WO2008136670A2 - Procédés et moyens améliorés pour une distribution de gène lentiviral - Google Patents

Procédés et moyens améliorés pour une distribution de gène lentiviral Download PDF

Info

Publication number
WO2008136670A2
WO2008136670A2 PCT/NL2008/050269 NL2008050269W WO2008136670A2 WO 2008136670 A2 WO2008136670 A2 WO 2008136670A2 NL 2008050269 W NL2008050269 W NL 2008050269W WO 2008136670 A2 WO2008136670 A2 WO 2008136670A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
stem cells
gene delivery
hematopoietic stem
delivery vehicle
Prior art date
Application number
PCT/NL2008/050269
Other languages
English (en)
Other versions
WO2008136670A3 (fr
Inventor
Nico Peter Van Til
Monique Maria Andrea Verstegen
Gerard Wagemaker
Original Assignee
Erasmus University Medical Center Rotterdam
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/NL2007/050193 external-priority patent/WO2008136656A1/fr
Application filed by Erasmus University Medical Center Rotterdam filed Critical Erasmus University Medical Center Rotterdam
Publication of WO2008136670A2 publication Critical patent/WO2008136670A2/fr
Publication of WO2008136670A3 publication Critical patent/WO2008136670A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the invention relates to the field of gene therapy and more in specific to lentiviral gene delivery vehicles and methods for efficient transduction of lentiviral gene delivery vehicles into hematopoietic stem cells and their descendants.
  • the invention provides in one of its embodiments a method of gene transfer into e.g. pluripotent hematopoietic stem cells and their descendants, enabling successful transduction of cells, including transplantable cell populations comprising hematopoietic stem cells that give rise to progeny expressing the transduced gene(s).
  • the invention further comprises a method for treating a variety of hereditary and acquired human disease by transfer of a therapeutically active gene into hematopoietic stem cells.
  • the invention shows that symptoms associated with Pompe disease are (completely) reduced and/or alleviated by treatment of a subject suffering from Pompe disease with hematopoietic stem cell transduced with an alpha-glucosidase comprising lentiviral vector.
  • HSC gamma retrovirus vector-mediated ex ⁇ i ⁇ o hematopoietic stem cell
  • ADA-SCID X-linked severe combined immunodeficiency SCID and adenosine deaminase deficiency
  • efficacy also revealed potential genotoxicity, essentially due to insertional transactivation of neighbouring genes by retroviral enhancer elements, resulting at a very low frequency in aberrant expression of neighboring genes and thereby in clonal dominance/leukemogenesis.
  • Vector systems with a reduced risk profile are therefore essential in the further development of gene therapy for inherited diseases.
  • HIV-I derived self- inactivating lentiviral vectors display a reduced risk pattern due to deletion of enhancer regions and a more favourable integration pattern as compared to gamma-retroviruses.
  • MOI multiplicity of infection
  • TU transducing units
  • the aim of the current invention is to improve lentiviral vector transduction efficiency of HSC and preferably to limit the number of integrations per cell.
  • VSV-G packaged LV expressing EGFP The inventors observed that retronectin and growth factor supported overnight transduction of lineage depleted (lin ⁇ ) mouse bone marrow cells (BMC) with low cell-density, but a high MOI of 30 yielded approximately 60% EGFP + cells.
  • the transduction efficiency of lirr 7 - cells could be markedly improved from 7% to 54% by increasing the cell density from 2 x 10 4 /ml to 6 x 10 5 /ml with a proportional increase of TU/ml.
  • Mouse lin '- BMCs transduced with LV at MOI 2 resulted in 98% EGFP + cells, with sustained reconstitution of up to 80% EGFP + blood cells after testing the gene-modified cells in a competitive repopulation assay in vivo.
  • the inventors of the current invention show that an increase in cell density of target cells with a fixed ratio of transducing units (TU) per cell (or MOI) improved vector transduction efficiency of HSC.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle.
  • this controlled transduction method results in the presence of essentially one therapeutic gene integration per target cell, which has been impossible with other lentiviral transduction methods.
  • another desirable effect of the method according to the invention is that only limited amount of therapeutic gene vector batches need to be produced, thus saving considerable time in respect of quality control and production time.
  • transduction refers to the stable transfer of genetic material from a viral particle to a hematopoietic stem cell genome.
  • the hematopoietic system produces perpetually large numbers of blood cells, which have a limited life span and need to be constantly renewed throughout the life of a mammal. This renewal is maintained through proliferation and differentiation of a small number of hematopoietic stem cells in the bone marrow.
  • the definition of stem cells is not always clear within the art.
  • a functional definition is used, which defines hematopoietic stem cells as those cells capable of (long term) reconstitution of a hematopoietic system. This definition is often felt to include at least some early progenitor cells.
  • hematopoietic stem cells are a highly suitable target for gene therapy for a variety of hereditary and acquired diseases within and outside the hematopoietic system.
  • lentiviral mediated gene transfer has met with only limited success due to the difficulty of obtaining sufficient numbers of successfully transduced, transplantable, single-copy of therapeutic gene containing, long-term repopulating hematopoietic stem cells.
  • HSC are stem cells and the early precursor cells which give rise to all the blood cell types that include both the myeloid (monocytes and macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets and some dendritic cells) and lymphoid lineages (T-cells, B-cells, NK-cells, some dendritic cells).
  • myeloid monocytes and macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets and some dendritic cells
  • T-cells, B-cells, NK-cells, some dendritic cells lymphoid lineages
  • the definition of HSC has undergone considerable revision in the last two decades.
  • the hematopoietic tissue have cells with long term and short term regeneration capacities and committed multipotent, oligopotent and unipotent progenitors.
  • HSC can be obtained from different sources and are, for example, found in the bone marrow of adults, which includes femurs, hip, ribs, sternum, and other bones.
  • Cells can be obtained directly by removal from the hip using a needle and syringe, or from the blood following pre-treatment with cytokines, such as G-CSF (granulocyte colony stimulating factor), that induces cells to be released from the bone marrow compartment into the blood (mobilized peripheral blood).
  • cytokines such as G-CSF (granulocyte colony stimulating factor)
  • Other sources for clinical and scientific use include umbilical cord blood, and placenta.
  • fetal liver, fetal spleen and AGM (Aorta-gonad-mesonephros) of animals are also useful sources of HSCs.
  • HSC are phenotypically identified by their small size, lack of lineage (lin) markers, low staining (side population) with vital dyes such, as rhodamine 123 (rhodamine DULL , also called rho 10 ) or Hoechst 33342, and presence of various antigenic markers on their surface many of which belongs to the cluster of differentiation series, such as: CD34, CD38, CD90, CD133, CD105, CD45 and also c-kit- the receptor for stem cell factor.
  • The_hematopoietic stem cells are negative for the markers which are used for detection of lineage commitment and are thus called Lin-, and during their purification by FACS, a bunch of up to 13 to 14 different mature blood-lineage marker e.g.
  • CD13 and CD33 for myeloid, CD71 for erythroid, CD 19 for B cells, CD61 for megakaryocyte etc for humans; and, B220 (murine CD45) for B cells, Mac-1 (CDllb/CD18) for monocytes, Gr-I for Granulocytes, Terll9 for erythroid cells, 117Ra, CD3, CD4, CD5, CD8 for T cells etc for mice) antibodies are used as a mixture to deplete the lin+ cells or late multipotent progenitors (MPP)s.
  • MPP multipotent progenitors
  • the VSV-g pseudotyped lentiviral vector described in the experimental part can in principle transduce all kinds of cell types because the VSV-g provides the lentiviral particles with a broad tropism.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said HSC are murine, human or primate cells. More preferred said HSC are bone marrow cells, umbilical cord blood cells or mobilized peripheral blood stem cells. Even more preferred said HSC are CD34 positive (CD34 + ) cells or CD34 positive / CD38 dull cells.
  • HSC hematopoietic stem cells
  • Gene delivery vehicles of lentiviral origin are all vehicles comprising genetic material and/or proteinaceous material derived from lentiviruses.
  • the most important features of such vehicles are the integration of their genetic material into the genome of a target cell and their capability to transduce stem cells. These elements are deemed essential in a functional manner, meaning that the sequences need not be identical to lentiviral sequences as long as the essential functions are present.
  • the methods of the invention are however especially suitable for recombinant lentiviral particles, which have most if not all of the replication and reproduction features of a lentivirus, typically in combination with a producer cell having some complementing elements. Normally the lentiviral particles making up the gene delivery vehicle are replication defective on their own.
  • a gene delivery vehicle is intended to read on any vehicle capable of delivering genetic material to a target cell, whether the genetic material is actually a gene, an antisense molecule or a cosuppressive nucleic acid (encoding molecule), etc.
  • Useful nucleic acids to be provided to target cells e.g.
  • stem cells are well known in the art and include such molecules as to replace inborn errors/deficiencies of the hematopoietic system, which may include hemoglobin genes and their regulatory elements for the thalassemia's and sickle cell anemia's and sequences to repair the various forms of severe combined immunodeficiency, such as caused by adenosine deaminase deficiency and that known as severe X linked immunodeficiency, or genes encoding enzymes for diseases known as lysosomal storage diseases, such as Hurler's, Hunter's, Krabbe's and in particular Gaucher's disease and metachromatic leukodystrophy, or by introducing sequences that confer resistance of the progeny of hematopoietic stem cells to infectious agents, such as HIV, as well as the introduction of suicide genes for cancer therapy and marker genes to track the progeny of transplanted normal and/or malignant hematopoietic stem cells.
  • hematopoietic stem cells typically use a target cell concentration of for example IxIO 5 to 4xlO 5 in combination with a relatively high multiplicity of infection (i.e., around 20 to 100).
  • a target cell concentration of for example IxIO 5 to 4xlO 5 in combination with a relatively high multiplicity of infection (i.e., around 20 to 100).
  • Another published used ratio of target cell concentration versus MOI is 5-2OxIO 4 cells versus a MOI of 100.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said amount of HSC is at least 6xlO 5 /mL.
  • HSC hematopoietic stem cells
  • This concentration of cells is especially useful for human and murine cells.
  • transduction percentages comparable to the mentioned human and murine percentage were obtained by using a cell density of 2.5 ⁇ lO 6 cells/mL. It is clear for the skilled person that a suitable target cell density depends on the kind of host used as a source, for example human, murine or primate.
  • the terms “amount of HSC” or “number of HSC” or “concentration of HSC” or “density of HSC” are used herein interchangeably.
  • the upper limit of the concentration of target cells is for example dependent on the amount of cells present in an organism used for isolation. For clinical purposes this will be approximately IxIO 8 cells. Based on the herein disclosed results an upper limit of 10 7 to 2xlO 7 should be feasible.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said amount of HSC is maximal 10 7 to 2xlO 7 cells/mL.
  • HSC hematopoietic stem cells
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said amount of HSC is at least 6xlO 5 cells/mL and maximal 10 7 to 2xlO 7 cells/mL.
  • HSC hematopoietic stem cells
  • the herein used concentration of target cells are for example obtained by isolating said target cells from a suitable source (for example bone marrow or umbilical cord or peripheral blood) and subsequently centrifuging the obtained cells to a pellet and finally resuspending the obtained pellet in a small(er) volume (when compared to the original, i.e., before centrifugation, volume).
  • a suitable source for example bone marrow or umbilical cord or peripheral blood
  • multiplicity of infection or concentration of transducing units (TU) are used interchangeable herein and refers to the number of viruses or virus particles that infect a single cell on average.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said relatively low amount of transducing units per cell is 1-10.
  • a method of the invention preferably further comprises selecting the cells that have been transduced.
  • the MOI of the used viral particles is determined on
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said relatively low amount of transducing units per cell is 1-5, more preferably 1-4, even more preferably 1-3 and most preferably 1-2.
  • HSC hematopoietic stem cells
  • producer cells Methods and means (such as producer cells) for producing the desired lentiviral particles are well known in the art.
  • preferred producer cells are 293T cells that are c ⁇ -transfected with VSV-G (Vesicular stomatitis virus- G-protein envelope).
  • VSV-G Vesicular stomatitis virus- G-protein envelope
  • Other pseudotypes used in this setting are RDl 14 (Feline-immunodeficiency virus).
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said amount of HSC is at least 6xl0 5 cells/mL and wherein said relatively low amount of transducing units per cell is 1-10.
  • HSC hematopoietic stem cells
  • the term "gene delivery vehicle of lentiviral origin” refers to a gene delivery of any lentiviral origin.
  • the used "gene delivery vehicle of lentiviral origin” is a "gene delivery vehicle of HIV lentiviral origin” and even more preferred are HIV-I derived self-inactivating lentiviral vectors.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, which is an ex vivo or in vitro method.
  • HSC hematopoietic stem cells
  • compositions obtainable by the methods of the invention.
  • compositions comprising HSC transduced with a gene delivery vehicle of lentiviral origin.
  • the invention further provides a composition comprising lentiviral particles wherein said composition has a relatively low amount of transducing units per amount of target cell.
  • said lentiviral particles are gene delivery vehicles and capable of transducing HSC and/or progenitor cells. Even more preferably said lentiviral particles are capable of transducing bone marrow cells, umbilical cord blood cells or mobilized peripheral blood stem cells.
  • multiplicity of infection or concentration of transducing units (TU) are used interchangeable herein and refers to the number of viruses or virus particles that infect a single cell on average.
  • the invention provides a composition comprising lentiviral particles wherein said composition has a relatively low amount of transducing units per amount of target cell, wherein said relatively low amount of transducing units per cell is 1-10.
  • the MOI of the used viral particles is determined on
  • the invention provides a composition comprising lentiviral particles wherein said composition has a relatively low amount of transducing units per amount of target cell, wherein said relatively low amount of transducing units per cell is 1-5, more preferably 1-4, even more preferably 1-3 and most preferably 1-2.
  • the invention also provides the pharmaceutical use of the described compositions, particularly in the treatment of diseases having a genetic component, such as the various genetic hemoglobin orders, the large group of rare diseases collectively known as severe combined immune deficiencies, the group of lysosomal storage diseases, especially with a strong hematopoietic and/or visceral expression, such as Gaucher' s disease, but also possibly Hurler's or Pompe's diseases, as well as in the treatment of infectious disease, notably HIV infection, or cancer.
  • a composition comprising lentiviral particles involves the transduction of HSC such as bone marrow cells, umbilical cord blood cells or mobilized peripheral blood stem cells (for example CD34 positive target cells).
  • Such transduced cells are typically made ex vivo and are also part of the present invention.
  • the invention provides a composition for the treatment of a hereditary disease or a pathological condition related to a genetic defect or a genetic aberration, comprising a plurality of, for example, CD34 positive cells transduced with a composition of lentiviral particles according to the invention, or a composition for the treatment of a hereditary disease or a pathological condition related to a genetic defect or a genetic aberration, comprising a plurality of, for example, CD34 positive cells, said composition being obtainable by a method according to the invention.
  • the invention provides the use of a composition as described above in the preparation of a medicament for the treatment of a hereditary disease or pathological condition related to a genetic defect or a genetic aberration.
  • Such a use is particularly useful in the treatment of diseases having a genetic component, such as the various genetic hemoglobin orders, the large group of rare diseases collectively known as severe combined immune deficiencies, the group of lysosomal storage diseases, especially with a strong hematopoietic and/or visceral expression, such as Gaucher's disease, but also possibly Hurler's or Pompe's diseases, as well as in the treatment of infectious disease, notably HIV infection, or cancer.
  • a composition comprising retroviral particles involves the transduction of, for example, CD34 positive target cells. Such transduced cells are typically made ex vivo and are also part of the present invention.
  • gene therapy is within reach for a variety of monogenic inherited rare diseases, clinical implementation requiring selection of diseases in which (1) the genetic defect is identified, (2) the diagnosis is made sufficiently early for meaningful therapeutic intervention, (3) a specific animal model is available for efficacy and safety evaluation, (4) strict regulation of transgene product levels is not required, (5) the transgene produces levels sufficient for sustained alleviation of symptoms or cure, (6) adverse immune responses to the transgene product are either not expected or do not interfere with efficacy. Lysosomal storage diseases, a group of over 50 different enzyme deficiencies, meet these requirements. The deficiencies result in intracellular deposition of storage material and loss of cell function. Clinical features display a broad spectrum of severity reflecting the extent of the enzyme deficiency.
  • allogeneic hematopoietic stem cell (HSC) transplantation is an effective treatment, even in diseases with brain involvement.
  • alloSCT requires intensive conditioning, an HLA identical donor, is dependent on endogenous enzyme levels of the hematopoietic system which may be insufficient, carries immunological risks with undue morbidity and mortality, and is applicable only to selected patients and diseases.
  • the alternative enzyme replacement (ERT) therapy is currently or shortly available for some ten of the lysosomal enzyme deficiencies, however, requires life-long administration, does not provide cure, is not effective in all patients, does not pass the blood brain barrier, and is extremely expensive.
  • the invention therefore provides the use of a composition as described above in the preparation of a medicament for the treatment of a lysosomal storage disease or a pathological condition related to a lysosomal storage disease.
  • said lysosomal storage disease is Pompe disease, i.e. in a preferred embodiment, the invention provides the use of a composition as described above in the preparation of a medicament for the treatment of Pompe disease.
  • a typical pathological condition associated with Pompe disease is an increased storage of glycogen, which causes progressive muscle weakness throughout the body. As a result thereof-cardiac as well as respiratory complications arise.
  • kits preferably a closed clinical grade device, which comprises means to purify and then transduce target cells (hematopoietic stem / progenitor cells).
  • the invention further comprises a kit comprising means to isolate and/or purify hematopoietic stem cells and a therapeutic gene delivery vehicle.
  • the other component of the described kit is a gene delivery vehicle that comprises a therapeutically interesting gene.
  • said gene delivery vehicle is of lentiviral origin.
  • the most important features of such vehicles are the integration of their genetic material into the genome of a target cell and their capability to transduce stem cells. These elements are deemed essential in a functional manner, meaning that the sequences need not be identical to lentiviral sequences as long as the essential functions are present.
  • the methods of the invention are however especially suitable for recombinant lentiviral particles, which have most if not all of the replication and reproduction features of a lentivirus, typically in combination with a producer cell having some complementing elements. Normally the lentiviral particles making up the gene delivery vehicle are replication defective on their own.
  • a gene delivery vehicle is intended to read on any vehicle capable of delivering genetic material to a target cell, whether the genetic material is actually a gene, an antisense molecule or a cosuppressive nucleic acid (encoding molecule), etc.
  • Useful nucleic acids to be provided to target cells e.g.
  • stem cells are well known in the art and include such molecules as to replace inborn errors/deficiencies of the hematopoietic system, which may include hemoglobin genes and their regulatory elements for the thalassemia's and sickle cell anemia's and sequences to repair the various forms of severe combined immunodeficiency, such as caused by adenosine deaminase deficiency and that known as severe X linked immunodeficiency, or genes encoding enzymes for diseases known as lysosomal storage diseases, such as.
  • Hurler's, Hunter's, Krabbe's and in particular Gaucher's disease and metachromatic-leukodystrophy or by introducing sequences that confer resistance of the progeny of hematopoietic stem cells to infectious agents, such as HIV, as well as the introduction of suicide genes for cancer therapy and marker genes to track the progeny of transplanted normal and/or malignant hematopoietic stem cells.
  • the invention comprises a kit comprising means to isolate and/or purify hematopoietic stem cells and a therapeutic gene delivery vehicle, further comprising instructions to transduce an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle.
  • the instructions can provide guidelines in respect of suggested reaction/transduction/culture conditions such as temperature, medium and incubation time.
  • the currently used lentiviral transduction systems are based on essentially random integration of the to be transferred nucleic acid sequences into the genomic sequence of the target cells (hematopoietic stem / progenitor cells).
  • the transduction method of the invention is not limited to this, because the method of the invention can equally well be performed in a transduction method in which the transferred nucleic acid sequence is incorporated into the genome of the target cell via homologous recombination processes.
  • Gene addition involves the delivery of corrective DNA (usually composed of the entire coding region of a gene and appropriate regulatory sequences) that compensates for or overrides the defective gene.
  • the defective gene unless it is completely absent, remains in the affected cells.
  • genome editing uses DNA repair and/or homologous recombination processes to correct an existing defective gene sequence so that the defective or mutated area of the gene is restored to a corrected normal state. Repairing the defective sequence itself maintains the corrected genetic material within its normal chromatin environment, ensuring appropriate genetic regulation and expression in the cell.
  • Genome editing may be the only suitable strategy in situations in which mutant gene product exercises a dominant negative influence over the normal gene product.
  • the invention provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively low amount of transducing units per cell of said gene delivery vehicle, wherein said gene delivery vehicle is provided with means for homologous recombination.
  • HSC hematopoietic stem cells
  • means for homologous recombination are nucleic acid sequences encoding (at least part of) a gene involved in a hereditary disease, such as X-linked SCID, sickle cell anemia, and lysosomal enzyme deficiencies.
  • the underlying mutation(s) in such hereditary disease is/are known or can easily be determined on a case-by-case basis.
  • Editing of the human genome in vi ⁇ o is somewhat hindered by the low frequency of homologous recombination. This can be circumvented by using specially engineered 'zinc-finger' nucleases as molecular scissors to cut DNA inside cells at a specific sequence. The DNA break is then patched using new genetic information. The efficiency of this process combined with the ability to design zinc-finger nucleases that target almost any DNA sequence mean that genome editing in human cells is likely to become- an important research tool and potentially a powerful way of treating disease.
  • the features of the present invention i.e. efficiency of transduction is greatly enhanced by increasing the target cell density at low multiplicity of infection, are also useful in genome editing.
  • the invention thus provides a method for transducing hematopoietic stem cells (HSC) with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells with a relatively- low amount of transducing units per cell of said gene delivery vehicle, wherein said gene delivery vehicle is provided with means for genome editing.
  • HSC hematopoietic stem cells
  • a means for genome editing is a Zinc finger endonuclease.
  • lenti ⁇ iral vectors Production of lenti ⁇ iral vectors.
  • Third generation lentiviral vector batches were produced by standard transient calcium phosphate transfection of HEK 293T cells as described before 1 ⁇ 5 , subsequently ultracentrifugated and titers were determined by end- point titration on HeLa cells.
  • the packaging plasmids pMDL-g/pRRE, pMD2-VSVg and pRSV-REV in combination with the self inactivating lentiviral transfer vectors pRRL.PPT.SF.EGFP.WPRE4*.SIN (SF-GFP), containing the HIV central polypurine tract, the spleen focus forming virus promoter or the phosphoglycerate kinase (PGK) promoter driving EGFP expression (PGK-GFP).
  • the lentiviral vector contained a modified woodchuck posttranslational regulatory element (WPRE4*) 4 containing 4 deleted ATG-sites and a large deletion of the Woodchuck hepatitis X-protein sequence.
  • Mouse Un-/ - bone marrow cells Lineage depleted (lin-/-) mouse bone marrow cells were obtained with the mouse hematopoietic progenitor (stem) cell enrichment set according to the manufacturer's protocol (BD Biosciences).
  • Murine lin-/- were cultured in our standard stem cell medium (Stem cell activating; SAF) 3 (see below) supplemented with 100 ng/ml murine stem cell factor (SCF), 10 ng/ml murine thro mbopoie tin (TPO) and 50 ng/ml human recombinant fms-like tyrosine kinase 3-ligand (Flt3L).
  • SCF murine stem cell factor
  • TPO murine thro mbopoie tin
  • Flt3L human recombinant fms-like tyrosine kinase 3-ligand
  • Low-density cells were isolated using Ficoll separation (1.077 g/cm 2 , Nycomed, Pharma, AS, Oslo, Norway) and cryopreserved until subset purification or directly continued towards purification and transduction.
  • CD34 + rhesus cells Purification of CD34 + rhesus cells was performed by positive selection using Dynalbeads (Dynal, Oslo, Norway). Briefly, low-density cells were incubated with an IgG2A antibody against CD 34 (mAb 561; from G. Gaudernack and T. Egeland, Rikshospitalet, Oslo, Norway) covalently linked to rat anti-mouse IgG2A beads. CD34 + cells devoid of the CD34-antibody were recovered using polyclonal antibodies against the Fab part of the CD34 antibody (Detachebead, Dynal). Purified rhesus CD34+ cells were analyzed by flow cytometry.
  • IgG2A antibody against CD 34 mAb 561; from G. Gaudernack and T. Egeland, Rikshospitalet, Oslo, Norway
  • CD34 + cells devoid of the CD34-antibody were recovered using polyclonal antibodies against the Fab part of the CD34 antibody (
  • Rhesus CD34+ cells were cultured in stem cell medium, supplemented with human Flt3-L (50 ng/ml, Thousand Oaks, CA, USA), rhTPO (10 ng/ml, Genentech, South San Francisco, CA, USA) and SCF (100 ng/ml).
  • UCB samples were obtained after informed consent in conformity with legal regulations in The Netherlands from placentas of full-term normal pregnancies.
  • Mononucleate d cells were isolated by Ficoll density gradient centrifugation (1.077 g/cm 2 , Nycomed .Pharma AS, Oslo, Norway), and were cryopreserved in 10% dime thy lsulphoxide, 20% heat-inactivated fetal calf serum (FCS) and 70% Hanks Balanced Salt Solution (HBSS, Gibco, Breda, The Netherlands) at -196°C. After thawing by stepwise dilution in HBSS containing 2% FCS, the cells were washed with HBSS containing 1% FCS.
  • FCS heat-inactivated fetal calf serum
  • HBSS Hanks Balanced Salt Solution
  • CD34 + cells were purified according to the manufacturer's instructions by magnetic selection using a biotinylated CD34 + antibody (clone 12.8 CellPro Inc, Bothell, USA), streptavidin MicroBeads, and MAC separation columns (Miltenyi Biotec Inc, Auburn, USA).
  • the percentage CD34 + cells in the unseparated population (unfractionated UCB) and in the purified CD34 + and CD34- fractions was determined by FACS-analysis with fluorescein isothiocyanate (FITC) conjugated antibodies against human CD34 (Becton Dickinson) for 30', on ice in HBN (HBBS, 2% (wt/vol) FCS, 0.05% (wt/vol) sodium-azide) containing 2% (vol/vol) normal human serum (NHS). The cells were washed twice and analysed by flow cytometry.
  • FITC fluorescein isothiocyanate
  • the CD34 + cells were cultured in our standardized serum free medium (SAF, stem cell activating, see below) supplemented with human recombinant growth factors fetal liver tyrosine kinase 3-ligand (Flt3-L; 50 ng/ml, Amgen, Thousand Oaks, CA, USA), thrombopoietin (TPO; 10 ng/ml, R&D Systems, Abingdon, UK) and stem cell factor (SCF; 100 ng/ml, R&D Systems, Abingdon, UK).
  • SAF serum free medium
  • Dulbecco's modified Eagle's medium (Gibco, Life Technologies Inc., Paisley, Scotland) supplemented with 2.8xlO 4 M L-alanine, 3.3xlO 4 M L-asparagine, 2.3xlO 4 M L-aspartic acid,.5.8xl ⁇ - 4 M L-cysteine, 5-lxlO 4 M L-glutamic acid, 3.5 xlO 4 M L-proline, l. ⁇ xlO" 5 M cholesterol, 4 ⁇ M cytidine, 4 ⁇ M adenosine, 4 ⁇ M uridine, 3.5 ⁇ M guanosine, 4.4 ⁇ M 2'-deoxycytidine, 4 ⁇ M
  • lentiviral vector supernatant was mixed with the enriched hematopoietic stem cells at increasing cell-density of 2xlO 4 , 6xlO 4 , 1.2xlO 5 , 2 ⁇ l0 5 or 6xlO 5 in 1 ml culture medium and these cells were subsequently seeded in retronectin (human recombinant fibronectin fragment CH-296, Takara Bio Inc) coated 24-well culture plates (surface area 2cm 2 ).
  • the multiplicity of infection (the number of viral transducing units per cell) was maintained at 1.
  • the EGFP positive fraction of the transduced cells was determined after at least 4 days after the start of transduction.
  • rhesus CD34+ cells were also transduced in 96-well plates (surface area 0.32 cm 2 ) at Ix 10 5 and 5 ⁇ lO 5 cells in 200 ⁇ L SAF medium.
  • Lin-/- bone marrow cells were isolated as described above and transduced overnight at an MOI of 2. Subsequently, BALB/c mice were 6Gy irradiated and 3 ⁇ lO 6 or Ix 10 6 cells were injected intravenously in the tail vein. Blood was collected by orbital puncture 4 times until 190 days after transplantation and EGFP expression was determined in leucocytes.
  • the ⁇ -glucosidase (GAA) cDNA was removed from the pSHAG2-GAA vector previously described 7 and cloned in the pSUPER vector containing a polylinker with EcoRI and Xbal sites (pSli).
  • the pSli-GAA plasmid was digested with Agel and Xbal.
  • the GAA cDNA fragment was ligated into the Agel-Nhel sites of the pRRL.PPT.PGK.EGFP.WPRE4*.SIN lentiviral transfer vector (LV-PGK- GAA) after removal of the enhanced Green Fluorescent Protein (eGFP) cDNA.
  • the Agel-Swal fragment from the LV-PGK-GAA was cloned in place of the EGFP of the pRRL.PPT.SF.EGFP.WPRE4*.SIN lentiviral transfer vector to result in LV-SF-GAA lentiviral vector.
  • Lentiviral vector batches were produced as described above, except that the lentiviral vector preparations were titrated on mouse GAA '- mesenchymal stem cells.
  • mice All mice were of FVB background and GAA '- mice were described previously 8 .
  • GAA ⁇ mice have a complete deficiency in the alpha- glucosidase gene and therefore mimic the early onset-type of the disease, in clinical symptoms, and in pathological as well as biochemical findings 8 .
  • the animal experiments were approved by an ethical committee of Erasmus Medical Center, Rotterdam in accordance with legal regulations in The Netherlands.
  • mice Male GAA-'- mice were used as bone marrow donor for transplantation. Total bone marrow was flushed from both femora and tibias and of 8-12 week-old male GAA-/- mice, and lineage depleted (lirr'-) according to the manufacturer's protocol (BD).
  • cells were transduced by LV-SF-GFP of LV- SF-GAA lentiviral vectors overnight at IxIO 6 cells/mL at multiplicity of infection (MOI) of 9-10, in serum-free modified Dulbecco's (Stem Cell Activating, SAF) medium supplemented as described and containing growth factors (m-SCF lOOng/ml, hu-Flt3 50ng/ml, m-TPO lOng/ml).
  • SAF Stem-SCF lOOng/ml, hu-Flt3 50ng/ml, m-TPO lOng/ml.
  • Glycogen content was determined as described 10 by treatment of samples by amyloglucosidase. Resulting glucose was measured by treatment of glucose- oxidase and 2,2'-azino-di-(ethyl-benzthiazolinsulfonate) (ABTS). Final glycogen content values were determined after untreated glucose levels were subtracted. Both GAA and glycogen assays were measured by VarioSkan. GAA activity and glycogen content were corrected for protein content using the bicinchoninic acid (BCA) protein assay (Pierce).
  • BCA bicinchoninic acid
  • GAA +/+ , and GAA '- mice, LV-SF-GFP of LV-SF-GAA treated were sacrificed and both left ventricular and right ventricular mass were determined by echography.
  • Mouse lin-/- cells could be efficiently transduced with SF-GFP and PGK-GFP lentiviral vectors.
  • An increase of 2 ⁇ lO 4 to 6 ⁇ lO 5 Hn-/- cells/mL at an MOI of 1 resulted in an increase of 4% to 41% and 7% to 54% of EGFP positive cells for SF-GFP and PGK-GFP respectively ( Figure 1).
  • Similar transduction efficiencies could be achieved with UCB cells at 6 ⁇ lO 5 cells/mL, which were on average 62% for SF-GFP lentiviral vector transduction.
  • rhesus CD34+ cells transduced less efficiently, but on average 22% EGFP positive rhesus CD34+ cells were measured at an MOI of 1 at 6 ⁇ lO 5 cells/mL in 24 well plates ( Figure 1). Higher cell-densities in 96 well plates (5xlO 5 and 2.5 ⁇ lO 6 /mL) at an MOI of 1 resulted 31 and 50% EGFP positive cells respectively ( Figure 2). Lentiviral transduction efficiency in Retronectin-coated plates was not improved compared to non-coated plates. Increase of the MOI to 37 at 6xlO 5 cells/mL markedly increased the transduction efficiency from 21% to 80%.
  • skeletal muscle displayed a significant reduction of glycogen, although not as prominent as the other tissues (Figure 7).
  • the large activity increase of GAA in the heart tissue resulted in a near normalization of heart geometry and function as visualized by echography.
  • Heart rate (beats per minute) was reduced in LV-SF-GFP mice at 6.5 months of age (315 ⁇ 11) and significantly improved in LV-SF-GAA mice (385+27), similar to heart rate in healthy mice 363+17.
  • LV-SF-GFP mice develop ventricular hypertrophy due to cardiac muscle weakness.
  • mice and LV-SF-GAA mice had significantly reduced left ventricular weight (82.219.37 mg and 96.01 ⁇ 7.73 mg, respectively) compared to LV-SF-GFP mice (119.96+11.16 mg) (Figure 8).
  • LV-SF-GAA treated mice older than 150 days showed significantly improved muscle strength determined by grip strength measurements.
  • healthy mice were significantly stronger than LV-SF-GAA treated mice. Further locomotor and respiratory functional tests are in progress. Adverse effects on the hematopoietic system were not observed.
  • Increase in transduction efficiency of enriched hematopoietic bone marrow cells by increasing the cell-density (2 ⁇ lO 4 ⁇ 6 ⁇ l0 5 /mL in 24-well format) with a fixed MOI of 1. Increase in transduction efficiency is depicted for mouse lin -/- cells, rhesus CD34+ cells and CD34+ umbilical cord blood cells. These cells were transduced with lentiviral vectors containing the SF-EGFP or PGK- EGFP promoter cassettes.
  • Retronectin is not required for efficient transduction of rhesus CD34+ cells. Filled bars: retronectin, open bars: no retronectin.
  • transduced mouse lin-/- BMCs (MOI 2) in irradiated mice and the EGFP positive fraction in peripheral blood.
  • A,B Percentage of EGFP positive cells of cultured mouse lin-/- BMCs 6 days after transduction of a lentiviral vector containing the SF-EGFP promoter construct,
  • C-H Percentage of EGLFP positive cells in peripheral blood of mice 46 days after transplantation of transduced lin-/- BMCs: C 1 D) Erythrocytes.
  • E,F Thrombocytes.
  • G,H ⁇ Leucocytes.
  • A,C,E,G Non-transduced controls.
  • GAA activity in tissues Upper graph. At 8 months after treatment high GAA activity was observed in leukocytes of GAA-/- LV-SF-GAA treated mice, as well as bone marrow (BM) and spleen. GAA+/+ mice GAA activity was detectable in BM and spleen, but this was low. No GAA activity was detected in GAA-/- LV- SF-GFP treated mice. Lower graph. High GAA activity was observed in GAA-/- LV-SF-GAA mice in heart, lung, diafragm, liver, stomach , uterus and quadriceps femoris (QF) similar to GAA+/+ mice.
  • QF quadriceps femoris
  • Glycogen content in affected tisues was decreased in heart, lung, diafragm, liver, stomach and uterus and moderately reduced in quadriceps femoris (QF).
  • GM-CFU murine bone marrow granulocyte/macrophage progenitor cells

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention porte sur le domaine de la thérapie génique et plus spécifiquement sur des véhicules de distribution de gène lentiviral et sur des procédés pour une transduction efficace de véhicules de distribution de gène lentiviral dans des cellules souches hématopoïétiques et leurs descendants. De préférence, l'invention concerne dans l'un de ces modes de réalisation un procédé de transfert de gène dans, par exemple, des cellules souches hématopoïétiques pluripotentes et leurs descendants, permettant une transduction réussie des cellules, comprenant des populations cellulaires transplantables comprenant des cellules souches hématopoïétiques, qui donnent lieu à une descendance exprimant le ou les gènes transduits. L'invention comprend en outre un procédé pour traiter une diversité de maladies humaines héréditaires et acquises par transfert de gènes thérapeutiquement actifs dans les cellules souches hématopoïétiques. En tant qu'exemple non limitatif, l'invention montre que des symptômes associés à la maladie de Pompe sont (entièrement) réduits et/ou soulagés par traitement d'un sujet souffrant de la maladie de Pompe avec une cellule souche hématopoïétique transduite par un vecteur lentiviral comprenant une alpha-glucosidase.
PCT/NL2008/050269 2007-05-02 2008-05-02 Procédés et moyens améliorés pour une distribution de gène lentiviral WO2008136670A2 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
NLPCT/NL2007/050193 2007-05-02
PCT/NL2007/050193 WO2008136656A1 (fr) 2007-05-02 2007-05-02 Procédés perfectionnés et moyens pour l'administration de gènes lentiviraux
EP07118404.8 2007-10-12
EP07118404 2007-10-12

Publications (2)

Publication Number Publication Date
WO2008136670A2 true WO2008136670A2 (fr) 2008-11-13
WO2008136670A3 WO2008136670A3 (fr) 2009-02-19

Family

ID=39944128

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2008/050269 WO2008136670A2 (fr) 2007-05-02 2008-05-02 Procédés et moyens améliorés pour une distribution de gène lentiviral

Country Status (1)

Country Link
WO (1) WO2008136670A2 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019050406A1 (fr) * 2017-09-08 2019-03-14 Erasmus University Medical Center Rotterdam Nouvelle thérapie pour la maladie de pompe
US10617721B2 (en) 2013-10-24 2020-04-14 Ospedale San Raffaele S.R.L. Methods for genetic modification of stem cells
US11286501B2 (en) 2016-04-20 2022-03-29 Centro De Investigaciones Energeticas, Medioambientales Y Tecnologicas O.A, M.P. Methods of treating or preventing pyruvate kinase deficiency
WO2022229227A1 (fr) * 2021-04-28 2022-11-03 Ospedale San Raffaele S.R.L. Compositions pour améliorer la transduction de cellules par des vecteurs viraux
US11642422B2 (en) 2017-10-16 2023-05-09 Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, O.A, M.P. Lentiviral vectors for delivery of PKLR to treat pyruvate kinase deficiency
WO2023204714A1 (fr) 2022-04-22 2023-10-26 Erasmus University Medical Center Rotterdam Thérapie génique pour la maladie de pompe
WO2024096738A1 (fr) 2022-11-01 2024-05-10 Erasmus University Medical Center Rotterdam Constructions de thérapie génique pour troubles métaboliques

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRENNER SEBASTIAN ET AL: "The late dividing population of gamma-retroviral vector transduced human mobilized peripheral blood progenitor cells contributes most to gene-marked cell engraftment in nonobese diabetic/severe combined immunodeficient mice." STEM CELLS (DAYTON, OHIO) JUL 2007, vol. 25, no. 7, July 2007 (2007-07), pages 1807-1813, XP009095635 ISSN: 1066-5099 *
Cell density and viral vector load are determinant factors for ex vivo lentiviral transduction effeciency of hematopoietic stem cells. June 2, 2007 XP002468030 *
SU LISHAN ET AL: "Hematopoietic stem cell-based gene therapy for acquired immunodeficiency syndrome: Efficient transduction and expression of RevM10 in myeloid cells in vivo and in vitro" BLOOD, vol. 89, no. 7, 1997, pages 2283-2290, XP002468028 ISSN: 0006-4971 *
SUTTON R E ET AL: "TRANSDUCTION OF HUMAN PROGENITOR HEMATOPOIETIC STEM CELLS BY HUMAN IMMUNODEFICIENCY VIRUS TYPE 1-BASED VECTORS IS CELL CYCLE DEPENDENT" JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 73, no. 5, May 1999 (1999-05), pages 3649-3660, XP002937676 ISSN: 0022-538X *
ZHANG B ET AL: "The significance of controlled conditions in lentiviral vector titration and in the use of multiplicity of infection (MOI) for predicting gene transfer events" GENETIC VACCINES AND THERAPY 04 AUG 2004 UNITED KINGDOM, vol. 2, 4 August 2004 (2004-08-04), page 10p, XP002468027 ISSN: 1479-0556 1479-0556 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10617721B2 (en) 2013-10-24 2020-04-14 Ospedale San Raffaele S.R.L. Methods for genetic modification of stem cells
US11286501B2 (en) 2016-04-20 2022-03-29 Centro De Investigaciones Energeticas, Medioambientales Y Tecnologicas O.A, M.P. Methods of treating or preventing pyruvate kinase deficiency
WO2019050406A1 (fr) * 2017-09-08 2019-03-14 Erasmus University Medical Center Rotterdam Nouvelle thérapie pour la maladie de pompe
NL2019517B1 (en) * 2017-09-08 2019-03-19 Univ Erasmus Med Ct Rotterdam New therapy for Pompe disease
US11642422B2 (en) 2017-10-16 2023-05-09 Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, O.A, M.P. Lentiviral vectors for delivery of PKLR to treat pyruvate kinase deficiency
WO2022229227A1 (fr) * 2021-04-28 2022-11-03 Ospedale San Raffaele S.R.L. Compositions pour améliorer la transduction de cellules par des vecteurs viraux
WO2023204714A1 (fr) 2022-04-22 2023-10-26 Erasmus University Medical Center Rotterdam Thérapie génique pour la maladie de pompe
NL2031676B1 (en) 2022-04-22 2023-11-07 Univ Erasmus Med Ct Rotterdam Gene therapy for Pompe Disease
WO2024096738A1 (fr) 2022-11-01 2024-05-10 Erasmus University Medical Center Rotterdam Constructions de thérapie génique pour troubles métaboliques

Also Published As

Publication number Publication date
WO2008136670A3 (fr) 2009-02-19

Similar Documents

Publication Publication Date Title
Reiser et al. Potential of mesenchymal stem cells in gene therapy approaches for inherited and acquired diseases
Naldini Ex vivo gene transfer and correction for cell-based therapies
Kohn The current status of gene therapy using hematopoietic stem cells
JP2019511240A (ja) ヌクレアーゼを使用するヒト神経幹細胞のゲノム編集
JP2014088432A (ja) Cd34幹細胞に関連した方法および組成物
WO2008136670A2 (fr) Procédés et moyens améliorés pour une distribution de gène lentiviral
Bedel et al. Metabolic correction of congenital erythropoietic porphyria with iPSCs free of reprogramming factors
JP2021517168A (ja) パーキンソン病を処置するための組成物及び方法
Clapp Somatic gene therapy into hematopoietic cells: current status and future implications
WO2008136656A1 (fr) Procédés perfectionnés et moyens pour l'administration de gènes lentiviraux
JP6676262B2 (ja) テロメラーゼ逆転写酵素に基づいた治療
JP2022512674A (ja) 人工トランス活性化因子による選択
Alenzi et al. Stem cells and gene therapy
JP2021522278A (ja) ライソゾーム病を改善するための、レンチウイルスベクターが形質導入されたt−rapa細胞の使用
Nienhuis et al. Gene transfer into hematopoietic cells
US20240093242A1 (en) Gene correction for scid-x1 in long-term hematopoietic stem cells
US20240122989A1 (en) Methods and compositions for production of genetically modified primary cells
US20230136849A1 (en) Capsid-modified raav vector compositions and methods therefor
US20220184137A1 (en) Correction of Beta-Thalassemia Phenotype by Genetically Engineered Hematopoietic Stem Cell
Trarbach et al. Optimized retroviral transduction protocol for human progenitor cells utilizing fibronectin fragments
Bueren et al. Genetic modification of hematopoietic stem cells: recent advances in the gene therapy of inherited diseases
Becker Hematopoietic stem cell gene therapy for inherited bone marrow disorders: past accomplishments and continued challenges
Das et al. An empowered, clinically viable hematopoietic stem cell gene therapy for the treatment of multisystemic mucopolysaccharidosis type II
CN113544279A (zh) 包含芳基硫酸酯酶a的重组载体及其在用于治疗异染性脑白质营养不良的干细胞治疗中的用途
KR20230131816A (ko) 저면역원성 줄기세포, 줄기세포로부터 분화되거나 유래된 저면역원성 세포및 이의 제조방법

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08741690

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08741690

Country of ref document: EP

Kind code of ref document: A2