+

WO2008136670A2 - Improved methods and means for lentiviral gene delivery - Google Patents

Improved methods and means for lentiviral gene delivery 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
French (fr)
Other versions
WO2008136670A3 (en
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/en
Application filed by Erasmus University Medical Center Rotterdam filed Critical Erasmus University Medical Center Rotterdam
Publication of WO2008136670A2 publication Critical patent/WO2008136670A2/en
Publication of WO2008136670A3 publication Critical patent/WO2008136670A3/en

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

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. Preferably, 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 therapeutically active genes into hematopoietic stem cells. As a non-limiting example, the invention shows that symptoms associated with Pompe disease are (completely) reduced and/or alleviated by treatment of a subject suffering from Pompe disease withJiematopoietic stem cell transduced with an alpha- glucosidase comprising lentiviral vector.

Description

Title: Improved methods and means for lentiviral gene delivery
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. Preferably, 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. As a non-limiting example, 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.
Successful correction of inherited hematopoietic disorders by gamma retrovirus vector-mediated ex υiυo hematopoietic stem cell (HSC) gene- modification has been demonstrated for X-linked severe combined immunodeficiency SCID and adenosine deaminase deficiency (ADA-SCID). However, 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 (LV) display a reduced risk pattern due to deletion of enhancer regions and a more favourable integration pattern as compared to gamma-retroviruses. With current LV transduction protocols, a high proportion of gene-modified HSCs requires a high multiplicity of infection (MOI), or rather a high concentration of transducing units (TU), which results in an undesirable high number of transgene copies per cell.
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. To test optimal transduction conditions the inventors used VSV-G packaged LV expressing EGFP. The inventors observed that retronectin and growth factor supported overnight transduction of lineage depleted (lin Λ) mouse bone marrow cells (BMC) with low cell-density, but a high MOI of 30 yielded approximately 60% EGFP+ cells.
Unexpectedly, at an MOI of 1, the transduction efficiency of lirr7- cells could be markedly improved from 7% to 54% by increasing the cell density from 2 x 104/ml to 6 x 105/ml with a proportional increase of TU/ml.
The increased cell density/viral load did not enhance the mean fluorescence of EGFP+ cells, suggesting that integrations per cell did not increase. Q-PCR confirmed an average of approximately 1 provirus copy per cell.
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.
Similar results were obtained for human CD34+ umbilical cord blood (UCB) cells. Transduction efficiencies of up to 70% at 6 x 105 cells/ml were observed in UCB cells by increasing TU/ml with proportionaHncreases in cell density, maintaining a low MOI of 1.
In summary, 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. In a first embodiment, 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.
Surprisingly, 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. Moreover, 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.
As used herein, the term 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. Herein 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. Since blood cells virtually reach every organ, 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. Unfortunately, until the present invention, 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.
In more detail, 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). 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). Other sources for clinical and scientific use include umbilical cord blood, and placenta. For experimental purposes, 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 (rhodamineDULL, also called rho10) 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. 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. In a preferred embodiment, 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.
Gene delivery vehicles of lentiviral origin are all vehicles comprising genetic material and/or proteinaceous material derived from lentiviruses.
Typically 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.
It is clear to the skilled person that 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 prior art in respect of lentiviral transduction. of hematopoietic stem cells typically use a target cell concentration of for example IxIO5 to 4xlO5 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 5-2OxIO4 cells versus a MOI of 100.
It is clear from the experimental part herein that an increase of 2xlO4 to 6xlO5 cells/mL at an MOI of 1 resulted in an increase of transduction percentage..
In a preferred embodiment, 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 6xlO5/mL. This concentration of cells is especially useful for human and murine cells. For rhesus macaque cells, transduction percentages comparable to the mentioned human and murine percentage were obtained by using a cell density of 2.5χlO6 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 IxIO8 cells. Based on the herein disclosed results an upper limit of 107 to 2xlO7 should be feasible. In a preferred embodiment, 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 107 to 2xlO7 cells/mL. In yet another preferred embodiment, 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 6xlO5 cells/mL and maximal 107 to 2xlO7 cells/mL.
The herein used concentration of target cells (i.e. HSC) 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).
The term multiplicity of infection (MOI) 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. In yet another preferred embodiment, 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.
It is clear to the skilled person that an MOI of between 0.1 and 0.9 can equally well be used. However, in such a case, a method of the invention preferably further comprises selecting the cells that have been transduced. Preferably, the MOI of the used viral particles is determined on
HeLa cells.
In a preferred embodiment, 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.
Methods and means (such as producer cells) for producing the desired lentiviral particles are well known in the art. Examples of preferred producer cells are 293T cells that are cα-transfected with VSV-G (Vesicular stomatitis virus- G-protein envelope). Other pseudotypes used in this setting are RDl 14 (Feline-immunodeficiency virus).
In a most preferred embodiment, 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 6xl05 cells/mL and wherein said relatively low amount of transducing units per cell is 1-10. Typically, the term "gene delivery vehicle of lentiviral origin" refers to a gene delivery of any lentiviral origin. Currently, five serogroups of lentiviruses are recognized, reflecting the vertebrate hosts with which they are associated (primates, sheep and goats, horses, cats, and cattle): bovine, equine, feline, ovinecaprine and HIV serogroup. In a preferred embodiment, 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.
In yet another preferred embodiment, 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.
The invention also includes compositions obtainable by the methods of the invention. Thus included are 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. Preferable, 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.
The term multiplicity of infection (MOI) 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. In yet another preferred embodiment, 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. Preferably, the MOI of the used viral particles is determined on
HeLa cells.
In a preferred embodiment, 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. Typically the use of 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. Thus 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.
In yet another preferred embodiment, 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. Typically the use of 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.
To provide insight in the useful applications of the current invention the following paragraphs are directed to genetic diseases in general and to the applicability of the current invention in respect of lysosomal storage diseases. Treatment directed to alleviation of symptoms associated-with Pompe's disease is provided as a non-limiting example.
Rare diseases, of which 80% are genetic diseases, affect some 6% of the human population, amounting in Europe to around 30 million people. Since many result in chronic disability and cost-intensive care, the impact is disproportional and may well be over 20% of health care costs. Most of these diseases lack a curative intervention, are managed by symptomatic treatment, and are amenable to gene therapy with curative intent. At the state-of-the-art, 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. For some, allogeneic hematopoietic stem cell (HSC) transplantation (alloSCT) is an effective treatment, even in diseases with brain involvement. However, 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. Ectopic production of enzyme by autologous HSC is anticipated to combine the advantages of SCT with those of enzyme therapy into a single curative procedure at limited cost. Pompe's disease (glycogen storage disease type II; acid maltase deficiency) is a lethal autosomal recessive disorder caused by acid a- glucosidase deficiency, which results in generalized storage of glycogen in many tissues such as brain, liver, spleen, kidney, endothelial cells, and most prominently in skeletal muscles, heart and smooth muscle. Symptoms arise from muscle weakness and wasting. Infants with complete enzyme deficiency present shortly after birth and become completely quadriplegic within 8 months. The natural course of Pompe's disease is invalidating by progressive loss of mobility and respiratory function and is lethal for severely affected infants in the first year of life. Older children and adults with residual activity show a more protracted course of disease and become wheelchair bound, dependent on artificial ventilation and die from cardiac/respiratory failure ranging from early childhood to late adulthood. Recently, the EMEA and FDA have approved ERT with recombinant human alpha-glucosidase for all patients. Enzyme therapy is well tolerated, its beneficial effects are unquestionable and prolong life significantly, but do not guarantee long-term symptom free survival. The therapy prolongs life of severely affected infants significantly and improves motor outcome, but during 8 years of experience approximately half of the treated infants ultimately died and all develop some form of residual disease. The beneficial effect of enzyme therapy in older children and adults still needs to be validated. The development of alternative gene therapy therefore would significantly contribute to the survival of 50% of the severely affected infants, and eventually would be particularly beneficial as a single treatment option for the juvenile form of Pompe's disease.
Previous experiments in which adenovirus and adeno-associated virus based vectors were used for systemic delivery of acid a-glucosidase gene constructs in mice have demonstrated its efficacy in long lasting reduction of glycogen storage. Importantly, its expression in muscle was not essential. Also, the liver could function as an enzyme -secreting depot supplying corrective enzyme to muscle and other tissues, warranting exploration of liver directed systemic administration of therapeutic vector. However, these vectors are applied via an in vivo approach and as a result shedding problems may arise as well as adverse immune reactions. Moreover, permanent integration is not necessarily obtained and hence treatment must be repeated in time.
In the experimental part herein, we demonstrate in an available mouse model that high level expression of human acid a-glucosidase in descendents of HSC transduced with SIN-lentiviral vectors provides a valid treatment option for alleviation of clinical symptoms of Pompe's disease. This approach does not require the administration of virus particles to the patient (compared to ade no- based treatment). 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. In a preferred embodiment, 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.
The particulars of the present invention are further used to develop a kit, preferably a closed clinical grade device, which comprises means to purify and then transduce target cells (hematopoietic stem / progenitor cells). Thus, the invention further comprises a kit comprising means to isolate and/or purify hematopoietic stem cells and a therapeutic gene delivery vehicle.
For selection purposes special columns are already on the market (Miltenyi; for research purposes Mini-Midi and-Maxi columns, for the clinical setting CliniMacs columns). However, other means for isolating and/or purifying HSC are equally suitable for incorporation in a kit.
The other component of the described kit is a gene delivery vehicle that comprises a therapeutically interesting gene. Preferably, said gene delivery vehicle is of lentiviral origin. Typically 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. It is clear to the skilled person that 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.
In a preferred embodiment, 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. Moreover, 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). However 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.
Moreover, two general approaches can be used to correct defective genes within stem cells: gene addition and genome editing. 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. In contrast, 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.
Hence, in yet another embodiment, 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. Examples of such means 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 viυo 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.
In yet another embodiment, 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. An example of such a means is a Zinc finger endonuclease.
The invention will be explained in more detail in the following, non- limiting examples. Experimental part Materials and methods
Production of lentiυiral 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. We used 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). In addition, 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.
Enrichment of stem cells.
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). Rhesus monkey bone marrow cells
Low-density cells were isolated using Ficoll separation (1.077 g/cm2, Nycomed, Pharma, AS, Oslo, Norway) and cryopreserved until subset purification or directly continued towards purification and transduction.
Subset purification
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. 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).
Human umbilical cord blood cells
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/cm2, 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.
Subset purification
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. 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).
Standard stem cell 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,.5.8xlθ-4 M L-cysteine, 5-lxlO 4 M L-glutamic acid, 3.5 xlO 4 M L-proline, l.δxlO"5 M cholesterol, 4 μM cytidine, 4 μM adenosine, 4 μM uridine, 3.5 μM guanosine, 4.4 μM 2'-deoxycytidine, 4 μM
2'deoxyadenosine, 4 μM thymidine, 3.7 μM 2'deoxy guanosine, 1.2xlO 7 M d- - biotin, 1% fraction V BSA, (all Sigma-Aldrich, Zwijndrecht, The Netherlands), 1.SxIO-8 M vitamin BΪ2, 10 3 M sodium pyruvate, 1.9xlO 2 M glucose (Merck), 4.4xlO-2 M NaHCO3, 10-* M β-mercapto-ethanol, KF M Na2SeO3, 1.5xl0 5 M linoleic acid (Merck, Darmstadt, Germany), 0.1 g/1 penicillin (Yamanouchi, Leiderdorp, The Netherlands), 105 IE/1 streptomycin (Fisiopharma, Milano, Italy), 2xlO'6 M iron saturated transferrin (Serologicals Proteins Inc. Kankakee, IL, USA), at an osmolarity of 300 mOsm/1. Transduction of hematopoietic stem cells.
Transduction of the cells was started at the same day of enrichment of the stem-cells. For the in vitro experiments lentiviral vector supernatant was mixed with the enriched hematopoietic stem cells at increasing cell-density of 2xlO4, 6xlO4, 1.2xlO5, 2χl05 or 6xlO5 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 2cm2). 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. In addition, rhesus CD34+ cells were also transduced in 96-well plates (surface area 0.32 cm2) at Ix 105 and 5χlO5 cells in 200μL SAF medium.
In vivo efficacy of transplanted lentiviral vector transduced hematopoietic stem cell.
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 3χlO6 or Ix 106 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.
Materials and methods for the treatment of Pompe disease
Development of lentiviral vector plasmids
The α-glucosidase (GAA) cDNA was removed from the pSHAG2-GAA vector previously described7 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.
Animals
All mice were of FVB background and GAA '- mice were described previously8. GAA Λ 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 findings8. The animal experiments were approved by an ethical committee of Erasmus Medical Center, Rotterdam in accordance with legal regulations in The Netherlands.
Lentiviral hematopoietic stem cell transduction
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). After enrichment, cells were transduced by LV-SF-GFP of LV- SF-GAA lentiviral vectors overnight at IxIO6 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). The following day, 6 Gy irradiated 8-12 week-old GAA '- mice were injected in tail vein with 5xlO5 transduced lin '- cells.
Enzymatic assays
All tissues were sonicated on ice until tissues were completely lysed (medium level, amplitude 5). Activity of GAAwas determined by fluorometry, according to a previous described protocol based on 4-methylumbelliferyl α-D- glucopyranoside (4MU, Sigma)9.
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).
Measurement of cardiac hypertrophy
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.
Experimental part Results
In vitro lentiviral vector transduction efficiency of hematopoietic stem cells.
Mouse lin-/- cells could be efficiently transduced with SF-GFP and PGK-GFP lentiviral vectors. An increase of 2χlO4 to 6χlO5 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 6χlO5 cells/mL, which were on average 62% for SF-GFP lentiviral vector transduction. In contrast, rhesus CD34+ cells transduced less efficiently, but on average 22% EGFP positive rhesus CD34+ cells were measured at an MOI of 1 at 6χlO5 cells/mL in 24 well plates (Figure 1). Higher cell-densities in 96 well plates (5xlO5 and 2.5χlO6/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 6xlO5 cells/mL markedly increased the transduction efficiency from 21% to 80%.
In vivo lentiviral vector transduction efficiency of hematopoietic stem cells.
Mouse lin-/- BMCs transduced with lentiviral vectors at an MOI of 2 resulted in 98% EGFP positive cells, with up to 80% EGFP positive leucocytes in peripheral blood 46 days after transplantation in irradiated mice (Figure 3). A large proportion of EGFP positive cells was also measured in erythrocytes and thrombocytes. Long-term EGFP positive cells were measured in leucocytes for up to 190 days after transplantation (Figure 4), which shows the potential of the high cell-density transduction efficiency at low MOI. Quantitative PCR oflentiviral integrations
We analysed the number of integrations in the transduced mouse lin-/- cells, rhesus macaque CD34+ cells and human CD34+ UCBs, transduced at an MOI of 1. The number of integrations was at maximum 3.1 integrations per cell at 6xlO5 cells/mL for mouse lin-/- cells. At a cell-density of 6xlO5 cells/mL in rhesus macaque cells the average number of integrations per cell was 1.1 and in human UCBs 2.7. All the lower cell-densities resulted in less integrations per cell compared to the highest cell-density (6xlO5 cells/mL).
Results treatment ofPompe disease
The stem cell gene therapy approach for Pompe's disease has been tested in experiments using the GAA-/- mouse model for Pompe's disease. Long-term (1 year) GAA activity in leukocytes was detected in ex vivo LV-SF-GAA-corrected mice (96.8±53 nmol 4MU/h/mg, n=31) compared to expression in LV-SF-GFP controls (1.4±0.8 nmol 4MU/h/mg, n=14, pO.OOl) (Figure 5), resulting in high levels of enzyme in affected organs (Figure 6). PAS staining demonstrated a near complete clearance of glycogen in liver, spleen and cardiac muscle (Figure 7). Also 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. In addition, LV-SF-GFP mice develop ventricular hypertrophy due to cardiac muscle weakness. At 8 months after treatment, both the healthy 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. However, consistent with the glycogen clearance levels, 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.
We conclude that ex vivo hematopoietic system mediated gene therapy corrects defects in the Pompe mouse model, indicating that the approach provides a valid alternative for enzyme replacement therapy in the treatment of Pompe's disease. Codon optimization of the transgene is anticipated to provide a further elevation of enzyme levels.
Description of figures
Figure 1.
Increase in transduction efficiency of enriched hematopoietic bone marrow cells by increasing the cell-density (2χlO4 →6χl05/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.
Figure 2.
Increase in lentiviral vector transduction efficiencies of rhesus CD34+ cells at high cell-densities at a fixed MOI of 1. Retronectin is not required for efficient transduction of rhesus CD34+ cells. Filled bars: retronectin, open bars: no retronectin.
Figure 3.
Transplantation of 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: C1D) Erythrocytes. E,F) Thrombocytes. G,H)~ Leucocytes. A,C,E,G) Non-transduced controls.
Figure 4.
Long-term EGFP expression in mouse leucocytes after high cell-density ex viυo lentiviral vector transduction. Transplantation of transduced mouse lin-/- BMCs (MOI 2) in 6Gy irradiated mice and the EGFP positive fraction in leucocytes. Percentage of EGFP positive leucocytes in mice transplanted with 3xlO6 and Ix 106 transduced lin-/- cells containing the SF-EGFP promoter construct. Long-term EGFP expression was observed until 190 days after transplantation.
Figure 5.
Long-term expression of GAA in blood. Long-term GAA activity was detected by enzymatic assay in leukocytes of GAA-/- LV-SF-GAA-treated mice (n=32) compared to LV-SF-GFP (n=15) treated mice. GAA activity of leukocytes in GAA+/+ mice (n=6) was comparable to GAA-/- mice.
Figure 6.
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.
Figure 7.
Glycogen content in affected tisues. Glycogen content of GAA-/- LV-SF-GAA treated mice was decreased in heart, lung, diafragm, liver, stomach and uterus and moderately reduced in quadriceps femoris (QF).
Figure 8.
Reduction in left ventricular cardiac hypertrophy after LV-SF-GAA treatment. Cardiac mass was determined in 10-month-old mice, eight months after hematopoietic transplantation. Left ventricular mass was significantly increased in GAA-/- LV-SF-GFP mice compared to GAA+/+ mice (*, p=0.001). In addition, GAA-/- LV-SF-GAA treated mice showed a significant reduction in left ventricular mass compared to GAA-/- LV-SF-GFP mice (f, p<0.05).
References
1. Dull, T., R. Zufferey, M. Kelly, R. J. Mandel, M. Nguyen, D. Trono, and L. Naldini. 1998. A third-generation lentivirus vector with a conditional packaging system. J. Virol. 72:8463-8471.
2. Follenzi, A. and L. Naldini. 2002. HIV-based vectors. Preparation and use. Methods MoI. Med. 69:259-274.
3. Merchav, S. and G. Wagemaker. 1984. Detection of murine bone marrow granulocyte/macrophage progenitor cells (GM-CFU) in serum-free cultures stimulated with purified M-CSF or GM-CSF. Int. J. Cell Cloning 2:356-367.
4. Schambach, A., J. Bohne, C. Baum, F. G. Hermann, L. Egerer, D. von Laer, and T. Giroglou. 2006. Woodchuck hepatitis virus post-transcriptional regulatory element deleted from X protein and promoter sequences enhances retroviral vector titer and expression. Gene Ther. 13:641-645.
5. Seppen, J., R. R. van der, N. Looije, N. P. van Til, W. H. Lamers, and R. P. Oude Elferink. 2003. Long-term correction of bilirubin UDPglucuronyltransferase deficiency in rats by in utero lentiviral gene transfer. MoI. Ther. 8:593-599. 6. Woods, N. B., A. Muessig, M. Schmidt, J. Flygare, K. Olsson, P.
Salmon, D. Trono, C. von Kalle, and S. Karlsson. 2003. Lentiviral vector transduction of NOD/SCID repopulating cells results in multiple vector integrations per transduced cell: risk of insertional mutagenesis. Blood 101:1284-1289. References - continued
7. Hoefsloot,L.H., WilIemsen,R., Kroos,M.A., Hoogeveen- Westerveld,M., Hermans,M.M., Van der Ploeg,A.T., Oostra,B-A., and Reuser,A.J.: Expression and routeing of human lysosomal alpha-glucosidase in transiently transfected mammalian cells. Biochem.J. 272:485-492, 1990.
8. BijvoetΛ-G-» van de Kamp,E.H., Kroos,M.A., Ding,J.H., Yang,B.Z., Visser,P., Bakker,C.E., Verbeet,M.P., Oostra,B.A., Reuser,A. J., and Van der Ploeg,A.T.: Generalized glycogen storage and cardiomegaly in a knockout mouse model of Pompe disease. Hum.Mol.Genet. 7:53-62, 1998.
9. de JongβjA-J-j de Smit,S., Kroos,M.A., and Reuser,A. J.: Cotransfer of syntenic human genes into mouse cells using isolated metaphase chromosomes or cellular DNA. Hum.Genet. 69:32-38,
1985. 10. BijvoetAG., Kroos,M.A., Pieper,F.R., Van,d., V, De Boer,H.A., Van der Ploeg,A'T., Verbeet,M.P., and Reuser^A. J.: Recombinant human acid alpha-glucosidase: high level production in mouse milk, biochemical characteristics, correction of enzyme deficiency in GSDII KO mice. Hum.Mol.Genet. 7:1815-1824, 1998.

Claims

Claims
1. A method for transducing hematopoietic stem cells with a gene delivery vehicle of lentiviral origin comprising contacting an increased amount of hematopoietic stem cells (HSC) with a relatively low amount of transducing units per cell of said gene delivery vehicle.
2. A method according to claim 1, wherein said HSC comprise bone marrow cells, umbilical cord blood cells or mobilized peripheral blood stem cells.
3. A method according to claim 1 or 2 wherein said HSC are CD 34 positive cells, or CD34 positive / CD38 dull cells.
4. A method according to any one of claims 1 to 3, wherein said amount of HSC is at least 6x105 cells/mL.
5. A method according to any one of claims 1 to 4, wherein said relatively low amount of transducing units per cell is 1-10.
6. A method according to any one of claims 1 to 5, wherein said gene delivery vehicle of lentiviral origin is a gene delivery vehicle of HIV lentiviral origin.
7. A method according to any one of claims 1 to 6 which is an ex vivo method.
8. A composition comprising lentiviral particles wherein said composition has a relatively low amount of transducing units per amount of target cell.
9. A composition according to claim 8, wherein said lentiviral particles are gene delivery vehicles.
10. A composition according to claim 8 or 9 for use as a pharmaceutical.
11. Use of a composition according to claim 8 or 9 in the transduction of hematopoietic stem cells.
12. A composition for the treatment of a hereditary disease or pathological condition related to a genetic defect or a genetic aberration, comprising a plurality of hematopoietic stem cells transduced with a composition according to claim 8 or 9.
13. A composition for the treatment of a hereditary disease or pathological condition related to a genetic defect or a genetic aberration, comprising a plurality of hematopoietic stem cells or of mesenchymal stem cells or of induced pluripotential stem cells, said composition being obtainable by a method according to any one of claims 1 to 7.
14. Use of composition according to claim 12 or 13 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.
15. Use of a composition according to claim 12 or 13 in the preparation of a medicament for the treatment of Pompe disease or a pathological condition related to Pompe disease.
16. A kit comprising means to isolate and/or purify hematopoietic stem cells and a therapeutic gene delivery vehicle.
17. A kit according to claim 16 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.
18. A method according to any one of claims 1 to 7, wherein said gene delivery vehicle is provided with means for homologous recombination.
19. A method according to any one of claims 1 to 7, wherein said gene delivery vehicle is provided with means for genome editing.
20. A method according to one of claims 1 to 7, wherein said gene delivery vehicle is provided with means to modify gene expression.
21. A method according to one of claims 1 to 7, wherein said gene delivery vehicle is mutated and provided with means for non-integrating gene expression.
PCT/NL2008/050269 2007-05-02 2008-05-02 Improved methods and means for lentiviral gene delivery WO2008136670A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
NLPCT/NL2007/050193 2007-05-02
PCT/NL2007/050193 WO2008136656A1 (en) 2007-05-02 2007-05-02 Improved methods and means for lentiviral gene delivery
EP07118404 2007-10-12
EP07118404.8 2007-10-12

Publications (2)

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

Family

ID=39944128

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2008/050269 WO2008136670A2 (en) 2007-05-02 2008-05-02 Improved methods and means for lentiviral gene delivery

Country Status (1)

Country Link
WO (1) WO2008136670A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019050406A1 (en) * 2017-09-08 2019-03-14 Erasmus University Medical Center Rotterdam New therapy for pompe disease
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 (en) * 2021-04-28 2022-11-03 Ospedale San Raffaele S.R.L. Compositions for improving the transduction of cells by viral vectors
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 (en) 2022-04-22 2023-10-26 Erasmus University Medical Center Rotterdam Gene therapy for pompe disease
WO2024096738A1 (en) 2022-11-01 2024-05-10 Erasmus University Medical Center Rotterdam Gene therapy constructs for metabolic disorders

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 (10)

* 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
US12163151B2 (en) 2016-04-20 2024-12-10 Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas O.A, M.P. Methods of treating or preventing pyruvate kinase deficiency
WO2019050406A1 (en) * 2017-09-08 2019-03-14 Erasmus University Medical Center Rotterdam New therapy for pompe disease
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 (en) * 2021-04-28 2022-11-03 Ospedale San Raffaele S.R.L. Compositions for improving the transduction of cells by viral vectors
WO2023204714A1 (en) 2022-04-22 2023-10-26 Erasmus University Medical Center Rotterdam Gene therapy for pompe disease
NL2031676B1 (en) 2022-04-22 2023-11-07 Univ Erasmus Med Ct Rotterdam Gene therapy for Pompe Disease
WO2024096738A1 (en) 2022-11-01 2024-05-10 Erasmus University Medical Center Rotterdam Gene therapy constructs for metabolic disorders

Also Published As

Publication number Publication date
WO2008136670A3 (en) 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
McClain et al. In utero stem cell transplantation and gene therapy: Recent progress and the potential for clinical application
Kohn The current status of gene therapy using hematopoietic stem cells
JP2014088432A (en) Cd34 stem cell-related method and composition
WO2008136670A2 (en) Improved methods and means for lentiviral gene delivery
JP2021517168A (en) Compositions and Methods for Treating Parkinson&#39;s Disease
JP2022512674A (en) Selection by Artificial TransActivator
US20230136849A1 (en) Capsid-modified raav vector compositions and methods therefor
US20250000912A1 (en) Hypoimmunogenic stem cells, hypoimmunogenic cells differentiated or derived from stem cells and method of preparing same
JP7586516B2 (en) Treatment of chronic granulomatous disease
WO2008136656A1 (en) Improved methods and means for lentiviral gene delivery
US20240122989A1 (en) Methods and compositions for production of genetically modified primary cells
JP6676262B2 (en) Telomerase reverse transcriptase-based therapy
Nyberg et al. In vivo engineering of murine T cells using the evolved adeno-associated virus variant Ark313
JP7469328B2 (en) Correction of the beta-thalassemia phenotype by genetically engineered hematopoietic stem cells
Nienhuis et al. Gene transfer into hematopoietic cells
CN113544279A (en) Recombinant vectors comprising arylsulfatase A and their use in stem cell therapy for the treatment of metachromatic leukodystrophy
Alenzi et al. Stem cells and gene therapy
US20240093242A1 (en) Gene correction for scid-x1 in long-term hematopoietic stem cells
Trarbach et al. Optimized retroviral transduction protocol for human progenitor cells utilizing fibronectin fragments
Gostynski et al. Long-term survival of type XVII collagen revertant cells in an animal model of revertant cell therapy
Becker Hematopoietic stem cell gene therapy for inherited bone marrow disorders: past accomplishments and continued challenges
WO2024197058A2 (en) Methods for creating regulatory t cells (tregs) using genome engineering
CN117624325A (en) Bcl2 mutants that can promote large gene expression and have low tumorigenicity and their applications
Bellantuono et al. Adult Stem Cells and Gene Therapy

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

点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载