scholarly journals The New Self-Inactivating Lentiviral Vector for Thalassemia Gene Therapy Combining Two HPFH Activating Elements Corrects Human Thalassemic Hematopoietic Stem Cells

2012 ◽  
Vol 23 (1) ◽  
pp. 15-31 ◽  
Author(s):  
Eleni Papanikolaou ◽  
Maria Georgomanoli ◽  
Evangelos Stamateris ◽  
Fottes Panetsos ◽  
Markisia Karagiorga ◽  
...  
Gene Therapy ◽  
2020 ◽  
Vol 27 (12) ◽  
pp. 545-556
Author(s):  
Yoonjeong Jang ◽  
Yoon-Sang Kim ◽  
Matthew M. Wielgosz ◽  
Francesca Ferrara ◽  
Zhijun Ma ◽  
...  

Blood ◽  
2010 ◽  
Vol 115 (26) ◽  
pp. 5329-5337 ◽  
Author(s):  
Niek P. van Til ◽  
Merel Stok ◽  
Fatima S. F. Aerts Kaya ◽  
Monique C. de Waard ◽  
Elnaz Farahbakhshian ◽  
...  

AbstractPompe disease (acid α-glucosidase deficiency) is a lysosomal glycogen storage disorder characterized in its most severe early-onset form by rapidly progressive muscle weakness and mortality within the first year of life due to cardiac and respiratory failure. Enzyme replacement therapy prolongs the life of affected infants and supports the condition of older children and adults but entails lifelong treatment and can be counteracted by immune responses to the recombinant enzyme. We have explored the potential of lentiviral vector–mediated expression of human acid α-glucosidase in hematopoietic stem cells (HSCs) in a Pompe mouse model. After mild conditioning, transplantation of genetically engineered HSCs resulted in stable chimerism of approximately 35% hematopoietic cells that overexpress acid α-glucosidase and in major clearance of glycogen in heart, diaphragm, spleen, and liver. Cardiac remodeling was reversed, and respiratory function, skeletal muscle strength, and motor performance improved. Overexpression of acid α-glucosidase did not affect overall hematopoietic cell function and led to immune tolerance as shown by challenge with the human recombinant protein. On the basis of the prominent and sustained therapeutic efficacy without adverse events in mice we conclude that ex vivo HSC gene therapy is a treatment option worthwhile to pursue.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2903-2903
Author(s):  
Zejin Sun ◽  
Rikki Enzor ◽  
Paula Rio ◽  
D. Wade Clapp ◽  
Helmut Hanenberg

Abstract Fanconi anemia (FA) is a recessive DNA repair disorder characterized by bone marrow (BM) failure, genomic instability, and a predisposition to malignancies. Natural gene therapy due to molecular self-correction of hematopoietic stem cells (HSCs) has been reported in a minority of FA patients, suggesting that due to the in vivo selection advantage of the corrected cells, FA is an excellent model disease for stem cell gene therapy. However, the scarcity of autologous HSCs from FA patients for research purposes is one of the major road blocks to preclinical studies with human cells. Here, we developed a lentiviral vector with EGFP as marker gene that co-expresses two distinct shRNA sequences against FANCA under two different human promoters (H1 and U6). In vitro analysis in primary human fibroblasts showed that stable integration of this construct was highly efficient to induce the typical FA cellular phenotypes as assessed by (1) FANCD2 ubiquitination deficiency and (2) a characteristic G2/M arrest upon DNA damage induced by DNA crosslinking reagent Mitomycin C (MMC). We then transduced human cord blood (CB) CD34+ cells with this lentiviral vector and demonstrated a reduced survival of clonogenic cells in progenitor assays at 20nM MMC: 70% (scrambled control shRNA) vs. 23% (FANCA shRNA). This vector pseudotyped with a foamyviral envelope was then used to transduce CD34+ CB cells on fibronectin CH296. The next day, genetically modified cells were transplanted into NOD.Cg---Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. When analyzing the percentage of EGFP+ cells in the human graft (hCD45+ cells), we noticed a progressive decline of EGFP+ cells from 29% on day 5 to 5% at 4 months after transplantation in the peripheral blood of the recipient mice, mimicking the progressive BM failure in FA patients. In contrast, engraftment over time was stable in CD34+ cells transduced with scrambled control shRNA vector (33% on day 5 vs. 34% at 4 months). The human progenitors isolated from the BM of NSG recipient mice at sacrifice 4 months after initial transduction and transplantation are still hypersensitive to MMC, with a much lower survival rate of 34% at 20nM MMC in the FANCA shRNA group as compared to 78% in the scrambled control shRNA group, thus confirming the knockdown by the lentiviral shRNA construct is stable. In summary, the novel double shRNA lentiviral vector is capable of inducing all major hallmarks of FA cells in normal human CB CD34+ cells, thus providing unlimited FA-like cellular materials including NSG mice-repopulating HSCs for preclinical gene therapy and basic stem cell biology research in FA. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-19-SCI-19
Author(s):  
Philip D. Gregory

In the more than 20 years since the first successful gene transfer was performed in a human subject (Blaese, R.M. et al. Science 1995; 270, 475-480), the promise of gene therapy has been at once tantalizingly close and just out of reach. Today, gene-modified therapies are being used to treat a variety of diseases, and direct manipulation of the human genome for therapeutic effect is becoming a reality. While many advances in gene-based therapeutics have come in the oncology space, non-malignant hematologic and primary immune disorders remain important targets for these approaches. The ability to manipulate hematopoietic stem cells (HSCs) ex vivo and return the modified HSCs to the patient offers many potential advantages over allogeneic HSC transplantation. Additionally, the potential to introduce new genes with therapeutic potential, or to modify genes to modulate protein expression, may open new avenues for transformative therapies for genetic diseases. The progress of gene therapy for hemoglobinopathies - from the γ-retroviral vector technology that established the field, to the accumulating clinical experience with lentiviral vector-based gene therapy and the potential for gene editing-based approaches to address these diseases - provides insight into the development of genetic therapeutics. Transfusion-dependent β-thalassemia (TDT) and sickle cell disease (SCD) result from pathogenic mutations in the β-globin gene, HBB . Addition of functional HBB genes into autologous hematopoietic stem cells has the potential to offer the long-term therapeutic benefits of allogeneic HSC transplantation without the complications of graft vs host disease. The first human proof of concept of this approach came in a study of the HPV569 lentiviral vector coding for therapeutic β-globin, which was successfully introduced into the HSCs of patients with TDT and resulted in sustained clinical benefit in some patients (Cavazzana-Calvo et al., Nature 2010; 467(7313):318-22). Beyond restoring normal β-globin production, studies with HPV569 and its improved variant BB305, have shown that it is possible to drive expression of a β-globin variant with a point mutation, T87Q, designed to mimic the anti-sickling effect of γ-globin. This innovation may have important implications for patients with SCD, where introduction of normal β-globin may be insufficient to ameliorate the red blood cell sickling and polymerization that can cause painful and damaging vaso-occlusive crises. It has taken more than 20 years for HSC gene addition to reach safety and efficacy thresholds that may allow it to be used routinely for patients with hemoglobinopathies, but this approach is now nearing maturity. Important refinements in LVV architecture and advances in HBB gene cassette design have yielded promising results in multiple clinical studies of TDT and SCD (e.g. Ribeil J.A. et al., N Engl J Med 2017; 376:848-855). There is also significant excitement about the potential for gene editing approaches to address the hemoglobinopathies even as these technologies are just beginning to transition from lab to clinic. Critical questions of both efficacy and safety remain regarding the path forward for nuclease-based editing technologies such as CRISPR, ZFN, and megaTALs. Key lessons from the development of clinical gene addition therapies in the hemoglobinopathies may help chart the path forward for gene editing technologies. Disclosures Gregory: bluebirdbio: Employment; Merck KGaA: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2584-2584 ◽  
Author(s):  
Nestor W. Meza ◽  
Maria Eugenia Alonso ◽  
Susana Navarro ◽  
Guillermo Guenechea ◽  
Oscar Quintana-Bustamante ◽  
...  

Abstract Human erythrocyte R-type pyruvate kinase deficiency (PKD) is an autosomal recessive disorder produced by mutations in the PKLR gene. Haemolytic anaemia is the major symptom of the disease. A severe deficiency in PKD causes ATP depletion in the RBC metabolism, which ultimately leads to haemolysis that may require periodical blood transfusion, splenectomy, and in some cases bone marrow transplantation. These clinical features make this disease a good candidate for gene therapy. With this aim, we have developed different gammaretroviral and lentiviral vectors expressing the human RPK and have characterized the functionality of the erythroid specific expression regulatory sequences of the human RPK gene in a lentiviral backbone. The transduction of mouse hematopoietic stem cells from a mouse strain deficient in the pklr gene [AcB55: pklr269A/269A mice], which mainly resembles the human RPK deficiency, with a retroviral vector expressing the human RPK, followed by the transplantation into irradiated syngenic recipients completely recovered the red cell parameters in peripheral blood, spleen and bone marrow. Also, intracellular values of ATP, plasmatic iron and circulating erythropoietin levels were recovered to normal values. After 100 days of transplantation, treated mice did not show any clinical symptom of the disease. Secondary pklr269A/269A recipients were also transplanted and their hematological symptoms were also reverted, demonstrating the stable therapeutic efficacy of the vector. To specifically express the RPK in the erythroid lineage, a lentiviral vector expressing the EGFP marker gene under the above RPK promoter (LVpRPKEG) was constructed to test its specificity. EGFP expression was detected in erythroid, but not in non-erythroid cell lines, transduced with this vector. Human CD34+ cells were also transduced with the LVpRPKEG vector and transplanted into NOD/SCID mice. Forty days post-transplantation human bone marrow cells were obtained and seeded in semisolid media. Significantly, only human erythroid colonies expressed the EGFP protein, demonstrating the efficacy of the RPK promoter to specifically express proteins in the erythroid lineage. Experiments using a lentiviral vector expressing the human RPK gene under the control of the human RPK promoter are now in progress. Overall, the transfer of lentiviral vectors harbouring the hRPK cDNA driven by its own promoter in PKLR mutated hematopoietic stem cells could represent an efficient therapeutic treatment of severe clinical cases of human erythrocyte PKD.


Sign in / Sign up

Export Citation Format

Share Document