Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cytotherapy ; 26(6): 586-591, 2024 06.
Article in English | MEDLINE | ID: mdl-38551525

ABSTRACT

BACKGROUND AIMS: Gene therapy using lentiviral vectors (LVs) that harbor a functional ß-globin gene provides a curative treatment for hemoglobinopathies including beta-thalassemia and sickle cell disease. Accurate quantification of the vector copy number (VCN) and/or the proportion of transduced cells is critical to evaluate the efficacy of transduction and stability of the transgene during treatment. Moreover, commonly used techniques for LV quantification, including real-time quantitative polymerase chain reaction (PCR) or fluorescence-activated cell sorting, require either a standard curve or expression of a reporter protein for the detection of transduced cells. In the present study, we describe a digital droplet PCR (ddPCR) technique to measure the lentiviral VCN in transduced hematopoietic stem and progenitor cells (HSPCs). METHODS: After HSPCs were transduced with an LV encoding the therapeutic ß-globin (ßA-T87Q) gene, the integrated lentiviral sequence in the host genome was amplified with primers that targeted a sequence within the vector and the human RPP30 gene. The dynamic range of ddPCR was between 5 × 10-3 ng and 5 × 10-6 ng of target copy per reaction. RESULTS: We found that the ddPCR-based approach was able to estimate VCN with high sensitivity and a low standard deviation. Furthermore, ddPCR-mediated quantitation of lentiviral copy numbers in differentiated erythroblasts correlated with the level of ßA-T87Q protein detected by reverse-phase high-performance liquid chromatography. CONCLUSIONS: Taken together, the ddPCR technique has the potential to precisely detect LV copy numbers in the host genome, which can be used for VCN estimation, calculation of infectious titer and multiplicity of infection for HSPC transduction in a clinical setting.


Subject(s)
Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cells , Lentivirus , Transduction, Genetic , beta-Globins , Humans , Lentivirus/genetics , Hematopoietic Stem Cells/metabolism , Genetic Vectors/genetics , beta-Globins/genetics , Transduction, Genetic/methods , Genetic Therapy/methods , beta-Thalassemia/therapy , beta-Thalassemia/genetics , Polymerase Chain Reaction/methods , Gene Dosage/genetics
3.
Nat Med ; 28(1): 81-88, 2022 01.
Article in English | MEDLINE | ID: mdl-35075288

ABSTRACT

Sickle cell disease (SCD) and transfusion-dependent ß-thalassemia (TDT) are the most prevalent monogenic disorders worldwide. Trial HGB-205 ( NCT02151526 ) aimed at evaluating gene therapy by autologous CD34+ cells transduced ex vivo with lentiviral vector BB305 that encodes the anti-sickling ßA-T87Q-globin expressed in the erythroid lineage. HGB-205 is a phase 1/2, open-label, single-arm, non-randomized interventional study of 2-year duration at a single center, followed by observation in long-term follow-up studies LTF-303 ( NCT02633943 ) and LTF-307 ( NCT04628585 ) for TDT and SCD, respectively. Inclusion and exclusion criteria were similar to those for allogeneic transplantation but restricted to patients lacking geno-identical, histocompatible donors. Four patients with TDT and three patients with SCD, ages 13-21 years, were treated after busulfan myeloablation 4.6-7.9 years ago, with a median follow-up of 4.5 years. Key primary endpoints included mortality, engraftment, replication-competent lentivirus and clonal dominance. No adverse events related to the drug product were observed. Clinical remission and remediation of biological hallmarks of the disease have been sustained in two of the three patients with SCD, and frequency of transfusions was reduced in the third. The patients with TDT are all transfusion free with improvement of dyserythropoiesis and iron overload.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy , Lentivirus/genetics , beta-Thalassemia/therapy , Adolescent , Female , Genetic Therapy/adverse effects , Humans , Male , Treatment Outcome , Young Adult
4.
Mol Ther ; 29(9): 2841-2853, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33940155

ABSTRACT

A primary challenge in lentiviral gene therapy of ß-hemoglobinopathies is to maintain low vector copy numbers to avoid genotoxicity while being reliably therapeutic for all genotypes. We designed a high-titer lentiviral vector, LVß-shα2, that allows coordinated expression of the therapeutic ßA-T87Q-globin gene and of an intron-embedded miR-30-based short hairpin RNA (shRNA) selectively targeting the α2-globin mRNA. Our approach was guided by the knowledge that moderate reduction of α-globin chain synthesis ameliorates disease severity in ß-thalassemia. We demonstrate that LVß-shα2 reduces α2-globin mRNA expression in erythroid cells while keeping α1-globin mRNA levels unchanged and ßA-T87Q-globin gene expression identical to the parent vector. Compared with the first ßA-T87Q-globin lentiviral vector that has received conditional marketing authorization, BB305, LVß-shα2 shows 1.7-fold greater potency to improve α/ß ratios. It may thus result in greater therapeutic efficacy and reliability for the most severe types of ß-thalassemia and provide an improved benefit/risk ratio regardless of the ß-thalassemia genotype.


Subject(s)
Genetic Vectors/administration & dosage , RNA, Small Interfering/genetics , alpha-Globins/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , Cell Line , Cells, Cultured , Down-Regulation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Genotype , Humans , K562 Cells , Lentivirus/genetics , Lentivirus/physiology , MicroRNAs/antagonists & inhibitors , Primary Cell Culture , Viral Load , beta-Thalassemia/therapy
5.
Viruses ; 12(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33353185

ABSTRACT

Human parvovirus B19 (B19V) causes various human diseases, ranging from childhood benign infection to arthropathies, severe anemia and fetal hydrops, depending on the health state and hematological status of the patient. To counteract B19V blood-borne contamination, evaluation of B19 DNA in plasma pools and viral inactivation/removal steps are performed, but nucleic acid testing does not correctly reflect B19V infectivity. There is currently no appropriate cellular model for detection of infectious units of B19V. We describe here an improved cell-based method for detecting B19V infectious units by evaluating its host transcription. We evaluated the ability of various cell lines to support B19V infection. Of all tested, UT7/Epo cell line, UT7/Epo-STI, showed the greatest sensitivity to B19 infection combined with ease of performance. We generated stable clones by limiting dilution on the UT7/Epo-STI cell line with graduated permissiveness for B19V and demonstrated a direct correlation between infectivity and S/G2/M cell cycle stage. Two of the clones tested, B12 and E2, reached sensitivity levels higher than those of UT7/Epo-S1 and CD36+ erythroid progenitor cells. These findings highlight the importance of cell cycle status for sensitivity to B19V, and we propose a promising new straightforward cell-based method for quantifying B19V infectious units.


Subject(s)
Cell Cycle , Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology , Parvovirus B19, Human/physiology , Biomarkers , Cell Line , Erythroid Cells/metabolism , Erythroid Cells/virology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/virology , Gene Expression , Gene Expression Regulation, Viral , Humans , Molecular Diagnostic Techniques , Parvoviridae Infections/metabolism , Sensitivity and Specificity , Viral Tropism
6.
BioDrugs ; 34(5): 625-647, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32897504

ABSTRACT

ß-Globin gene transfer has been used as a paradigm for hematopoietic stem cell (HSC) gene therapy, but is subject to major difficulties, such as the lack of selection of genetically corrected HSCs, the need for high-level expression of the therapeutic gene, and cell-specific transgene expression. It took more than 40 years for scientists and physicians to advance from the cloning of globin gene and discovering globin gene mutations to improving our understanding of the pathophysiological mechanisms involved, the detection of genetic modifiers, the development of animal models and gene transfer vectors, comprehensive animal testing, and demonstrations of phenotypic improvement in clinical trials, culminating in the authorization of the first gene therapy product for ß-thalassemia in 2019. Research has focused mostly on the development of lentiviral gene therapy vectors expressing variants of the ß-globin gene or, more recently, targeting a γ-globin repressor, some of which have entered clinical testing and should soon diversify the available treatments and promote price competition. These results are encouraging, but we have yet to reach the end of the story. New molecular and cellular tools, such as gene editing or the development of induced pluripotent stem cells, are being developed, heralding the emergence of alternative products, the efficacy and safety of which are being studied. Hemoglobin disorders constitute an important model for testing the pros and cons of these advanced technologies, some of which are already in the clinical phase. In this review, we focus on the development of the advanced products and recent technological innovations that could lead to clinical trials in the near future, and provide hope for a definitive cure of these severe conditions.


Subject(s)
Genetic Therapy , beta-Thalassemia , Animals , Gene Editing , Genetic Vectors , Therapies, Investigational , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy
7.
Hum Gene Ther ; 30(10): 1306-1323, 2019 10.
Article in English | MEDLINE | ID: mdl-30848170

ABSTRACT

Recent marketing approval for genetically engineered hematopoietic stem and T cells bears witness to the substantial improvements in lentiviral vectors over the last two decades, but evaluations of the long-term efficacy and toxicity of gene and cell therapy products will, nevertheless, require further studies in nonhuman primate models. Macaca fascicularis monkeys from Mauritius have a low genetic diversity and are particularly useful for reproducible drug testing. In particular, they have a genetically homogeneous class I major histocompatibility complex system that probably mitigates the variability of the response to simian immunodeficiency virus infection. However, the transduction of simian cells with human immunodeficiency virus type 1 (HIV-1)-derived vectors is inefficient due to capsid-specific restriction factors, such as the tripartite motif-containing protein tripartite motif 5α, which prevent infection with non-host-adapted retroviruses. This study introduced the modified capsid of the macaque-trophic HIV-1 clone MN4/LSQD into the packaging system and compared transduction efficiencies between hematopoietic cells transduced with this construct and cells transduced with HIV-1 NL4-3-derived packaging constructs. Capsid modification increased transduction efficiency in all hematopoietic cells tested (by factors of up to 10), including hematopoietic progenitor cells, repopulating cells, and T cells from Mauritian Macaca fascicularis, regardless of vector structure or purification method. The study also established culture conditions similar to those used in clinical practice for the efficient transduction of hematopoietic stem and progenitor CD34+ cells. These results suggest that the procedure is suitable for use in Mauritian Macaca fascicularis, which can therefore be used as a model in preclinical studies for hematopoietic gene and cell therapy.


Subject(s)
Capsid/immunology , Genetic Vectors/metabolism , HIV-1/immunology , Hematopoietic Stem Cells/immunology , Macaca fascicularis/immunology , Transduction, Genetic/methods , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Biomarkers/metabolism , Capsid/chemistry , Female , Gene Expression , Genetic Vectors/immunology , HIV-1/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/virology , Male , Mice , Mice, Inbred NOD , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Transplantation, Heterologous , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
8.
Exp Hematol ; 64: 12-32, 2018 08.
Article in English | MEDLINE | ID: mdl-29807062

ABSTRACT

The ß-hemoglobinopathies, transfusion-dependent ß-thalassemia and sickle cell disease, are the most prevalent inherited disorders worldwide and affect millions of people. Many of these patients have a shortened life expectancy and suffer from severe morbidity despite supportive therapies, which impose an enormous financial burden to societies. The only available curative therapy is allogeneic hematopoietic stem cell transplantation, although most patients do not have an HLA-matched sibling donor, and those who do still risk life-threatening complications. Therefore, gene therapy by one-time ex vivo modification of hematopoietic stem cells followed by autologous engraftment is an attractive new therapeutic modality. The first proof-of-principle of conversion to transfusion independence by means of a lentiviral vector expressing a marked and anti-sickling ßT87Q-globin gene variant was reported a decade ago in a patient with transfusion-dependent ß-thalassemia. In follow-up multicenter Phase II trials with an essentially identical vector (termed LentiGlobin BB305) and protocol, 12 of the 13 patients with a non-ß0/ß0 genotype, representing more than half of all transfusion-dependent ß-thalassemia cases worldwide, stopped red blood cell transfusions with total hemoglobin levels in blood approaching normal values. Correction of biological markers of dyserythropoiesis was achieved in evaluated patients. In nine patients with ß0/ß0 transfusion-dependent ß-thalassemia or equivalent severity (ßIVS1-110), median annualized transfusion volume decreased by 73% and red blood cell transfusions were stopped in three patients. Proof-of-principle of therapeutic efficacy in the first patient with sickle cell disease was also reported with LentiGlobin BB305. Encouraging results were presented in children with transfusion-dependent ß-thalassemia in another trial with the GLOBE lentiviral vector and several other gene therapy trials are currently open for both transfusion-dependent ß-thalassemia and sickle cell disease. Phase III trials are now under way and should help to determine benefit/risk/cost ratios to move gene therapy toward clinical practice.


Subject(s)
Genetic Vectors/therapeutic use , Hemoglobinopathies/therapy , Lentivirus/genetics , Anemia, Sickle Cell/therapy , Blood Transfusion , Clinical Trials as Topic , Developing Countries , Gene Editing , Genetic Vectors/genetics , Global Burden of Disease , Hematopoietic Stem Cell Transplantation , Hemoglobinopathies/epidemiology , Hemoglobinopathies/genetics , Humans , Iron Overload/etiology , Iron Overload/prevention & control , Mutagenesis, Site-Directed , Prevalence , Recombinant Proteins/genetics , Transplantation Conditioning/methods , beta-Globins/genetics , beta-Thalassemia/therapy
9.
N Engl J Med ; 378(16): 1479-1493, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29669226

ABSTRACT

BACKGROUND: Donor availability and transplantation-related risks limit the broad use of allogeneic hematopoietic-cell transplantation in patients with transfusion-dependent ß-thalassemia. After previously establishing that lentiviral transfer of a marked ß-globin (ßA-T87Q) gene could substitute for long-term red-cell transfusions in a patient with ß-thalassemia, we wanted to evaluate the safety and efficacy of such gene therapy in patients with transfusion-dependent ß-thalassemia. METHODS: In two phase 1-2 studies, we obtained mobilized autologous CD34+ cells from 22 patients (12 to 35 years of age) with transfusion-dependent ß-thalassemia and transduced the cells ex vivo with LentiGlobin BB305 vector, which encodes adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q). The cells were then reinfused after the patients had undergone myeloablative busulfan conditioning. We subsequently monitored adverse events, vector integration, and levels of replication-competent lentivirus. Efficacy assessments included levels of total hemoglobin and HbAT87Q, transfusion requirements, and average vector copy number. RESULTS: At a median of 26 months (range, 15 to 42) after infusion of the gene-modified cells, all but 1 of the 13 patients who had a non-ß0/ß0 genotype had stopped receiving red-cell transfusions; the levels of HbAT87Q ranged from 3.4 to 10.0 g per deciliter, and the levels of total hemoglobin ranged from 8.2 to 13.7 g per deciliter. Correction of biologic markers of dyserythropoiesis was achieved in evaluated patients with hemoglobin levels near normal ranges. In 9 patients with a ß0/ß0 genotype or two copies of the IVS1-110 mutation, the median annualized transfusion volume was decreased by 73%, and red-cell transfusions were discontinued in 3 patients. Treatment-related adverse events were typical of those associated with autologous stem-cell transplantation. No clonal dominance related to vector integration was observed. CONCLUSIONS: Gene therapy with autologous CD34+ cells transduced with the BB305 vector reduced or eliminated the need for long-term red-cell transfusions in 22 patients with severe ß-thalassemia without serious adverse events related to the drug product. (Funded by Bluebird Bio and others; HGB-204 and HGB-205 ClinicalTrials.gov numbers, NCT01745120 and NCT02151526 .).


Subject(s)
Genetic Therapy , beta-Globins/genetics , beta-Thalassemia/therapy , Adolescent , Adult , Antigens, CD34 , Child , Erythrocyte Transfusion/statistics & numerical data , Female , Gene Transfer Techniques , Genetic Vectors , Hemoglobins/analysis , Hemoglobins/genetics , Humans , Lentivirus/genetics , Male , Mutation , Transplantation, Autologous , Young Adult , beta-Thalassemia/genetics
10.
Mol Ther ; 26(2): 480-495, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29221807

ABSTRACT

Although gene transfer to hematopoietic stem cells (HSCs) has shown therapeutic efficacy in recent trials for several individuals with inherited disorders, transduction incompleteness of the HSC population remains a hurdle to yield a cure for all patients with reasonably low integrated vector numbers. In previous attempts at HSC selection, massive loss of transduced HSCs, contamination with non-transduced cells, or lack of applicability to large cell populations has rendered the procedures out of reach for human applications. Here, we fused codon-optimized puromycin N-acetyltransferase to herpes simplex virus thymidine kinase. When expressed from a ubiquitous promoter within a complex lentiviral vector comprising the ßAT87Q-globin gene, viral titers and therapeutic gene expression were maintained at effective levels. Complete selection and preservation of transduced HSCs were achieved after brief exposure to puromycin in the presence of MDR1 blocking agents, suggesting the procedure's suitability for human clinical applications while affording the additional safety of conditional suicide.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Transduction, Genetic , beta-Globins/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Disease Models, Animal , Gene Expression , Gene Order , Genes, Transgenic, Suicide , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Mice , Mice, Transgenic , Transgenes
11.
N Engl J Med ; 376(9): 848-855, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28249145

ABSTRACT

Sickle cell disease results from a homozygous missense mutation in the ß-globin gene that causes polymerization of hemoglobin S. Gene therapy for patients with this disorder is complicated by the complex cellular abnormalities and challenges in achieving effective, persistent inhibition of polymerization of hemoglobin S. We describe our first patient treated with lentiviral vector-mediated addition of an antisickling ß-globin gene into autologous hematopoietic stem cells. Adverse events were consistent with busulfan conditioning. Fifteen months after treatment, the level of therapeutic antisickling ß-globin remained high (approximately 50% of ß-like-globin chains) without recurrence of sickle crises and with correction of the biologic hallmarks of the disease. (Funded by Bluebird Bio and others; HGB-205 ClinicalTrials.gov number, NCT02151526 .).


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy , beta-Globins/genetics , Adolescent , Anemia, Sickle Cell/blood , Clinical Trials as Topic , Gene Expression , Genetic Therapy/adverse effects , Genetic Vectors , Hemoglobin A/metabolism , Humans , Lentivirus , Male
12.
Sci Rep ; 6: 35495, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759036

ABSTRACT

We have reported that of the 10 commonly used AAV serotype vectors, AAV6 is the most efficient in transducing primary human hematopoietic stem/progenitor cells (HSPCs). However, the transduction efficiency of the wild-type (WT) AAV6 vector varies greatly in HSPCs from different donors. Here we report two distinct strategies to further increase the transduction efficiency in HSPCs from donors that are transduced less efficiently with the WT AAV6 vectors. The first strategy involved modifications of the viral capsid proteins where specific surface-exposed tyrosine (Y) and threonine (T) residues were mutagenized to generate a triple-mutant (Y705 + Y731F + T492V) AAV6 vector. The second strategy involved the use of ex vivo transduction at high cell density. The combined use of these strategies resulted in transduction efficiency exceeding ~90% in HSPCs at significantly reduced vector doses. Our studies have significant implications in the optimal use of capsid-optimized AAV6 vectors in genome editing in HSPCs.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Transduction, Genetic , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Culture Techniques , Cell Line , Dependovirus/classification , Gene Editing , Gene Expression , Genes, Reporter , Hematopoietic Stem Cells/virology , Host-Pathogen Interactions , Humans , Species Specificity
13.
Biomed J ; 39(1): 24-38, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27105596

ABSTRACT

Beta-thalassemia is a group of frequent genetic disorders resulting in the synthesis of little or no ß-globin chains. Novel approaches are being developed to correct the resulting α/ß-globin chain imbalance, in an effort to move beyond the palliative management of this disease and the complications of its treatment (e.g. life-long red blood cell transfusion, iron chelation, splenectomy), which impose high costs on healthcare systems. Three approaches are envisaged: fetal globin gene reactivation by pharmacological compounds injected into patients throughout their lives, allogeneic hematopoietic stem cell transplantation (HSCT), and gene therapy. HSCT is currently the only treatment shown to provide an effective, definitive cure for ß-thalassemia. However, this procedure remains risky and histocompatible donors are identified for only a small fraction of patients. New pharmacological compounds are being tested, but none has yet made it into common clinical practice for the treatment of beta-thalassemia major. Gene therapy is in the experimental phase. It is emerging as a powerful approach without the immunological complications of HSCT, but with other possible drawbacks. Rapid progress is being made in this field, and long-term efficacy and safety studies are underway.


Subject(s)
Complementary Therapies , Hematopoietic Stem Cell Transplantation , Time , beta-Thalassemia/therapy , Animals , Complementary Therapies/methods , Humans , Tissue Donors , Treatment Outcome , beta-Thalassemia/diagnosis
14.
Hum Gene Ther ; 27(2): 148-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26886832

ABSTRACT

ß-globin gene disorders are the most prevalent inherited diseases worldwide and result from abnormal ß-globin synthesis or structure. Novel therapeutic approaches are being developed in an effort to move beyond palliative management. Gene therapy, by ex vivo lentiviral transfer of a therapeutic ß-globin gene derivative (ß(AT87Q)-globin) to hematopoietic stem cells, driven by cis-regulatory elements that confer high, erythroid-specific expression, has been evaluated in human clinical trials over the past 8 years. ß(AT87Q)-globin is used both as a strong inhibitor of HbS polymerization and as a biomarker. While long-term studies are underway in multiple centers in Europe and in the United States, proof-of-principle of efficacy and safety has already been obtained in multiple patients with ß-thalassemia and sickle cell disease.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , beta-Globins/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Clinical Trials as Topic , Gene Expression , Gene Transfer Techniques , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hematopoietic Stem Cells , Humans , Lentivirus/genetics , Lentivirus/metabolism , Patient Safety , Transgenes , Transplantation Conditioning/methods , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
15.
Mol Ther Methods Clin Dev ; 2: 15039, 2015.
Article in English | MEDLINE | ID: mdl-26528487

ABSTRACT

RNA delivery is an attractive strategy to achieve transient gene expression in research projects and in cell- or gene-based therapies. Despite significant efforts investigating vector-directed RNA transfer, there is still a requirement for better efficiency of delivery to primary cells and in vivo. Retroviral platforms drive RNA delivery, yet retrovirus RNA-packaging constraints limit gene transfer to two genome-molecules per viral particle. To improve retroviral transfer, we designed a dimerization-independent MS2-driven RNA packaging system using MS2-Coat-retrovirus chimeras. The engineered chimeric particles promoted effective packaging of several types of RNAs and enabled efficient transfer of biologically active RNAs in various cell types, including human CD34(+) and iPS cells. Systemic injection of high-titer particles led to gene expression in mouse liver and transferring Cre-recombinase mRNA in muscle permitted widespread editing at the ROSA26 locus. We could further show that the VLPs were able to activate an osteoblast differentiation pathway by delivering RUNX2- or DLX5-mRNA into primary human bone-marrow mesenchymal-stem cells. Thus, the novel chimeric MS2-lentiviral particles are a versatile tool for a wide range of applications including cellular-programming or genome-editing.

17.
Curr Gene Ther ; 15(1): 64-81, 2015.
Article in English | MEDLINE | ID: mdl-25429463

ABSTRACT

A previously published clinical trial demonstrated the benefit of autologous CD34(+) cells transduced with a selfinactivating lentiviral vector (HPV569) containing an engineered ß-globin gene (ß(A-T87Q)-globin) in a subject with ß thalassemia major. This vector has been modified to increase transduction efficacy without compromising safety. In vitro analyses indicated that the changes resulted in both increased vector titers (3 to 4 fold) and increased transduction efficacy (2 to 3 fold). An in vivo study in which 58 ß-thalassemic mice were transplanted with vector- or mock-transduced syngenic bone marrow cells indicated sustained therapeutic efficacy. Secondary transplantations involving 108 recipients were performed to evaluate long-term safety. The six month study showed no hematological or biochemical toxicity. Integration site (IS) profile revealed an oligo/polyclonal hematopoietic reconstitution in the primary transplants and reduced clonality in secondary transplants. Tumor cells were detected in the secondary transplant mice in all treatment groups (including the control group), without statistical differences in the tumor incidence. Immunohistochemistry and quantitative PCR demonstrated that tumor cells were not derived from transduced donor cells. This comprehensive efficacy and safety data provided the basis for initiating two clinical trials with this second generation vector (BB305) in Europe and in the USA in patients with ß-thalassemia major and sickle cell disease.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/genetics , Animals , Antigens, CD34/metabolism , Computational Biology , DNA Damage , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation , Male , Mice , Mice, Inbred C57BL , beta-Thalassemia/genetics
18.
Nat Med ; 20(4): 398-407, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24658077

ABSTRACT

The pathophysiology of ineffective erythropoiesis in ß-thalassemia is poorly understood. We report that RAP-011, an activin receptor IIA (ActRIIA) ligand trap, improved ineffective erythropoiesis, corrected anemia and limited iron overload in a mouse model of ß-thalassemia intermedia. Expression of growth differentiation factor 11 (GDF11), an ActRIIA ligand, was increased in splenic erythroblasts from thalassemic mice and in erythroblasts and sera from subjects with ß-thalassemia. Inactivation of GDF11 decreased oxidative stress and the amount of α-globin membrane precipitates, resulting in increased terminal erythroid differentiation. Abnormal GDF11 expression was dependent on reactive oxygen species, suggesting the existence of an autocrine amplification loop in ß-thalassemia. GDF11 inactivation also corrected the abnormal ratio of immature/mature erythroblasts by inducing apoptosis of immature erythroblasts through the Fas-Fas ligand pathway. Taken together, these observations suggest that ActRIIA ligand traps may have therapeutic relevance in ß-thalassemia by suppressing the deleterious effects of GDF11, a cytokine which blocks terminal erythroid maturation through an autocrine amplification loop involving oxidative stress and α-globin precipitation.


Subject(s)
Activin Receptors, Type II/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Erythroblasts/metabolism , Erythropoiesis/drug effects , Growth Differentiation Factors/antagonists & inhibitors , Hematinics/pharmacology , Recombinant Fusion Proteins/pharmacology , beta-Thalassemia/metabolism , Animals , Apoptosis/physiology , Autocrine Communication/physiology , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Disease Models, Animal , Fas Ligand Protein , Gene Amplification/physiology , Growth Differentiation Factors/metabolism , Ligands , Mice , Oxidative Stress/physiology , Reactive Oxygen Species , Signal Transduction , fas Receptor
19.
Stem Cells ; 31(9): 1785-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712774

ABSTRACT

A patient with ß(E)/ß(0) -thalassemia major was converted to transfusion-independence 4.5 years ago by lentiviral gene transfer in hematopoietic stem cells while showing a myeloid-biased cell clone. Induced pluripotent stem cells (iPSCs) are a potential alternative source of hematopoietic stem cells. If fetal to adult globin class, switching does not occur in vivo in iPSC-derived erythroid cells, ß-globin gene transfer would be unnecessary. To investigate both vector integration skewing and the potential use of iPSCs for the treatment of thalassemia, we derived iPSCs from the thalassemia gene therapy patient and compared iPSC-derived hematopoietic cells to their natural isogenic somatic counterparts. In NSG immunodeficient mice, embryonic to fetal and a partial fetal to adult globin class switching were observed, indicating that the gene transfer is likely necessary for iPSC-based therapy of the ß-hemoglobinopathies. Lentivector integration occurred in regions of low and high genotoxicity. Surprisingly, common integration sites (CIS) were identified across those iPSCs and cells retrieved from isogenic and nonisogenic gene therapy patients with ß-thalassemia and adrenoleukodystrophy, respectively. This suggests that CIS observed in the absence of overt tumorigenesis result from nonrandom lentiviral integration rather than oncogenic in vivo selection. These findings bring the use of iPSCs closer to practicality and further clarify our interpretation of genome-wide lentivector integration.


Subject(s)
Globins/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Lentivirus/metabolism , Transduction, Genetic , beta-Thalassemia/pathology , Adult , Animals , Cell Differentiation/drug effects , Erythroid Cells/cytology , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Gene Expression Regulation/drug effects , Genetic Vectors/metabolism , Globins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Mutagens/toxicity , Regeneration/drug effects , Virus Integration/drug effects
20.
Stem Cells ; 31(10): 2162-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23554255

ABSTRACT

Our understanding of system dynamics of mixed cell populations in whole organisms has benefited from the advent of individual cell marking by nonarrayed DNA barcodes subsequently analyzed by high-throughput DNA sequencing. However, key limitations include statistical biases compromising quantification and the lack of applicability to deconvolute individual cell fate in vivo after pooling single cells differentially exposed to different conditions ex vivo. Here, we have derived an arrayed lentiviral library of DNA barcodes and obtained a proof-of-concept of its resolving capacity by quantifying hematopoietic regeneration after engraftment of mice with genetically modified autologous cells. This method has helped clarify and bridge the seemingly opposed clonal-succession and continuous-recruitment models of hematopoietic stem cell behavior and revealed that myeloid-lymphoid biases are common occurrences in steady-state hematopoiesis. Arrayed lentiviral barcoding should prove a versatile and powerful approach to deconvolute cell dynamics in vivo with applications in hematology, embryology, and cancer biology.


Subject(s)
Hematopoietic Stem Cells/physiology , Lentivirus/genetics , Animals , Cell Tracking/methods , DNA Barcoding, Taxonomic , Genetic Vectors , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...