Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Blood ; 141(9): 1007-1022, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36332160

ABSTRACT

X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene, resulting in the inability of phagocytic cells to eliminate infections. To design a lentiviral vector (LV) capable of recapitulating the endogenous regulation and expression of CYBB, a bioinformatics-guided approach was used to elucidate the cognate enhancer elements regulating the native CYBB gene. Using this approach, we analyzed a 600-kilobase topologically associated domain of the CYBB gene and identified endogenous enhancer elements to supplement the CYBB promoter to develop MyeloVec, a physiologically regulated LV for the treatment of X-CGD. When compared with an LV currently in clinical trials for X-CGD, MyeloVec showed improved expression, superior gene transfer to hematopoietic stem and progenitor cells (HSPCs), corrected an X-CGD mouse model leading to complete protection against Burkholderia cepacia infection, and restored healthy donor levels of antimicrobial oxidase activity in neutrophils derived from HSPCs from patients with X-CGD. Our findings validate the bioinformatics-guided design approach and have yielded a novel LV with clinical promise for the treatment of X-CGD.


Subject(s)
Granulomatous Disease, Chronic , Animals , Mice , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/therapy , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NADPH Oxidase 2/genetics , Genetic Therapy/methods , Mutation
2.
N Engl J Med ; 384(21): 2002-2013, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33974366

ABSTRACT

BACKGROUND: Severe combined immunodeficiency due to adenosine deaminase (ADA) deficiency (ADA-SCID) is a rare and life-threatening primary immunodeficiency. METHODS: We treated 50 patients with ADA-SCID (30 in the United States and 20 in the United Kingdom) with an investigational gene therapy composed of autologous CD34+ hematopoietic stem and progenitor cells (HSPCs) transduced ex vivo with a self-inactivating lentiviral vector encoding human ADA. Data from the two U.S. studies (in which fresh and cryopreserved formulations were used) at 24 months of follow-up were analyzed alongside data from the U.K. study (in which a fresh formulation was used) at 36 months of follow-up. RESULTS: Overall survival was 100% in all studies up to 24 and 36 months. Event-free survival (in the absence of reinitiation of enzyme-replacement therapy or rescue allogeneic hematopoietic stem-cell transplantation) was 97% (U.S. studies) and 100% (U.K. study) at 12 months; 97% and 95%, respectively, at 24 months; and 95% (U.K. study) at 36 months. Engraftment of genetically modified HSPCs persisted in 29 of 30 patients in the U.S. studies and in 19 of 20 patients in the U.K. study. Patients had sustained metabolic detoxification and normalization of ADA activity levels. Immune reconstitution was robust, with 90% of the patients in the U.S. studies and 100% of those in the U.K. study discontinuing immunoglobulin-replacement therapy by 24 months and 36 months, respectively. No evidence of monoclonal expansion, leukoproliferative complications, or emergence of replication-competent lentivirus was noted, and no events of autoimmunity or graft-versus-host disease occurred. Most adverse events were of low grade. CONCLUSIONS: Treatment of ADA-SCID with ex vivo lentiviral HSPC gene therapy resulted in high overall and event-free survival with sustained ADA expression, metabolic correction, and functional immune reconstitution. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01852071, NCT02999984, and NCT01380990.).


Subject(s)
Agammaglobulinemia/therapy , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Lentivirus/genetics , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adolescent , Child , Child, Preschool , Genetic Therapy/adverse effects , Humans , Infant , Lymphocyte Count , Progression-Free Survival , Prospective Studies , Transplantation, Autologous
3.
Blood ; 138(15): 1304-1316, 2021 10 14.
Article in English | MEDLINE | ID: mdl-33974038

ABSTRACT

Patients lacking functional adenosine deaminase activity have severe combined immunodeficiency (ADA SCID), which can be treated with ADA enzyme replacement therapy (ERT), allogeneic hematopoietic stem cell transplantation (HSCT), or autologous HSCT with gene-corrected cells (gene therapy [GT]). A cohort of 10 ADA SCID patients, aged 3 months to 15 years, underwent GT in a phase 2 clinical trial between 2009 and 2012. Autologous bone marrow CD34+ cells were transduced ex vivo with the MND (myeloproliferative sarcoma virus, negative control region deleted, dl587rev primer binding site)-ADA gammaretroviral vector (gRV) and infused following busulfan reduced-intensity conditioning. These patients were monitored in a long-term follow-up protocol over 8 to 11 years. Nine of 10 patients have sufficient immune reconstitution to protect against serious infections and have not needed to resume ERT or proceed to secondary allogeneic HSCT. ERT was restarted 6 months after GT in the oldest patient who had no evidence of benefit from GT. Four of 9 evaluable patients with the highest gene marking and B-cell numbers remain off immunoglobulin replacement therapy and responded to vaccines. There were broad ranges of responses in normalization of ADA enzyme activity and adenine metabolites in blood cells and levels of cellular and humoral immune reconstitution. Outcomes were generally better in younger patients and those receiving higher doses of gene-marked CD34+ cells. No patient experienced a leukoproliferative event after GT, despite persisting prominent clones with vector integrations adjacent to proto-oncogenes. These long-term findings demonstrate enduring efficacy of GT for ADA SCID but also highlight risks of genotoxicity with gRVs. This trial was registered at www.clinicaltrials.gov as #NCT00794508.


Subject(s)
Agammaglobulinemia/therapy , Genetic Therapy , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/genetics , Adolescent , Agammaglobulinemia/genetics , Child , Child, Preschool , Follow-Up Studies , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Severe Combined Immunodeficiency/genetics , Transplantation, Autologous/methods , Treatment Outcome
4.
Mol Ther ; 28(1): 328-340, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31628051

ABSTRACT

ß-globin lentiviral vectors (ß-LV) have faced challenges in clinical translation for gene therapy of sickle cell disease (SCD) due to low titer and sub-optimal gene transfer to hematopoietic stem and progenitor cells (HSPCs). To overcome the challenge of preserving efficacious expression while increasing vector performance, we used published genomic and epigenomic data available through ENCODE to redefine enhancer element boundaries of the ß-globin locus control region (LCR) to construct novel ENCODE core sequences. These novel LCR elements were used to design a ß-LV of reduced proviral length, termed CoreGA-AS3-FB, produced at higher titers and possessing superior gene transfer to HSPCs when compared to the full-length parental ß-LV at equal MOI. At low vector copy number, vectors containing the ENCODE core sequences were capable of reversing the sickle phenotype in a mouse model of SCD. These studies provide a ß-LV that will be beneficial for gene therapy of SCD by significantly reducing the cost of vector production and extending the vector supply.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Locus Control Region/genetics , Transduction, Genetic/methods , beta-Globins/genetics , Animals , Bone Marrow Cells/metabolism , Disease Models, Animal , HEK293 Cells , Healthy Volunteers , Hematopoietic Stem Cells/metabolism , Humans , Mice , Phenotype , Transfection
5.
Stem Cells ; 37(2): 284-294, 2019 02.
Article in English | MEDLINE | ID: mdl-30372555

ABSTRACT

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated system (Cas9)-mediated gene editing of human hematopoietic stem cells (hHSCs) is a promising strategy for the treatment of genetic blood diseases through site-specific correction of identified causal mutations. However, clinical translation is hindered by low ratio of precise gene modification using the corrective donor template (homology-directed repair, HDR) to gene disruption (nonhomologous end joining, NHEJ) in hHSCs. By using a modified version of Cas9 with reduced nuclease activity in G1 phase of cell cycle when HDR cannot occur, and transiently increasing the proportion of cells in HDR-preferred phases (S/G2), we achieved a four-fold improvement in HDR/NHEJ ratio over the control condition in vitro, and a significant improvement after xenotransplantation of edited hHSCs into immunodeficient mice. This strategy for improving gene editing outcomes in hHSCs has important implications for the field of gene therapy, and can be applied to diseases where increased HDR/NHEJ ratio is critical for therapeutic success. Stem Cells 2019;37:284-294.


Subject(s)
DNA Repair/genetics , Gene Editing/methods , Hematopoietic Stem Cell Transplantation/methods , Stem Cells/metabolism , Transplantation Conditioning/methods , Animals , Humans , Mice
6.
Mol Ther ; 27(8): 1389-1406, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31178391

ABSTRACT

Site-specific correction of a point mutation causing a monogenic disease in autologous hematopoietic stem and progenitor cells (HSPCs) can be used as a treatment of inherited disorders of the blood cells. Sickle cell disease (SCD) is an ideal model to investigate the potential use of gene editing to transvert a single point mutation at the ß-globin locus (HBB). We compared the activity of zinc-finger nucleases (ZFNs) and CRISPR/Cas9 for editing, and homologous donor templates delivered as single-stranded oligodeoxynucleotides (ssODNs), adeno-associated virus serotype 6 (AAV6), integrase-deficient lentiviral vectors (IDLVs), and adenovirus 5/35 serotype (Ad5/35) to transvert the base pair responsible for SCD in HBB in primary human CD34+ HSPCs. We found that the ZFNs and Cas9 directed similar frequencies of nuclease activity. In vitro, AAV6 led to the highest frequencies of homology-directed repair (HDR), but levels of base pair transversions were significantly reduced when analyzing cells in vivo in immunodeficient mouse xenografts, with similar frequencies achieved with either AAV6 or ssODNs. AAV6 also caused significant impairment of colony-forming progenitors and human cell engraftment. Gene correction in engrafting hematopoietic stem cells may be limited by the capacity of the cells to mediate HDR, suggesting additional manipulations may be needed for high-efficiency gene correction in HSPCs.


Subject(s)
Anemia, Sickle Cell/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , Mutation , beta-Globins/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/therapy , CRISPR-Cas Systems , Dependovirus , Endonucleases/genetics , Gene Expression , Gene Targeting , Genetic Therapy , Genetic Vectors/genetics , Humans , Parvovirinae/genetics , Tissue Donors , Transduction, Genetic , Zinc Finger Nucleases/genetics
7.
Mol Ther ; 26(2): 468-479, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29221806

ABSTRACT

The use of engineered nucleases combined with a homologous DNA donor template can result in targeted gene correction of the sickle cell disease mutation in hematopoietic stem and progenitor cells. However, because of the high homology between the adjacent human ß- and δ-globin genes, off-target cleavage is observed at δ-globin when using some endonucleases targeted to the sickle mutation in ß-globin. Introduction of multiple double-stranded breaks by endonucleases has the potential to induce intergenic alterations. Using a novel droplet digital PCR assay and high-throughput sequencing, we characterized the frequency of rearrangements between the ß- and δ-globin paralogs when delivering these nucleases. Pooled CD34+ cells and colony-forming units from sickle bone marrow were treated with nuclease only or including a donor template and then analyzed for potential gene rearrangements. It was observed that, in pooled CD34+ cells and colony-forming units, the intergenic ß-δ-globin deletion was the most frequent rearrangement, followed by inversion of the intergenic fragment, with the inter-chromosomal translocation as the least frequent. No rearrangements were observed when endonuclease activity was restricted to on-target ß-globin cleavage. These findings demonstrate the need to develop site-specific endonucleases with high specificity to avoid unwanted gene alterations.


Subject(s)
Gene Editing , Genetic Variation , Hematopoietic Stem Cells/metabolism , beta-Globins/genetics , Gene Conversion , Gene Rearrangement , Gene Targeting , High-Throughput Nucleotide Sequencing , Humans , Nucleic Acid Amplification Techniques , Translocation, Genetic
8.
Mol Ther ; 25(9): 2163-2175, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28663101

ABSTRACT

Lentiviral vector (LV)-based hematopoietic stem cell (HSC) gene therapy is becoming a promising clinical strategy for the treatment of genetic blood diseases. However, the current approach of modifying 1 × 108 to 1 × 109 CD34+ cells per patient requires large amounts of LV, which is expensive and technically challenging to produce at clinical scale. Modification of bulk CD34+ cells uses LV inefficiently, because the majority of CD34+ cells are short-term progenitors with a limited post-transplant lifespan. Here, we utilized a clinically relevant, immunomagnetic bead (IB)-based method to purify CD34+CD38- cells from human bone marrow (BM) and mobilized peripheral blood (mPB). IB purification of CD34+CD38- cells enriched severe combined immune deficiency (SCID) repopulating cell (SRC) frequency an additional 12-fold beyond standard CD34+ purification and did not affect gene marking of long-term HSCs. Transplant of purified CD34+CD38- cells led to delayed myeloid reconstitution, which could be rescued by the addition of non-transduced CD38+ cells. Importantly, LV modification and transplantation of IB-purified CD34+CD38- cells/non-modified CD38+ cells into immune-deficient mice achieved long-term gene-marked engraftment comparable with modification of bulk CD34+ cells, while utilizing ∼7-fold less LV. Thus, we demonstrate a translatable method to improve the clinical and commercial viability of gene therapy for genetic blood cell diseases.


Subject(s)
Hematopoietic Stem Cells/metabolism , Transduction, Genetic , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD34/metabolism , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Genetic Therapy , Genetic Vectors/genetics , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Immunomagnetic Separation , Immunophenotyping , Lentivirus/genetics , Mice , Transgenes
9.
Blood ; 125(17): 2597-604, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25733580

ABSTRACT

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the ß-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the ß-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Subject(s)
Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Mutation , beta-Globins/genetics , Anemia, Sickle Cell/pathology , Animals , Antigens, CD34/analysis , Base Sequence , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Endodeoxyribonucleases/metabolism , Fetal Blood/transplantation , Genetic Loci , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Zinc Fingers
10.
Cytotherapy ; 19(9): 1096-1112, 2017 09.
Article in English | MEDLINE | ID: mdl-28733131

ABSTRACT

BACKGROUND AIMS: Gene therapy by autologous hematopoietic stem cell transplantation (HSCT) represents a new approach to treat sickle cell disease (SCD). Optimization of the manufacture, characterization and testing of the transduced hematopoietic stem cell final cell product (FCP), as well as an in depth in vivo toxicology study, are critical for advancing this approach to clinical trials. METHODS: Data are shown to evaluate and establish the feasibility of isolating, transducing with the Lenti/ßAS3-FB vector and cryopreserving CD34+ cells from human bone marrow (BM) at clinical scale. In vitro and in vivo characterization of the FCP was performed, showing that all the release criteria were successfully met. In vivo toxicology studies were conducted to evaluate potential toxicity of the Lenti/ßAS3-FB LV in the context of a murine BM transplant. RESULTS: Primary and secondary transplantation did not reveal any toxicity from the lentiviral vector. Additionally, vector integration site analysis of murine and human BM cells did not show any clonal skewing caused by insertion of the Lenti/ßAS3-FB vector in cells from primary and secondary transplanted mice. CONCLUSIONS: We present here a complete protocol, thoroughly optimized to manufacture, characterize and establish safety of a FCP for gene therapy of SCD.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells , Adult , Animals , Antigens, CD34/metabolism , Bone Marrow Cells , Bone Marrow Transplantation , Case-Control Studies , Clinical Trials, Phase I as Topic , Genetic Vectors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Lentivirus/genetics , Mice, Inbred NOD , Transduction, Genetic , Transplantation, Autologous/methods
11.
Mol Ther ; 24(9): 1561-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27406980

ABSTRACT

Targeted genome editing technology can correct the sickle cell disease mutation of the ß-globin gene in hematopoietic stem cells. This correction supports production of red blood cells that synthesize normal hemoglobin proteins. Here, we demonstrate that Transcription Activator-Like Effector Nucleases (TALENs) and the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system can target DNA sequences around the sickle-cell mutation in the ß-globin gene for site-specific cleavage and facilitate precise correction when a homologous donor template is codelivered. Several pairs of TALENs and multiple CRISPR guide RNAs were evaluated for both on-target and off-target cleavage rates. Delivery of the CRISPR/Cas9 components to CD34+ cells led to over 18% gene modification in vitro. Additionally, we demonstrate the correction of the sickle cell disease mutation in bone marrow derived CD34+ hematopoietic stem and progenitor cells from sickle cell disease patients, leading to the production of wild-type hemoglobin. These results demonstrate correction of the sickle mutation in patient-derived CD34+ cells using CRISPR/Cas9 technology.


Subject(s)
Anemia, Sickle Cell/genetics , CRISPR-Cas Systems , Gene Editing , Hematopoietic Stem Cells/metabolism , Mutation , Targeted Gene Repair , beta-Globins/genetics , Anemia, Sickle Cell/therapy , Base Sequence , Cell Line , DNA Cleavage , Gene Targeting , Genetic Loci , Humans , Protein Binding , RNA, Guide, Kinetoplastida , Transcription Activator-Like Effector Nucleases/metabolism
12.
Stem Cells ; 33(5): 1532-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25588820

ABSTRACT

Autologous hematopoietic stem cell (HSC) gene therapy for sickle cell disease has the potential to treat this illness without the major immunological complications associated with allogeneic transplantation. However, transduction efficiency by ß-globin lentiviral vectors using CD34-enriched cell populations is suboptimal and large vector production batches may be needed for clinical trials. Transducing a cell population more enriched for HSC could greatly reduce vector needs and, potentially, increase transduction efficiency. CD34(+) /CD38(-) cells, comprising ∼1%-3% of all CD34(+) cells, were isolated from healthy cord blood CD34(+) cells by fluorescence-activated cell sorting and transduced with a lentiviral vector expressing an antisickling form of beta-globin (CCL-ß(AS3) -FB). Isolated CD34(+) /CD38(-) cells were able to generate progeny over an extended period of long-term culture (LTC) compared to the CD34(+) cells and required up to 40-fold less vector for transduction compared to bulk CD34(+) preparations containing an equivalent number of CD34(+) /CD38(-) cells. Transduction of isolated CD34(+) /CD38(-) cells was comparable to CD34(+) cells measured by quantitative PCR at day 14 with reduced vector needs, and average vector copy/cell remained higher over time for LTC initiated from CD34(+) /38(-) cells. Following in vitro erythroid differentiation, HBBAS3 mRNA expression was similar in cultures derived from CD34(+) /CD38(-) cells or unfractionated CD34(+) cells. In vivo studies showed equivalent engraftment of transduced CD34(+) /CD38(-) cells when transplanted in competition with 100-fold more CD34(+) /CD38(+) cells. This work provides initial evidence for the beneficial effects from isolating human CD34(+) /CD38(-) cells to use significantly less vector and potentially improve transduction for HSC gene therapy.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Transduction, Genetic , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Cell Separation , Erythroid Cells/cytology , Genetic Vectors/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Mice, Inbred NOD , Receptors, LDL/metabolism
13.
Stem Cells ; 32(6): 1503-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24677652

ABSTRACT

Unlimited self renewal capacity and differentiation potential make human pluripotent stem cells (PSC) a promising source for the ex vivo manufacture of red blood cells (RBCs) for safe transfusion. Current methods to induce erythropoiesis from PSC suffer from low yields of RBCs, most of which are immature and contain embryonic and fetal rather than adult hemoglobins. We have previously shown that homodimerization of the intracellular component of MPL (ic-MPL) induces erythropoiesis from human cord blood progenitors. The goal of this study was to investigate the potential of ic-MPL dimerization to induce erythropoiesis from human embryonic stem cells (hESCs) and to identify the signaling pathways activated by this strategy. We present here the evidence that ic-MPL dimerization induces erythropoietin (EPO)-independent erythroid differentiation from hESC by inducing the generation of erythroid progenitors and by promoting more efficient erythroid maturation with increased RBC enucleation as well as increased gamma:epsilon globin ratio and production of beta-globin protein. ic-MPL dimerization is significantly more potent than EPO in inducing erythropoiesis, and its effect is additive to EPO. Signaling studies show that dimerization of ic-MPL, unlike stimulation of the wild type MPL receptor, activates AKT in the absence of JAK2/STAT5 signaling. AKT activation upregulates GATA-1 and FOXO3 transcriptional pathways with resulting inhibition of apoptosis, modulation of cell cycle, and enhanced maturation of erythroid cells. These findings open up potential new targets for the generation of therapeutically relevant RBC products from hPSC.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Erythropoiesis , Erythropoietin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Protein Multimerization , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism
14.
Mol Ther ; 22(3): 607-622, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24256635

ABSTRACT

Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA(-/-) mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA(-/-) mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34(+) cells transduced with 1-5 × 10(7) TU/ml had 1-3 vector copies/cell and expressed 1-2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Agammaglobulinemia/immunology , Agammaglobulinemia/therapy , Genetic Vectors/adverse effects , Lentivirus/genetics , Peptide Elongation Factor 1/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/immunology , Adenosine Deaminase/metabolism , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Animals , B-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Female , HEK293 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , Transduction, Genetic , Virus Integration
15.
Mol Ther ; 21(9): 1705-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23857176

ABSTRACT

We investigated the use of integrase-defective lentiviral vectors (IDLVs) for transient delivery of zinc finger nucleases (ZFNs) and donor templates for site-specific modification of the human adenosine deaminase (hADA) gene. Initially, we constructed IDLVs carrying ZFN monomers (Single-IDLVs) and found them to be able to deliver their gene-editing payload to K562 cells successfully upon cotransduction, with minimal cytotoxicity. To simplify delivery, we designed an IDLV construct to deliver both ZFN monomers from the same vector (Double-IDLV). However, this construct in its original state was prone to rearrangements of the vector genome, resulting in greatly reduced functionality; this was due to recombination between highly similar ZFN monomers arranged in tandem. We modified the Double-IDLV constructs to reduce recombination and restored simultaneous delivery of both ZFNs. We also tested an IDLV construct for delivery of donor templates and demonstrated its efficacy for gene modification. In summary, we highlighted the importance of modifying vector design for co-delivery of highly similar sequences inherent to genome-editing nucleases, and demonstrated significant improvement in the use of IDLVs for delivery of ZFNs and donor templates for genome modification.


Subject(s)
Adenosine Deaminase/genetics , Endonucleases/metabolism , Genetic Vectors , Integrases/genetics , Lentivirus/genetics , Transduction, Genetic , Virus Integration , Endonucleases/genetics , Genetic Loci , Humans , K562 Cells , Lentivirus/metabolism , Zinc Fingers
16.
Mol Ther Methods Clin Dev ; 32(2): 101254, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38745893

ABSTRACT

A major limitation of gene therapy for sickle cell disease (SCD) is the availability and access to a potentially curative one-time treatment, due to high treatment costs. We have developed a high-titer bifunctional lentiviral vector (LVV) in a vector backbone that has reduced size, high vector yields, and efficient gene transfer to human CD34+ hematopoietic stem and progenitor cells (HSPCs). This LVV contains locus control region cores expressing an anti-sickling ßAS3-globin gene and two microRNA-adapted short hairpin RNA simultaneously targeting BCL11A and ZNF410 transcripts to maximally induce fetal hemoglobin (HbF) expression. This LVV induces high levels of anti-sickling hemoglobins (HbAAS3 + HbF), while concurrently decreasing sickle hemoglobin (HbS). The decrease in HbS and increased anti-sickling hemoglobin impedes deoxygenated HbS polymerization and red blood cell sickling at low vector copy per cell in transduced SCD patient CD34+ cells differentiated into erythrocytes. The dual alterations in red cell hemoglobins ameliorated the SCD phenotype in the SCD Berkeley mouse model in vivo. With high titer and enhanced transduction of HSPC at a low multiplicity of infection, this LVV will increase the number of patient doses of vector from production lots to decrease costs and help improve accessibility to gene therapy for SCD.

17.
J Transl Med ; 11: 23, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23360526

ABSTRACT

BACKGROUND: Chimeric Antigen Receptors (CARs) consist of the antigen-recognition portion of a monoclonal antibody fused to an intracellular signaling domain capable of activating T-cells. CARs displayed on the surface of transduced cells perform non-MHC-restricted antigen recognition and activating intracellular signaling pathways for induction of target cytolysis, cytokine secretion and proliferation. Clinical trials are in progress assessing the use of mature T-lymphocytes transduced with CARs targeting CD19 antigen to treat B-lineage malignancies. CD19 is an attractive target for immunotherapy because of its consistent and specific expression in most of the stages of maturation and malignancies of B-lymphocyte origin, but not on hematopoietic stem cells. Antibodies against the extracellular domain of the CAR molecule (anti-Fab, Fc or idiotype) have been used for detection of CAR expression in research and clinical samples by flow cytometry, but may need development for each construct and present significant background in samples from xenograft models. METHODS: A specific reagent for the detection of anti-CD19 CAR expression was developed, a fusion protein consisting of human CD19 extracellular domains and the Fc region of human IgG1 (CD19sIg). Genes encoding CD19sIg fusion proteins were constructed by fusing either exons 1 to 3 (CD19sIg1-3) or exons 1 to 4 (CD19sIg1-4) of the human CD19 cDNA to a human IgG1Fc fragment. These fusion proteins are intended to work in similar fashion as the MHC Tetramers used for identification of antigen-specific T-cells, and may also have other applications in studies of activation of anti-CD19 CAR bearing cells. The CD19sIg proteins were produced from 293 T cells by stable lentiviral vector transduction and purification from culture medium. RESULTS: ELISA assays using several different monoclonal antibodies to CD19 demonstrated dose-related specific binding by the fusion molecule CD19sIg1-4, but no binding by CD19sIg1-3. Conjugation of the CD19sIg1-4 fusion protein to Alexa Fluor 488 allowed specific and sensitive staining of anti-CD19 CAR-bearing cells for flow cytometry assays, detecting as low as 0.5% of CAR-modified primary cells with minimal background staining. CONCLUSIONS: This fusion molecule is a sensitive reagent for detection of anti-CD19 CAR derived from any monoclonal antibody present in CAR-modified T-cells.


Subject(s)
Antigens, CD19/chemistry , Immunoglobulin Fragments/chemistry , Receptors, Antigen/chemistry , Recombinant Fusion Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Cell Proliferation , Cell Transplantation , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/cytology
19.
Mol Ther Oncolytics ; 23: 582-592, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34938858

ABSTRACT

Lentiviral vectors (LVs) are robust delivery vehicles for gene therapy as they can efficiently integrate transgenes into host cell genomes. However, LVs with lengthy or complex expression cassettes typically are produced at low titers and have reduced gene transfer capacity, creating barriers for clinical and commercial applications. Modifications of the packaging cell line and methods may be able to produce complex vectors at higher titer and infectivity and may improve production of many different LVs. In this study, we identified two host restriction factors in HEK293T packaging cells that impeded LV production, 2'-5'-oligoadenylate synthetase 1 (OAS1) and low-density lipoprotein receptor (LDLR). Knocking out these two genes separately led to ∼2-fold increases in viral titer. We created a monoclonal cell line, CRISPRed HEK293T to Disrupt Antiviral Response (CHEDAR), by successively knocking out OAS1, LDLR, and PKR, a previously identified factor impeding LV titers. Packaging in CHEDAR yielded ∼7-fold increases in physical particles, full-length vector RNA, and vector titers. In addition, overexpressing transcription elongation factors, SPT4 and SPT5, during packaging improved the production of full-length vector RNA, thereby increasing titers by ∼2-fold. Packaging in CHEDAR with over-expression of SPT4 and SPT5 led to ∼11-fold increases of titers. These approaches improved the production of a variety of LVs, especially vectors with low titers or with internal promoters in the reverse orientation, and may be beneficial for multiple gene therapy applications.

20.
Stem Cell Reports ; 16(1): 198-211, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33186538

ABSTRACT

Lentiviral vectors (LVs) commonly used for the treatment of hemoglobinopathies often have low titers and sub-optimal gene transfer efficiency for human hematopoietic stem and progenitor cells (HSPCs), hindering clinical translation and commercialization for ex vivo gene therapy. We observed that a high percentage of ß-globin LV viral genomic RNAs were incomplete toward the 3' end in packaging cells and in released vector particles. The incomplete vector genomes impeded reverse transcription in target cells, limiting stable gene transfer to HSPCs. By combining three modifications to vector design and production (shortening the vector length to 5.3 kb; expressing HIV-1 Tat protein during packaging; and packaging in PKR-/- cells) there was a 30-fold increase in vector titer and a 3-fold increase in vector infectivity in HSPCs. These approaches may improve the manufacturing of ß-globin and other complex LVs for enhanced gene delivery and may facilitate clinical applications.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/genetics , RNA/metabolism , Virion/physiology , beta-Globins/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , beta-Globins/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL