Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Blood Adv ; 4(4): 687-695, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32084259

ABSTRACT

The fetal-to-adult hemoglobin switch has been a focus of a long-standing effort to potentially treat sickle cell disease and ß thalassemia by induction of fetal hemoglobin. In a continuation of this effort, we designed specific transcriptional activator-like effectors (TALEs) to target both the Gγ and Aγ-globin promoters. We fused the TALEs to a LIM domain binding protein (Ldb1) dimerization domain, followed by a T2A green fluorescent protein (GFP) cassette, which were assembled into a lentiviral vector. To prevent deletions caused by the repeats of TALEs during the lentivirus packing process, we changed the TALE encoding DNA by codon optimization. Intriguingly, 5 of 14 TALEs showed forced reactivation of fetal-globin expression in human umbilical cord blood-derived erythroid progenitor (HUDEP-2) cells, with a significant increase in the γ-globin mRNA level by more than 70-fold. We also observed a more than 50% reduction of ß-globin mRNA. High-performance liquid chromatography analysis revealed more than 30% fetal globin in TALE-induced cells compared with the control of 2%. Among several promoters studied, the ß-globin gene promoter with the locus control region (LCR) enhancer showed the highest TALE expression during CD34 erythroid differentiation. At day 19 of differentiation, 2 TALEs increased fetal-globin expression more than 40-fold in the mRNA level and up to 70% of the total globin protein. These TALEs have potential for clinical translation.


Subject(s)
Transcription Activator-Like Effectors , gamma-Globins , Adult , Fetal Hemoglobin/genetics , Humans , Promoter Regions, Genetic , beta-Globins/genetics , gamma-Globins/genetics
2.
Mol Ther ; 25(5): 1163-1167, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28411016

ABSTRACT

The X-linked bleeding disorder hemophilia causes frequent and exaggerated bleeding that can be life-threatening if untreated. Conventional therapy requires frequent intravenous infusions of the missing coagulation protein (factor VIII [FVIII] for hemophilia A and factor IX [FIX] for hemophilia B). However, a lasting cure through gene therapy has long been sought. After a series of successes in small and large animal models, this goal has finally been achieved in humans by in vivo gene transfer to the liver using adeno-associated viral (AAV) vectors. In fact, multiple recent clinical trials have shown therapeutic, and in some cases curative, expression. At the same time, cellular immune responses against the virus have emerged as an obstacle in humans, potentially resulting in loss of expression. Transient immune suppression protocols have been developed to blunt these responses. Here, we provide an overview of the clinical development of AAV gene transfer for hemophilia, as well as an outlook on future directions.


Subject(s)
Factor IX/genetics , Factor VIII/genetics , Genetic Therapy/methods , Hemophilia A/therapy , Hemophilia B/therapy , Blood Transfusion , Dependovirus/genetics , Dependovirus/immunology , Factor IX/metabolism , Factor VIII/metabolism , Gene Expression , Genetic Therapy/trends , Genetic Vectors/chemistry , Genetic Vectors/immunology , Hemophilia A/genetics , Hemophilia A/metabolism , Hemophilia A/pathology , Hemophilia B/genetics , Hemophilia B/metabolism , Hemophilia B/pathology , Humans , Lentivirus/genetics , Lentivirus/immunology , Mutation
3.
Mol Ther ; 25(3): 593-605, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28190779

ABSTRACT

Recently, an engineered Homeobox-nucleoporin fusion gene, NUP98-HOXA10HD or NA10HD, was reported to expand and maintain murine hematopoietic stem cells (HSCs). We postulated that NA10HD would increase the number of human γ-globin-expressing cells to therapeutic levels. We developed a double gene lentiviral vector encoding both human γ-globin and NA10HD, which was used to transduce human peripheral blood CD34+ cells and increased engraftment 2- to 2.5-fold at 15 weeks post-transplantation in immunodeficient mice. In ß-thalassemic mice transplanted with ß-thalassemic HSCs transduced with the γ-globin/NA10HD vector, the number of fetal hemoglobin (HbF)-expressing cells was significantly increased after 3 months, leading to resolution of the anemia. Furthermore, the increases in HbF were maintained at 6 months and persisted after secondary transplantation. In addition, NA10HD enrichment of transduced HSCs led to HbF increases without affecting homeostasis of the white blood cell lineages. Our results suggest that NA10HD increases the number of γ-globin-transduced HSCs that engraft, leading to an elevated number of fetal hemoglobin-containing red cells. These effects of NA10HD provide an improved platform for testing of the therapeutic efficacy of novel globin vectors and provide further impetus to develop safe and effective methods for selective expansion of genetically modified cells.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Lentivirus/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics , Animals , Disease Models, Animal , Erythrocytes/cytology , Erythrocytes/metabolism , Fetal Hemoglobin/metabolism , Gene Order , Gene Transfer Techniques , Genetic Loci , Graft Survival , Hematopoietic Stem Cell Transplantation , Homeobox A10 Proteins , Humans , Mice , Transduction, Genetic , Transplantation, Heterologous , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
4.
Mol Ther Methods Clin Dev ; 3: 16077, 2016.
Article in English | MEDLINE | ID: mdl-28035317

ABSTRACT

Chromosome Conformation Capture (3C) technology was used to identify physical interactions between the proximal Wiskott-Aldrich Syndrome protein (WASp) promoter and its distant DNA segments in Jurkat-T cells. We found that two hematopoietic specific DNase I hypersensitive (DHS) sites (proximal DHS-A, and distal DHS-B) which had high interaction frequencies with the proximal WASp promoter indicating potential regulatory activity for these DHS sites. Subsequently, we cloned several DNA fragments around the proximal DHS-A site into a luciferase reporter vector. Interestingly, no fragments showed enhancer activity, but two fragments exhibited strong silencing activity in Jurkat-T cells. After aligning the chromatin state profiling for hematopoietic and nonhematopoietic cells using the human genome browser (UCSC), we found a 5 kb putative hematopoietic specific enhancer region located 250 kb downstream of the WAS gene. This putative enhancer region contains two hematopoietic cell specific DHS sites. Subsequently, the hematopoietic specific DHS sites enhanced luciferase expression from the proximal WASp promoter in all hematopoietic cells we tested. Finally, using a lentiviral vector stable expression system, the hematopoietic specific-enhancer(s) increased GFP reporter gene expression in hematopoietic cells, and increased WASp gene expression in WASp deficient cells. This enhancer may have the potential to be used in gene therapy for hematological diseases.

5.
Hum Gene Ther ; 27(4): 305-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27021603

ABSTRACT

Adeno-associated viral vectors have been developed for the treatment of hemophilia A and B. Derivation of vector particles is achieved after multiplasmid transfection of cells that package the vector genome to yield vector particles. To date, three clinical trials have been performed for hemophilia B. The results of these trials are described. The trial that we conducted with our collaborators has yielded evidence of clinical efficacy for hemophilia B. A vector for treating hemophilia A has been developed and a clinical trial is planned.


Subject(s)
Genetic Therapy , Hemophilia A/therapy , Hemophilia B/therapy , Animals , Clinical Trials as Topic , Dependovirus/metabolism , Humans
7.
Mol Ther Methods Clin Dev ; 2: 14063, 2015.
Article in English | MEDLINE | ID: mdl-26052531

ABSTRACT

We have developed a producer cell line that generates lentiviral vector particles of high titer. The vector encodes the Wiskott-Aldrich syndrome (WAS) protein. An insulator element has been added to the long terminal repeats of the integrated vector to limit proto-oncogene activation. The vector provides high-level, stable expression of WAS protein in transduced murine and human hematopoietic cells. We have also developed a monoclonal antibody specific for intracellular WAS protein. This antibody has been used to monitor expression in blood and bone marrow cells after transfer into lineage negative bone marrow cells from WAS mice and in a WAS negative human B-cell line. Persistent expression of the transgene has been observed in transduced murine cells 12-20 weeks following transplantation. The producer cell line and the specific monoclonal antibody will facilitate the development of a clinical protocol for gene transfer into WAS protein deficient stem cells.

9.
N Engl J Med ; 371(21): 1994-2004, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25409372

ABSTRACT

BACKGROUND: In patients with severe hemophilia B, gene therapy that is mediated by a novel self-complementary adeno-associated virus serotype 8 (AAV8) vector has been shown to raise factor IX levels for periods of up to 16 months. We wanted to determine the durability of transgene expression, the vector dose-response relationship, and the level of persistent or late toxicity. METHODS: We evaluated the stability of transgene expression and long-term safety in 10 patients with severe hemophilia B: 6 patients who had been enrolled in an initial phase 1 dose-escalation trial, with 2 patients each receiving a low, intermediate, or high dose, and 4 additional patients who received the high dose (2×10(12) vector genomes per kilogram of body weight). The patients subsequently underwent extensive clinical and laboratory monitoring. RESULTS: A single intravenous infusion of vector in all 10 patients with severe hemophilia B resulted in a dose-dependent increase in circulating factor IX to a level that was 1 to 6% of the normal value over a median period of 3.2 years, with observation ongoing. In the high-dose group, a consistent increase in the factor IX level to a mean (±SD) of 5.1±1.7% was observed in all 6 patients, which resulted in a reduction of more than 90% in both bleeding episodes and the use of prophylactic factor IX concentrate. A transient increase in the mean alanine aminotransferase level to 86 IU per liter (range, 36 to 202) occurred between week 7 and week 10 in 4 of the 6 patients in the high-dose group but resolved over a median of 5 days (range, 2 to 35) after prednisolone treatment. CONCLUSIONS: In 10 patients with severe hemophilia B, the infusion of a single dose of AAV8 vector resulted in long-term therapeutic factor IX expression associated with clinical improvement. With a follow-up period of up to 3 years, no late toxic effects from the therapy were reported. (Funded by the National Heart, Lung, and Blood Institute and others; ClinicalTrials.gov number, NCT00979238.).


Subject(s)
Factor IX/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Hemophilia B/therapy , Adult , Alanine Transaminase/blood , Dependovirus/genetics , Factor IX/metabolism , Follow-Up Studies , Gene Expression , Genetic Therapy/adverse effects , Hemophilia B/blood , Hemophilia B/genetics , Humans , Infusions, Intravenous , Male , Middle Aged , Transgenes , Young Adult
10.
Blood ; 122(9): 1556-64, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23843498

ABSTRACT

This review addresses the current status of gene therapy for immunodeficiencies, chronic granulomatous disease, suicide gene therapy for graft-versus-host disease, viral infections, malignant hematologic disorders, hemophilia, and the hemoglobin disorders. New developments in vector design have fostered improved expression as well as enhanced safety, particularly of integrating retroviral vectors. Several immunodeficiencies have been treated successfully by stem cell-targeted, retroviral-mediated gene transfer with reconstitution of the immune system following infusion of the transduced cells. In a trial for hemophilia B, long-term expression of human FIX has been observed following adeno-associated viral vector-mediated gene transfer into the liver. This approach should be successful in treating any disorder in which liver production of a specific protein is therapeutic.


Subject(s)
Genetic Therapy/methods , Hematologic Diseases/therapy , Adenoviridae , Animals , Dependovirus , Genetic Vectors , Humans , Retroviridae
11.
Hum Gene Ther Clin Dev ; 24(2): 77-85, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23786330

ABSTRACT

Gene therapy for the treatment of Wiskott-Aldrich syndrome (WAS) presents an alternative to the current use of allogeneic bone marrow transplantation. We describe the development of a self-inactivating lentiviral vector containing chromatin insulators for treatment of WAS and compare a gammaretroviral (MND), human cellular (EF1α), and the human WASp gene promoter for expression patterns in vivo during murine hematopoiesis using the green fluorescent protein (GFP) marker. Compared with the EF1α and the WASp promoters, expression from the MND promoter in mouse transplant recipients was much higher in all lineages examined. Importantly, there was sustained expression in the platelets of secondary recipient animals, necessary to correct the thrombocytopenia defect in WAS patients. Analysis of WAS protein expression in transduced human EBV-immortalized B-cells and transduced patient peripheral blood mononuclear cells also demonstrated stronger expression per copy from the MND promoter compared with the other promoters. In addition, when analyzed in an LM02 activation assay, the addition of an insulator to MND-promoter-containing constructs reduced transactivation of the LM02 gene. We propose a clinical trial design in which cytokine-mobilized, autologous, transduced CD34(+) cells are administered after myelosuppression.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/therapy , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blood Platelets/cytology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cells, Cultured , Chromatin/metabolism , Gammaretrovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Platelet Count , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transduction, Genetic , Wiskott-Aldrich Syndrome Protein/genetics
12.
Blood ; 121(17): 3335-44, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23426947

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors encoding human factor VIII (hFVIII) were systematically evaluated for hemophilia A (HA) gene therapy. A 5.7-kb rAAV-expression cassette (rAAV-HLP-codop-hFVIII-N6) containing a codon-optimized hFVIII cDNA in which a 226 amino acid (aa) B-domain spacer replaced the entire B domain and a hybrid liver-specific promoter (HLP) mediated 10-fold higher hFVIII levels in mice compared with non-codon-optimized variants. A further twofold improvement in potency was achieved by replacing the 226-aa N6 spacer with a novel 17-aa peptide (V3) in which 6 glycosylation triplets from the B domain were juxtaposed. The resulting 5.2-kb rAAV-HLP-codop-hFVIII-V3 cassette was more efficiently packaged within AAV virions and mediated supraphysiologic hFVIII expression (732 ± 162% of normal) in HA knock-out mice following administration of 2 × 10(12) vector genomes/kg, a vector dose shown to be safe in subjects with hemophilia B. Stable hFVIII expression at 15 ± 4% of normal was observed at this dose in a nonhuman primate. hFVIII expression above 100% was observed in 3 macaques that received a higher dose of either this vector or the N6 variant. These animals developed neutralizing anti-FVIII antibodies that were abrogated with transient immunosuppression. Therefore, rAAV-HLP-codop-hFVIII-V3 substantially improves the prospects of effective HA gene therapy.


Subject(s)
Dependovirus/genetics , Factor VIII/pharmacology , Genetic Therapy , Genetic Variation/genetics , Genetic Vectors/administration & dosage , Hemophilia A/therapy , Animals , Blotting, Western , Factor VIII/genetics , Factor VIII/immunology , Glycosylation , Hemophilia A/genetics , Humans , Immune Tolerance , Liver/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/genetics , Peptide Fragments/metabolism , Promoter Regions, Genetic/genetics
13.
Cold Spring Harb Perspect Med ; 2(12): a011726, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23209183

ABSTRACT

The ß-thalassemia syndromes reflect deficient or absent ß-globin synthesis usually owing to a mutation in the ß-globin locus. The relative excess of α-globin results in the formation of insoluble aggregates leading to ineffective erythropoiesis and shortened red cell survival. A relatively high capacity for fetal hemoglobin synthesis is a major genetic modifier of disease severity, with polymorphisms in other genes also having a significant role. Iron overload secondary to enhanced absorption and red cell transfusions causes an increase in liver iron and in various other tissues, leading to endocrine and cardiac dysfunction. Modern chelation regimens are effective in removing iron and preserving or restoring organ function.


Subject(s)
beta-Thalassemia/genetics , Anemia/genetics , Endocrine System Diseases/genetics , Globins/biosynthesis , Heart Diseases/genetics , Hemoglobin E/genetics , Heterozygote , Homozygote , Humans , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Iron Overload/genetics , Liver Diseases/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , beta-Thalassemia/drug therapy , beta-Thalassemia/metabolism
14.
Article in English | MEDLINE | ID: mdl-23125203

ABSTRACT

Retroviral vector-mediated gene transfer into hematopoietic stem cells provides a potentially curative therapy for severe ß-thalassemia. Lentiviral vectors based on human immunodeficiency virus have been developed for this purpose and have been shown to be effective in curing thalassemia in mouse models. One participant in an ongoing clinical trial has achieved transfusion independence after gene transfer into bone marrow stem cells owing, in part, to a genetically modified, dominant clone. Ongoing efforts are focused on improving the efficiency of lentiviral vector-mediated gene transfer into stem cells so that the curative potential of gene transfer can be consistently achieved.


Subject(s)
Genetic Therapy , Genetic Vectors , Globins/genetics , Thalassemia/therapy , Animals , Clinical Trials as Topic , Disease Models, Animal , Gammaretrovirus , Genetic Vectors/adverse effects , Humans , Lentivirus , Locus Control Region , Stem Cells
15.
Hum Gene Ther Methods ; 23(1): 1-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22428975

ABSTRACT

Self-complementary AAV (scAAV) vector genomes contain a covalently closed hairpin derived from a mutated inverted terminal repeat that connects the two monomer single-stranded genomes into a head-to-head or tail-to-tail dimer. We found that during quantitative PCR (qPCR) this structure inhibits the amplification of proximal amplicons and causes the systemic underreporting of copy number by as much as 10-fold. We show that cleavage of scAAV vector genomes with restriction endonuclease to liberate amplicons from the covalently closed terminal hairpin restores quantitative amplification, and we implement this procedure in a simple, modified qPCR titration method for scAAV vectors. In addition, we developed and present an AAV genome titration procedure based on gel electrophoresis that requires minimal sample processing and has low interassay variability, and as such is well suited for the rigorous quality control demands of clinical vector production facilities.


Subject(s)
DNA Copy Number Variations/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Polymerase Chain Reaction/methods , Research Design , DNA Primers/genetics , DNA Restriction Enzymes/metabolism , Dimerization , Electrophoresis/methods , Immunoblotting , Mutation/genetics , Nucleic Acid Amplification Techniques/methods , Quality Control , Terminal Repeat Sequences/genetics
16.
N Engl J Med ; 365(25): 2357-65, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22149959

ABSTRACT

BACKGROUND: Hemophilia B, an X-linked disorder, is ideally suited for gene therapy. We investigated the use of a new gene therapy in patients with the disorder. METHODS: We infused a single dose of a serotype-8-pseudotyped, self-complementary adenovirus-associated virus (AAV) vector expressing a codon-optimized human factor IX (FIX) transgene (scAAV2/8-LP1-hFIXco) in a peripheral vein in six patients with severe hemophilia B (FIX activity, <1% of normal values). Study participants were enrolled sequentially in one of three cohorts (given a high, intermediate, or low dose of vector), with two participants in each group. Vector was administered without immunosuppressive therapy, and participants were followed for 6 to 16 months. RESULTS: AAV-mediated expression of FIX at 2 to 11% of normal levels was observed in all participants. Four of the six discontinued FIX prophylaxis and remained free of spontaneous hemorrhage; in the other two, the interval between prophylactic injections was increased. Of the two participants who received the high dose of vector, one had a transient, asymptomatic elevation of serum aminotransferase levels, which was associated with the detection of AAV8-capsid-specific T cells in the peripheral blood; the other had a slight increase in liver-enzyme levels, the cause of which was less clear. Each of these two participants received a short course of glucocorticoid therapy, which rapidly normalized aminotransferase levels and maintained FIX levels in the range of 3 to 11% of normal values. CONCLUSIONS: Peripheral-vein infusion of scAAV2/8-LP1-hFIXco resulted in FIX transgene expression at levels sufficient to improve the bleeding phenotype, with few side effects. Although immune-mediated clearance of AAV-transduced hepatocytes remains a concern, this process may be controlled with a short course of glucocorticoids without loss of transgene expression. (Funded by the Medical Research Council and others; ClinicalTrials.gov number, NCT00979238.).


Subject(s)
Dependovirus , Factor IX/genetics , Genetic Therapy , Genetic Vectors , Hemophilia B/therapy , Adult , Dependovirus/genetics , Factor IX/therapeutic use , Genetic Therapy/adverse effects , Genetic Vectors/immunology , Humans , Infusions, Intravenous , Middle Aged , Transgenes/immunology
17.
Hum Gene Ther ; 22(5): 595-604, 2011 May.
Article in English | MEDLINE | ID: mdl-21410419

ABSTRACT

To generate sufficient clinical-grade vector to support a phase I/II clinical trial of adeno-associated virus serotype 8 (AAV8)-mediated factor IX (FIX) gene transfer for hemophilia B, we have developed a large-scale, good manufacturing practice (GMP)-compatible method for vector production and purification. We used a 293T-based two-plasmid transient transfection system coupled with a three-column chromatography purification process to produce high-quality self-complementary AAV2/8 FIX clinical-grade vector. Two consecutive production campaigns using a total of 432 independent 10-stack culture chambers produced a total of ∼2 × 10(15) vector genomes (VG) by dot-blot hybridization. Benzonase-treated microfluidized lysates generated from pellets of transfected cells were purified by group separation on Sepharose beads followed by anion-exchange chromatography. The virus-containing fractions were further processed by gel filtration and ultrafiltration, using a 100-kDa membrane. The vector was formulated in phosphate-buffered saline plus 0.25% human serum albumin. Spectrophotometric analysis suggested ∼20% full particles, with only low quantities of nonviral proteins were visible on silver-stained sodium dodecyl sulfate-polyacrylamide gels. A sensitive assay for the detection of replication-competent AAV was developed, which did reveal trace quantities of such contaminants in the final product. Additional studies have confirmed the long-term stability of the vector at -80°C for at least 24 months and for at least 24 hr formulated in the clinical diluent and stored at room temperature within intravenous bags. This material has been approved for use in clinical trials in the United States and the United Kingdom.


Subject(s)
Biotechnology/methods , Dependovirus , Genetic Therapy/methods , Genetic Vectors/genetics , Hemophilia B/therapy , Cell Line , Chromatography, Gel , Chromatography, Ion Exchange , Clinical Trials as Topic , Hemophilia B/genetics , Humans , Immunoblotting , Spectrophotometry
18.
Blood ; 117(15): 3945-53, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21321359

ABSTRACT

In humans, embryonic, fetal, and adult hemoglobins are sequentially expressed in developing erythroblasts during ontogeny. For the past 40 years, this process has been the subject of intensive study because of its value to enlighten the biology of developmental gene regulation and because fetal hemoglobin can significantly ameliorate the clinical manifestations of both sickle cell disease and ß-thalassemia. Understanding the normal process of loss of fetal globin expression and activation of adult globin expression could potentially lead to new therapeutic approaches for these hemoglobin disorders. Herein, we briefly review the history of the study of hemoglobin switching and then focus on recent discoveries in the field that now make new therapeutic approaches seem feasible in the future. Erythroid-specific knockdown of fetal gene repressors or enforced expression of fetal gene activators may provide clinically applicable approaches for genetic treatment of hemoglobin disorders that would benefit from increased fetal hemoglobin levels.


Subject(s)
Fetal Hemoglobin/genetics , Gene Expression Regulation, Developmental/physiology , Transcription, Genetic/physiology , Humans
19.
Mol Ther ; 19(5): 876-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21245849

ABSTRACT

Adeno-associated virus vectors (AAV) show promise for liver-targeted gene therapy. In this study, we examined the long-term consequences of a single intravenous administration of a self-complementary AAV vector (scAAV2/ 8-LP1-hFIXco) encoding a codon optimized human factor IX (hFIX) gene in 24 nonhuman primates (NHPs). A dose-response relationship between vector titer and transgene expression was observed. Peak hFIX expression following the highest dose of vector (2 × 10(12) pcr-vector genomes (vg)/kg) was 21 ± 3 µg/ml (~420% of normal). Fluorescent in-situ hybridization demonstrated scAAV provirus in almost 100% of hepatocytes at that dose. No perturbations of clinical or laboratory parameters were noted and vector genomes were cleared from bodily fluids by 10 days. Macaques transduced with 2 × 10(11) pcr-vg/kg were followed for the longest period (~5 years), during which time expression of hFIX remained >10% of normal level, despite a gradual decline in transgene copy number and the proportion of transduced hepatocytes. All macaques developed serotype-specific antibodies but no capsid-specific cytotoxic T lymphocytes were detected. The liver was preferentially transduced with 300-fold more proviral copies than extrahepatic tissues. Long-term biochemical, ultrasound imaging, and histologic follow-up of this large cohort of NHP revealed no toxicity. These data support further evaluation of this vector in hemophilia B patients.


Subject(s)
Capsid Proteins/metabolism , Dependovirus/genetics , Factor IX/metabolism , Genetic Therapy/methods , Hemophilia B/therapy , Animals , Capsid Proteins/genetics , Factor IX/genetics , Gene Expression , Genetic Vectors , HEK293 Cells , Hemophilia B/genetics , Humans , In Situ Hybridization, Fluorescence , Liver/metabolism , Macaca , Mice
20.
Blood ; 117(10): 2817-26, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21156846

ABSTRACT

ß-Thalassemia major results from severely reduced or absent expression of the ß-chain of adult hemoglobin (α2ß2;HbA). Increased levels of fetal hemoglobin (α2γ2;HbF), such as occurs with hereditary persistence of HbF, ameliorate the severity of ß-thalassemia, raising the potential for genetic therapy directed at enhancing HbF. We used an in vitro model of human erythropoiesis to assay for enhanced production of HbF after gene delivery into CD34(+) cells obtained from mobilized peripheral blood of normal adults or steady-state bone marrow from patients with ß-thalassemia major. Lentiviral vectors encoding (1) a human γ-globin gene with or without an insulator, (2) a synthetic zinc-finger transcription factor designed to interact with the γ-globin gene promoters, or (3) a short-hairpin RNA targeting the γ-globin gene repressor, BCL11A, were tested. Erythroid progeny of normal CD34(+) cells demonstrated levels of HbF up to 21% per vector copy. For ß-thalassemic CD34(+) cells, similar gene transfer efficiencies achieved HbF production ranging from 45% to 60%, resulting in up to a 3-fold increase in the total cellular Hb content. These observations suggest that both lentiviral-mediated γ-globin gene addition and genetic reactivation of endogenous γ-globin genes have potential to provide therapeutic HbF levels to patients with ß-globin deficiency.


Subject(s)
Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Gene Transfer Techniques , Genetic Therapy , beta-Thalassemia/therapy , gamma-Globins/genetics , Antigens, CD34/metabolism , Blotting, Southern , Blotting, Western , Cell Separation , Erythropoiesis/physiology , Fetal Hemoglobin/genetics , Flow Cytometry , Genetic Vectors , Humans , Lentivirus/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...