Summary of opinion: Zynteglo,autologous CD34+ cell enriched population that contains hematopoietic stem cells transduced with lentiglobin BB305 lentiviral vector encoding the beta-A-T87Q-globin gene

2019 ◽  
Author(s):  
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.


2021 ◽  
Vol 12 ◽  
Author(s):  
Parin Rattananon ◽  
Usanarat Anurathapan ◽  
Kanit Bhukhai ◽  
Suradej Hongeng

β-thalassemia, a disease that results from defects in β-globin synthesis, leads to an imbalance of β- and α-globin chains and an excess of α chains. Defective erythroid maturation, ineffective erythropoiesis, and shortened red blood cell survival are commonly observed in most β-thalassemia patients. In severe cases, blood transfusion is considered as a mainstay therapy; however, regular blood transfusions result in chronic iron overload with life-threatening complications, e.g., endocrine dysfunction, cardiomyopathy, liver disease, and ultimately premature death. Therefore, transplantation of healthy hematopoietic stem cells (HSCs) is considered an alternative treatment. Patients with a compatible human leukocyte antigen (HLA) matched donor can be cured by allogeneic HSC transplantation. However, some recipients faced a high risk of morbidity/mortality due to graft versus host disease or graft failure, while a majority of patients do not have such HLA match-related donors. Currently, the infusion of autologous HSCs modified with a lentiviral vector expressing the β-globin gene into the erythroid progenitors of the patient is a promising approach to completely cure β-thalassemia. Here, we discuss a history of β-thalassemia treatments and limitations, in particular the development of β-globin lentiviral vectors, with emphasis on clinical applications and future perspectives in a new era of medicine.


2019 ◽  
Vol 3 (21) ◽  
pp. 3379-3392 ◽  
Author(s):  
Jean-Yves Métais ◽  
Phillip A. Doerfler ◽  
Thiyagaraj Mayuranathan ◽  
Daniel E. Bauer ◽  
Stephanie C. Fowler ◽  
...  

Key Points Cas9 editing of the γ-globin gene promoters in hematopoietic stem cells (HSCs) increases red cell HbF by ≤40%. No deleterious effects on hematopoiesis or off-target mutations were detected 16 weeks after xenotransplantation of edited HSCs.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1289-1289
Author(s):  
Ping Xia ◽  
Richard Emmanuel ◽  
Kuo Isabel ◽  
Malik Punam

Abstract We have previously shown that self-inactivating lentiviral vectors infect quiescent hematopoietic stem cells (HSC), express long-term, resist proviral silencing in HSC and express in a lineage specific manner. However, their random integration into the host chromosome results in variable expression, dependent upon the flanking host chromatin (Mohamedali et al, Mol. Therapy 2004). Moreover, the recent occurrence of leukemogenesis from activation of a cellular oncogene by the viral enhancer elements calls for safer vector designs, with expression cassettes that can be ‘insulated’ from flanking cellular genes. We analyzed the role of the chicken β-globin locus hypersensitive site 4 insulator element (cHS4) in a self-inactivating (SIN) lentiviral vector in the RBC progeny of hematopoietic stem cells (HSC) in long term in vivo. We designed an erythroid-specific SIN-lentiviral vector I8HKGW, expressing GFP driven by the human ankyrin gene promoter and containing two erythroid-specific enhancer elements and compared it to an analogous vector I8HKGW-I, where the cHS4 insulator was inserted in the SIN deletion to flank the I8HKGW expression cassette at both ends upon integration. First, murine erythroleukemia (MEL) cells were transduced at <5% transduction efficiency and GFP+ cells were sorted to generate clones. Single copy MEL clones showed no difference in the mean GFP fluorescence intensity (MFI) between the I8HKGW+ and the I8HKGW-I+ MEL clones. However, there was a reduction in the chromatin position effect variegation (PEV), reflected by reduced coefficient of variation of GFP expression (CV) in I8HKGW-I clones (n=115; P<0.01), similar to in vitro results reported by Ramezani et al (Blood 2003). Next, we examined for expression and PEV in the RBC progeny of HSC, using the secondary murine bone marrow transplant model. Lethally irradiated C57Bl6 (CD45.2) mice were transplanted with I8HKGW and I8HKGW-I transduced B6SJL (CD45.1) Sca+Lin- HSC and 4–6 months later, secondary transplants were performed. Mice were analyzed 3–4 months following secondary transplants (n=43). While expression from both I8HKGW and I8HKGW-I vectors appeared similar in secondary mice (46±6.0% vs. 48±3.6% GFP+ RBC; MFI 31±2.6 vs. 29±1.4), there were 0.37 vs. 0.22 copies/cell in I8HKGW and I8HKGW-I secondary recipients, respectively (n=43), suggesting that the probability of GFP expression from I8HKGW-I vectors was superior when equalized for vector copy. The CV of GFP fluorescence in RBC was remarkably reduced to 55±1.7 in I8HKGW-I vs. 196±32 in I8HKGW RBC (P<0.001). We therefore, analyzed these data at a clonal level in secondary CFU-S and tertiary CFU-S. The I8HKGW-I secondary CFU-S had more GFP+ cells (32.4±4.4%) vs. I8HKGW CFU-S (8.1±1.2%, n=143, P<0.1x10E-11). Similarly, I8HKGW-I tertiary CFU-S also had more GFP+ cells (25±1.8%) vs. I8HKGW CFU-S (6.3±0.8%, n=166, P<0.3x10E-10). We also plated bone marrow from secondary mice in methylcellulose and analyzed GFP expression in individual BFU-E. The I8HKGW-I tertiary BFU-E had more GFP+ cells (28±3.9%) vs. I8HKGW BFU-E (11±5%, n=50, P<0.03) with significantly reduced CV (67 vs 125, n=50, P<6.6X10E-7). Taken together, the ‘insulated’ erythroid-specific SIN-lentiviral vector increased the probability of expression of proviral integrants and reduced PEV in vivo, resulting in higher, consistent transgene expression in the erythroid cell progeny of HSC. In addition, the enhancer blocking effect of the cHS4, although not tested here, would further improve bio-safety of these vectors for gene therapy for RBC disorders.


Sign in / Sign up

Export Citation Format

Share Document