gene expression in humans.


Sign in / Sign up

Export Citation Format

Share Document