Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Publication year range
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
2.
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
3.
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
5.
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
6.
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
7.
Nature ; 467(7313): 318-22, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20844535

ABSTRACT

The ß-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of ß-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound ß(E)/ß(0)-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The ß(E)-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated ß(E)-globin with partial instability. When this is compounded with a non-functional ß(0) allele, a profound decrease in ß-globin synthesis results, and approximately half of ß(E)/ß(0)-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral ß-globin gene transfer, an adult patient with severe ß(E)/ß(0)-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl(-1), of which one-third contains vector-encoded ß-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Subject(s)
Blood Transfusion , Genetic Therapy , HMGA2 Protein/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Adolescent , Blood Cells/cytology , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Child, Preschool , Clone Cells/metabolism , Gene Expression , Genetic Vectors/genetics , HMGA2 Protein/genetics , Homeostasis , Humans , Lentivirus/genetics , Male , MicroRNAs/genetics , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/genetics , Time Factors , Transcriptional Activation , Young Adult , beta-Thalassemia/metabolism
8.
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
9.
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
10.
Blood ; 117(17): 4420-4, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21304104

ABSTRACT

The understanding of the hierarchical organization of the human hematopoietic system is of major biologic and clinical significance. The validity of the conventional model in which hematopoiesis is solely maintained by a pool of multipotent long-term hematopoietic stem cells (LT-HSCs) has been recently challenged by several mouse studies. These new data point to the existence of a heterogeneous stem cell population that consists of distinct subsets of LT-HSCs, which include stem cells biased toward lineage-specific differentiation programs. This review attempts to discuss the balanced versus biased patterns of lineage output of human LT-HSCs gathered in 3 different gene therapy trials on the basis of vector integration site analysis by deep sequencing. The distribution of integration sites observed tends to support the validity of the revised model.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Multipotent Stem Cells/physiology , Animals , Humans , Models, Biological
11.
Blood ; 117(20): 5321-31, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21436071

ABSTRACT

A challenge for gene therapy of genetic diseases is to maintain corrected cell populations in subjects undergoing transplantation in cases in which the corrected cells do not have intrinsic selective advantage over nontransduced cells. For inherited hematopoietic disorders, limitations include inefficient transduction of stem cell pools, the requirement for toxic myelosuppression, and a lack of optimal methods for cell selection after transduction. Here, we have designed a lentiviral vector that encodes human ß-globin and a truncated erythropoietin receptor, both under erythroid-specific transcriptional control. This truncated receptor confers enhanced sensitivity to erythropoietin and a benign course in human carriers. Transplantation of marrow transduced with the vector into syngenic thalassemic mice, which have elevated plasma erythropoietin levels, resulted in long-term correction of the disease even at low ratios of transduced/untransduced cells. Amplification of the red over the white blood cell lineages was self-controlled and averaged ∼ 100-fold instead of ∼ 5-fold for ß-globin expression alone. There was no detectable amplification of white blood cells or alteration of hematopoietic homeostasis. Notwithstanding legitimate safety concerns in the context of randomly integrating vectors, this approach may prove especially valuable in combination with targeted integration or in situ homologous recombination/repair and may lower the required level of pretransplantation myelosuppression.


Subject(s)
Genetic Therapy/methods , beta-Thalassemia/therapy , Animals , Base Sequence , DNA Primers/genetics , Disease Models, Animal , Erythropoiesis/genetics , Gene Expression , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Homeostasis , Humans , Lentivirus/genetics , Mice , Receptors, Erythropoietin/genetics , Recombinant Proteins/genetics , Transplantation, Isogeneic , beta-Globins/genetics , beta-Thalassemia/blood , beta-Thalassemia/genetics
12.
Mol Ther ; 19(7): 1273-86, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21386821

ABSTRACT

A lentiviral vector encoding ß-globin flanked by insulator elements has been used to treat ß-thalassemia (ß-Thal) successfully in one human subject. However, a clonal expansion was observed after integration in the HMGA2 locus, raising the question of how commonly lentiviral integration would be associated with possible insertional activation. Here, we report correcting ß-Thal in a murine model using the same vector and a busulfan-conditioning regimen, allowing us to investigate efficacy and clonal evolution at 9.2 months after transplantation of bone marrow cells. The five gene-corrected recipient mice showed near normal levels of hemoglobin, reduced accumulation of reticulocytes, and normalization of spleen weights. Mapping of integration sites pretransplantation showed the expected favored integration in transcription units. The numbers of gene-corrected long-term repopulating cells deduced from the numbers of unique integrants indicated oligoclonal reconstitution. Clonal abundance was quantified using a Mu transposon-mediated method, indicating that clones with integration sites near growth-control genes were not enriched during growth. No integration sites involving HMGA2 were detected. Cells containing integration sites in genes became less common after prolonged growth, suggesting negative selection. Thus, ß-Thal gene correction in mice can be achieved without expansion of cells harboring vectors integrated near genes involved in growth control.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , beta-Thalassemia/therapy , Animals , Bone Marrow Transplantation , Chromatography, High Pressure Liquid , Flow Cytometry , HMGA2 Protein/genetics , Mice , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
13.
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
15.
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
16.
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
17.
FASEB J ; 22(6): 1849-60, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18245171

ABSTRACT

Pulmonary arterial hypertension (PAH) is one of the leading causes of morbidity and mortality in adult patients with sickle cell disease (SCD). Here, we developed a model to study the early stage of PAH in SCD. We exposed wild-type and transgenic sickle cell SAD (Hbb(s)/Hbb(s)) mice to hypoxia (8% O(2)) for 7 days. Prolonged hypoxia in SAD mice only induced 1) increased neutrophil count in both bronchoalveolar lavage (BAL) and peripheral circulation; 2) increased BAL IL1beta, IL10, IL6, and TNF-alpha; and 3) up-regulation of the genes endothelin-1, cyclo-oxygenase-2, angiotensin-converting-enzyme, and IL-1beta, suggesting that amplified inflammatory response and activation of the endothelin-1 system may contribute to the early phase of PAH in SCD. Since phosphodiesterases (PDEs) are involved in pulmonary vascular tone regulation, we evaluated gene expression of phosphodiesterase-4 (PDE-4) isoforms and of PDE-1, -2, -3, -7, -8, which are the main cyclic-adenosine-monophosphate hydrolyzing enzymes. In SAD mouse lungs, prolonged hypoxia significantly increased PDE-4 and -1 gene expressions. The PDE-4 inhibitor, rolipram, prevented the hypoxia-induced PDE-4 and -1 gene up-regulation and interfered with the development of PAH, most likely through modulation of both vascular tone and inflammatory factors. This finding supports a possible therapeutic use of PDEs inhibitors in the earlier phases of PAH in SCD.


Subject(s)
Anemia, Sickle Cell/complications , Hypertension, Pulmonary/drug therapy , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Anemia, Sickle Cell/drug therapy , Animals , Disease Models, Animal , Hypoxia , Mice , Mice, Transgenic , Phosphodiesterase Inhibitors/therapeutic use , Rolipram/pharmacology , Up-Regulation/genetics
18.
Exp Hematol ; 36(4): 412-23, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18295963

ABSTRACT

OBJECTIVE: To investigate whether expression of a membrane-anchored form of erythropoietin (MbEpo) results in self-controlled, autocrine proliferation, and differentiation of erythroid cells. This would provide a possible approach to the selective expansion of genetically corrected erythroid cells in gene-therapy protocols. MATERIALS AND METHODS: We designed retroviral vectors encoding MbEpo or secreted erythropoietin (Epo) and enhanced green fluorescent protein. Several Epo-dependent cell lines were transduced and their proliferative capacity evaluated. This approach was also assessed in human bone marrow CD34(+) cells and mouse bone marrow transplants. RESULTS: Retroviral vector-mediated MbEpo expression induced autocrine proliferation of the Epo-dependent cell lines DAE7 and UT7/Epo. However, it blocked the Epo receptor (EpoR)-induced activation of granulocyte macrophage colony-stimulating factor-dependent UT7/GM cells and the erythroid differentiation of both human hematopoietic cells in vitro and of mouse bone marrow cells in transplant experiments. MbEpo was present at the surface of UT7/GM cells. It did not affect the membrane localization of the EpoR, but prevented its normal Epo-dependent phosphorylation and internalization. By contrast to these inhibitory effects, a higher rate of EpoR replenishment in UT7/GM cells before MbEpo production rendered cell proliferation independent of exogenous growth factor. CONCLUSIONS: Activation of EpoR gene expression before MbEpo-induced EpoR activation is essential for activation or inhibition of growth and differentiation of Epo-dependent cell lines. It will be necessary to delay MbEpo expression in late erythroid progenitors until after EpoR gene activation, for erythroid cell expansion to be achieved in vivo.


Subject(s)
Erythropoietin/metabolism , Hematopoietic Stem Cells/metabolism , Receptors, Cell Surface/metabolism , Receptors, Erythropoietin/metabolism , Animals , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, Cell Surface/drug effects , Receptors, Erythropoietin/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Up-Regulation
19.
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
20.
Hum Gene Ther ; 19(4): 331-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18341422

ABSTRACT

Numerous studies using erythropoietin (EPO) gene delivery vectors, either viral or nonviral, have shown uncontrolled EPO expression leading to transient or sustained erythrocytosis and, more recently, severe autoimmune anemia. Therefore, there is a need to develop other EPO gene delivery systems that allow sustained and adjustable expression of EPO. We have examined a new approach of delivering plasmid encoding mouse EPO cDNA into mouse skeletal muscle, using an amphiphilic block copolymer. Repeated injections of low doses of block copolymer-EPOcDNA formulations increased hematocrit in a dose-dependent manner for more than 9 months, without any initial overshoot. Low doses of block copolymer-EPOcDNA formulations prevented autoimmune anemia in immunocompetent Swiss mice and prevented or reversed chronic anemia in an acquired mouse model of renal failure. We conclude that repeated injections of low doses of block copolymer-DNA formulations that do not induce (1) inflammation at the injection site, (2) overexpression of EPO, or (3) the production of anti-EPO neutralizing auto-antibodies hold promise for in vivo expression of therapeutic proteins, in particular for systemic delivery.


Subject(s)
Anemia/complications , Anemia/therapy , Erythropoietin/genetics , Erythropoietin/therapeutic use , Genetic Therapy , Plasmids/genetics , Renal Insufficiency/complications , Anemia/chemically induced , Anemia/genetics , Animals , Creatinine/blood , Disease Models, Animal , Erythropoietin/immunology , Erythropoietin/metabolism , Female , Inflammation/pathology , Injections, Intramuscular , Kidney/pathology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Plasmids/administration & dosage , Polyethylene Glycols/pharmacology , Reticulocyte Count , Urea/blood
SELECTION OF CITATIONS
SEARCH DETAIL