WO1996009400A1 - Procedes permettant de modifier genetiquement des cellules souches hematopoietiques - Google Patents

Procedes permettant de modifier genetiquement des cellules souches hematopoietiques Download PDF

Info

Publication number
WO1996009400A1
WO1996009400A1 PCT/US1995/011892 US9511892W WO9609400A1 WO 1996009400 A1 WO1996009400 A1 WO 1996009400A1 US 9511892 W US9511892 W US 9511892W WO 9609400 A1 WO9609400 A1 WO 9609400A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
stem cells
cell
stem
vsv
Prior art date
Application number
PCT/US1995/011892
Other languages
English (en)
Inventor
William G. Kerr
Garry P. Nolan
James J. Mule
Original Assignee
Systemix, Inc.
The Board Of Trustees Of The Leland Stanford Junior University
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
Application filed by Systemix, Inc., The Board Of Trustees Of The Leland Stanford Junior University filed Critical Systemix, Inc.
Priority to AU36356/95A priority Critical patent/AU3635695A/en
Publication of WO1996009400A1 publication Critical patent/WO1996009400A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/20011Rhabdoviridae
    • C12N2760/20211Vesiculovirus, e.g. vesicular stomatitis Indiana virus
    • C12N2760/20222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • Mammalian hematopoietic cells provide a diverse range of physiological activities. These cells are divided into lymphoid, myeloid and erythroid lineages.
  • the lymphoid lineages comprising B cells and T cells, provides for the production of antibodies, regulation of the cellular immune system, detection of foreign agents in the blood, detection of cells foreign to the host, and the like.
  • the myeloid lineage which includes monocytes, granulocytes,
  • megakaryocytes as well as other cells, monitors for the presence of foreign bodies, provides protection against neoplastic cells, scavenges foreign materials, produces platelets, and the like.
  • the erythroid lineage provides the red blood cells, which act as oxygen carriers.
  • stem cells stem cells
  • Stem cells are capable of self-regeneration and may become lineage committed progenitors which are dedicated to differentiation and expansion into a specific lineage.
  • stem cells refers to hematopoietic cells and not stem cells of other cell types. Further, unless indicated otherwise, “stem cells” refers to human hematopoietic stem cells.
  • U.S. Patent No. 5,061,620 describes a substantially homogeneous stem cell composition and the manner of obtaining such a composition. See also the references cited therein.
  • Stem cells constitute only a small percentage of the total number of hematopoietic cells. Hematopoietic cells are identifiable by the presence of a variety of cell surface "markers.” Such markers may be either specific to a particular lineage or progenitor cell or be present on more than one cell type. Currently, it is not known how many of the markers associated with differentiated cells are also present on stem cells. One marker, which was previously indicated as present solely on stem cells, CD34, is also found on a significant number of lineage committed
  • U.S. Pat. No. 4,714,680 describes a population of cells expressing the CD34 marker.
  • Table 1 summarizes probable phenotypes of stem cells in fetal, adult, and mobilized peripheral blood.
  • myelomonocytic stands for myelomonocytic associated markers
  • NK stands for natural killer cells
  • AMPB adult mobilized peripheral blood.
  • the negative sign or, uppercase negative sign, (-) means that the level of the specified marker is undetectable above Ig isotype controls by FACS analysis, and includes cells with very low expression of the specified marker.
  • stem cells The ability of stem cells to undergo substantial self-renewal as well as the ability to proliferate and differentiate into all of the hematopoietic lineages makes stem cells the target of choice for a number of gene therapy applications.
  • Successful gene transfer into stem cells should provide long-term repopulation of an individual with the modified cells and their progeny, which will express the desired gene product.
  • gene transfer into more mature hematopoietic cells, such as T cells provides only transient therapeutic benefit.
  • Retroviral vectors have been the primary vehicle due to the generally high rate of gene transfer obtained in experiments with cell lines, and the ability to obtain stable integration of the genetic material, which ensures that the progeny of the modified cell will contain the transferred genetic material. Retroviral vectors and their use in the transfer and expression of foreign genes are reviewed in Gilboa (1988) Adv. Exp . Med. Biol . 241 : 29 ; Luskey et al. (1990) Ann . N. Y. Acad. Sci . 612 : 398 ; and Smith (1992) J. Hematother. 1:155-166.
  • retroviral vectors typically in the range of 10 5 to 10 6 infectious virions per milliliter.
  • primitive stem cells typically are quiescent in culture; retroviral vectors require target cells to be cycling for stable integration of the retroviral DNA.
  • Cytokines may be used to cause stem cells to cycle; however, the effect of various cytokines in driving stem cells to differentiation remains in question. Likewise, the effect of more differentiated cells in culture on the growth or
  • mMLV Moloney murine leukemia virus
  • the viral gag, pol and env sequences are removed from the virus, creating room for insertion of foreign DNA sequences. Genes encoded by the foreign DNA are usually expressed under the control of the strong viral promoter in the LTR.
  • Such a construct can be packed into viral particles efficiently if the gag, pol and env functions are provided in trans by a packaging cell line.
  • the gag-pol and env proteins produced by the cell assemble with the vector RNA to produce infectious virions that are secreted into the culture medium.
  • the virus thus produced can infect and integrate into the DNA of the target cell, but does not produce infectious viral particles since it is lacking essential packaging sequences.
  • Most of the packaging cell lines currently in use have been transfected with separate plasmids, each containing one of the necessary coding
  • the packaging cell line harbors an integrated provirus.
  • the provirus has been crippled so that, although it produces all the proteins required to assemble infectious viruses, its own RNA cannot be packaged into virus. Instead, RNA produced from the recombinant virus is packaged. The virus stock released from the packaging cells thus contains only recombinant virus.
  • the range of host cells that may be infected by a retrovirus or retroviral vector is determined by the viral envelope protein.
  • the recombinant virus can be used to infect virtually any other cell type recognized by the env protein provided by the packaging cell, resulting in the integration of the viral genome in the transduced cell and the stable production of the foreign gene product.
  • the efficiency of infection is also related to the level of expression of the receptor on the target cell.
  • murine ecotropic env of MoMLV allows infection of rodent cells
  • amphotropic env allows infection of rodent, avian and some primate cells, including human cells.
  • Amphotropic packaging cell lines for use with MoMLV systems are known in the art and include, but are not limited to, ⁇ AM, PA12, PA317, and ⁇ CRIP. Miller et al. (1985) Mol . Cell . Biol . 5:431-437; Miller et al. (1986) Mol . Cell . Biol.
  • Xenotropic vector systems exist which also allow infection of human cells.
  • the host range of retroviral vectors has been altered by substituting the env protein of the base virus with that of a second virus.
  • the resulting, "pseudotyped" virus has the host range of the virus donating the envelope protein and expressed by the packaging cell line.
  • VSV-G G-glycoprotein from vesicular stomatitis virus
  • VSV-G protein is thought to mediate viral infection by fusing with a phospholipid component of cell membranes rather than by recognition of a cell surface protein. Since infection is not dependent on a specific receptor, VSV-G pseudotyped vectors have a broad host range. Indeed, MoMLV-based retroviral vectors pseudotyped with VSV-G were shown to have a broad host range, infecting a number of cell lines derived from species such as hamster and fish.
  • VSV-G The mechanism of infection mediated by the VSV-G protein, however, has not been elucidated. It has been reported that VSV-G interacts with a phosphatidylserine component of cell membranes, but that observation has yet to be verified. Schlegal et al. (1983) Cell 32:639-646. As a result, it cannot be predicted with any certainty what cells or cell types would be infected by a VSV-G pseudotyped vector, or with what efficiency transduction might occur.
  • the invention provides improved methods of transducing hematopoietic stem cells with pseudotyped
  • retrovirus vectors containing the VSV-G protein comprise using the vectors to transduce a population of hematopoietic cells enriched for stem cells.
  • Figure 1 is a schematic depiction of the plasmid pME-VSV-G.
  • Figure 2 is a schematic depiction of the plasmid MFG-lac-Z.
  • CD34 + Thy-1 + mobilized peripheral blood (MPB) cells are transduced with surprisingly high efficiency by a VSV-G pseudotyped retroviral vector as compared to CD34 + adult bone marrow (ABM) cells and as compared to the transduction efficiency of a conventional amphotropic vector.
  • the improvement appears to be specific to the vector and cell type; an amphotropic vector containing the same viral genome showed a somewhat better transduction efficiency in CD34 + ABM cells compared to CD34 + Thy-1 + MPB cells.
  • the improved transduction efficiency described herein appears to be due either to the more purified stem cell population, represented by CD34 + Thy-1 + Lin- cells as compared to CD34 + cells, or due to the tissue cell source.
  • Stem cells that have been mobilized into the peripheral blood by chemotherapy and/or cytokines may undergo changes in membrane potential
  • VSV-G VSV-G
  • stem cells obtained from MPB may also be suitable for use herein.
  • more primitive hematopoietic cells represented by CD34 + Thy-1 + Lin- cells may be preferentially infected by the pseudotyped vector regardless of tissue source.
  • primitive cells may cycle more when purified from more mature
  • any retroviral vector will transduce pluripotent stem cells more efficiently when a purer stem cell population is transduced as compared to the more heterogenous CD34 + cell population.
  • stem cells will cycle less causing transduction frequency to represent disproportionate transduction of progenitor cells at the expense of stem cells.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • stem cells refers to a population of hematopoietic cells more highly enriched in pluripotent stem cells than the population characterized solely by CD34 expression.
  • the highly enriched stem cell population will typically have a LTCIC frequency in the range of 1/20 to 1/100; preferably it will have a frequency of at least 1/50.
  • Stem cells are exemplified by CD34 + Thy-1 + MPB cells and described more fully in the examples provided herein.
  • Other markers that have been reported to subdivide CD34 + cells, further enriching for stem cells include, but are not limited to, CD38-;
  • rhodamine 10 rhodamine 10 ; c-kit receptor + ; HLADR 10/- ; CD71-; and CD45RA-.
  • stem cells are highly enriched in the CD34 hi Lin- population as described by DiGiusto et al. (1993) Blood 84:421-432.
  • the highly enriched populations of stem cells may be transduced immediately after purification or maintained in long-term cultures and expanded in number in appropriate media, optionally in conjunction with hematopoietic factors such as LIF, stem cell factor, IL3, IL6, IL7, IL11, GCSF, GMCSF, EPO, MIP-lO and IFN ⁇ , under otherwise conventional conditions.
  • hematopoietic factors such as LIF, stem cell factor, IL3, IL6, IL7, IL11, GCSF, GMCSF, EPO, MIP-lO and IFN ⁇ , under otherwise conventional conditions.
  • transduced cells may then be grown under conditions similar to those for unmodified stem cells, whereby the modified stem cells may be expanded and used for a variety of purposes.
  • Stem cells may be isolated from any known source of stem cells, including, but not limited to, bone marrow, both adult and fetal, mobilized peripheral blood (MPB) and
  • bone marrow cells may be obtained from a source of bone marrow, including but not limited to, ilium (e.g. from the hip bone via the iliac crest), tibia, femora, spine, or other bone cavities.
  • Other sources of stem cells include, but are not limited to, embryonic yolk sac, fetal liver, and fetal spleen.
  • an appropriate solution may be used to flush the bone, including, but not limited to, salt solution, conveniently supplemented with fetal calf serum (FCS) or other naturally occurring factors, in conjunction with an acceptable buffer at low FCS.
  • FCS fetal calf serum
  • Convenient buffers include, but are not limited to, HEPES, phosphate buffers and lactate buffers. Otherwise bone marrow may be aspirated from the bone in accordance with conventional techniques.
  • Methods for mobilizing stem cells into the peripheral blood are known in the art and generally involve treatment with chemotherapeutic drugs, cytokines (e.g. GM- CSF, G-CSF or IL3), or combinations thereof.
  • cytokines e.g. GM- CSF, G-CSF or IL3
  • apheresis for total white cells begins when the total white cell count reaches 500-2000 cells/ ⁇ l and the platelet count reaches 50,000/ ⁇ l.
  • Monoclonal antibodies are particularly useful for identifying markers associated with particular cell lineages and/or stages of differentiation.
  • the antibodies may be attached to a solid support to allow for crude separation.
  • the separation techniques employed should maximize the retention of viability of the fraction to be collected.
  • Various techniques of different efficacy may be employed to obtain "relatively crude” separations. Such separations are where up to 10%, usually not more than about 5%, preferably not more than about 1%, of the total cells present not having the marker may remain with the cell population to be retained. The particular technique employed will depend upon efficiency of separation, associated
  • separation techniques include, but are not limited to, those based on differences in physical
  • Procedures for separation may include, but are not limited to, magnetic separation, using antibody-coated magnetic beads, affinity chromatography, cytotoxic agents joined to a monoclonal antibody or used in conjunction with a monoclonal antibody, including, but not limited to, complement and cytotoxins, and "panning" with antibody attached to a solid matrix, e.g., plate, elutriation or any other convenient technique.
  • Techniques providing accurate separation include, but are not limited to, FACS, which can have varying degrees of
  • a large proportion of the differentiated cells may be removed by initially using a relatively crude separation, where major cell population lineages of the hematopoietic system, such as lymphocytic and myelomonocytic, are removed, as well as minor populations, such as megakaryocytic, mast cells, eosinophils and basophils. Usually, at least about 70 to 90 percent of the hematopoietic cells will be removed. If desired, a prior separation may be employed to remove
  • erythrocytes by employing Ficoll-Hypaque separation.
  • the gross separation may be achieved using methods known in the art including, but not limited to, magnetic beads, cytotoxic agents, affinity chromatography or panning.
  • Antibodies which find use include antibodies to lineage specific markers which allow for removal of most, if not all, mature cells, while being absent on stem cells.
  • a negative selection may be carried out, where antibodies to lineage-specific markers present on dedicated cells are employed.
  • these markers include, but are not limited to, CD2-, CD3-, CD7-, CD8-, CD10-, CD14-, CD15-, CD16-, CD19-, CD20-, CD33- and glycophorin A; preferably including, but not limited to, at least CD2-, CD14-, CD15-, CD16-, CD19- and glycophorin A; and normally including at least CD14- and CD15-
  • Lin- refers to a cell population lacking at least one lineage-specific marker.
  • the hematopoietic cell composition substantially depleted of dedicated cells may then be further separated using a marker for Thy-1, whereby a substantially homogeneous stem cell population is achieved.
  • a marker for Thy-1 for Thy-1
  • population is a population which is CD34 + Thy-1 + Lin-, which provides an enriched stem cell composition.
  • the purified stem cells have low side scatter and low to medium forward scatter profiles by FACS analysis.
  • Cytospin preparations show the enriched stem cells to have a size between mature lymphoid cells and mature granulocytes .
  • Cells may be selected based on light-scatter properties as well as their expression of various cell surface antigens.
  • cells are initially separated by a coarse separation, followed by a fine separation
  • compositions highly enriched in stem cells may be achieved in this manner.
  • the desired stem cells are
  • negative selection lineage selection for lineage specific markers provide a greater enrichment in stem cells obtained from bone marrow than from MPB.
  • the majority of CD34 + cells that are mobilized into the peripheral blood do not express lineage-specific markers and, therefore, Lin. selection does not significantly enrich over CD34 + selection in the peripheral blood as it does in bone marrow.
  • Selection for Thy-1 + does enrich for stem cells in both mobilized peripheral blood and bone marrow.
  • a stem cell composition is characterized by being able to be maintained in culture for extended periods of time, being capable of selection and transfer to secondary and higher order cultures, and being capable of
  • myelomonocytic lineages particularly B and T lymphocytes, monocytes, macrophages, neutrophils, erythrocytes and the like.
  • the stem cells may be grown in culture in an appropriate nutrient medium, including, but not limited to, conditioned medium, a co-culture with an appropriate stromal cell line, adhesion molecules, or a medium comprising a synthetic combination of growth factors which are sufficient to maintain the growth of hematopoietic cells.
  • an appropriate nutrient medium including, but not limited to, conditioned medium, a co-culture with an appropriate stromal cell line, adhesion molecules, or a medium comprising a synthetic combination of growth factors which are sufficient to maintain the growth of hematopoietic cells.
  • stromal cell lines For conditioned media or co-cultures, various stromal cell lines may be used. Since human stromal cell lines are not required, other stromal cell lines may be employed, including but not limited to rodentiae,
  • Suitable murine stromal cell lines include AC3 and AC6, which are described in Whitlock et al. (1987) Cell 48 : 1009-1021. Other stromal cell lines may be developed, if desired.
  • the stromal cell line used is a passage of AC6, AC6.21 (otherwise referred to as SySl).
  • Various devices exist for co-culture of stem cells with stromal cells which allow for growth and maintenance of stem cells include devices employing mechanisms including, but not limited to, crossed threads, membranes and controlled medium flow. These may be employed for the growth of the cells for removal of waste products, and replenishment of the various factors associated with cell growth.
  • tissue culture plates or flasks may be employed where confluent stromal cell layers may be maintained for extended periods of time without passage, but with changing of the tissue culture medium about every five to seven days.
  • the stem cells may be grown in co-culture by placing the stem cells onto the stromal cell lines, either directly or separated by a porous membrane. For example, about 3 x 10 4 to 3 x 10 5 cells/ml are placed on a confluent stromal cell layer.
  • the media employed in the co-culture may be any convenient growth medium, including, but not limited to, RPMI-1640 and IMDM either individually or in combination, where appropriate antibiotics to prevent bacterial growth, e.g. penicillin, streptomycin (pen/strep) and other
  • Cytokines may also be added, including, but not limited to, LIF, interleukins, colony stimulating factors, steel factor. Of particular interest are LIF, steel factor, IL-3, IL-6, GM-CSF, G-CSF and MIP-lo.
  • the factors which are employed may be naturally occurring or synthetic, e . g. prepared recombinantly, and may be human or of other species, e . g. murine, preferably human.
  • the amount of the factors will generally be in the range of about 1 ng/ml to 100 ng/ml.
  • the factors for LIF, the
  • concentration will be in the range of about 1 ng/ml to 100 ng/mg, more usually 5 ng/ml to 30 ng/ml; for IL-3, the concentration will be in the range of about 5 ng/ml to 50 ng/ml, more usually 5 ng/ml to 100 ng/ml; for IL-6, the concentration will be in the range of about 5 ng/ml to 50 ng/ml, more usually 5 ng/ml to 20 ng/ml, and for GM-CSF, the concentration will generally be 5 ng/ml to 50 ng/ml, more usually 5 ng/ml to 20 ng/ml.
  • the stem cells are optionally expanded prior to or after transduction.
  • the growth factors may be present only during the initial course of the stem cell growth and expansion, usually at least 24 hours, more usually at least about 48 hours to 4 days or may be maintained during the course of the expansion.
  • the stem cells are cultured with or without cytokines in an appropriate medium, transduced with the appropriate vector, cultured for
  • Gene transfer into stem cells may be used to treat a variety of neoplastic, infectious or genetic diseases.
  • the mdrl gene may be introduced into stem cells to provide increased resistance to a wide variety of drugs including taxol, which are exported by the mdrl gene product, in combination with the administration of chemotherapeutics such as taxol, e.g. for breast cancer treatment.
  • genes that provide increased resistance to alkylating agents, such as melphalan may be introduced into stem cells in
  • Genes that provide resistance to alkylating agents include, but are not limited to, glutathione-S-transferase and methylpurine DNA
  • adenosine deaminase for resistance to purine analogs
  • 06- alkylguanine-DNA alkyltransferase for resistance to N-methyl- N-nitrosurea
  • methylguanine methyl transferase for resistance to nitrosurea BCNU
  • dihydrofolate reductase for resistance to methotrexate
  • methylpurine DNA glycosylate for resistance to radiation
  • glutacyanase transferase for resistance to platinum analogs .
  • stem cells may be modified to endow the progeny with resistance to the infectious agent.
  • HAV human immunodeficiency virus
  • specific antisense or ribozyme sequences may be introduced that interfere with viral infection or replication in the target cells.
  • the introduced gene products may serve as "decoys" by binding essential viral proteins, thereby interfering with the normal viral life cycle and inhibiting replication.
  • Introduction of the apoptosis modulating genes such as bcr-abl into stem cells may provide resistance to the cell death by apoptosis associated with HIV infection.
  • stem cells may be modified to produce a product to correct a genetic deficiency, or where the host has acquired a genetic deficiency through a
  • deficiency include, but are not limited to, adenosine
  • glucocerebrosidase for the treatment of Gaucher's disease
  • beta-globin for the treatment of sickle cell anemia
  • Factor VIII or Factor IX for the treatment of hemophilia.
  • Suitable viral constructs are discussed in Burns et al. (1993) and PCT application no. WO 92/14829 although any functional pseudotype virus containing the VSV-G gene and capable of being packaged by a packaging cell line is
  • the viral constructs employed will normally include the VSV-G gene, the foreign gene(s) and a marker gene, which allows for selection of cells into which the DNA has been integrated, as against cells which have not integrated the DNA construct.
  • Various marker genes include, but are not limited to, antibiotic resistance markers, such as resistance to G418 or hygromycin. Less conveniently, negative selection may be used, including, but not limited to, where the marker is the HSV-tk gene, which will make the cells sensitive to agents such as acyclovir and gancyclovir.
  • selections could be accomplished by employment of a stable cell surface marker to select for transgene expressing stem cells by FACS sorting.
  • the viral constructs can be prepared in a variety of conventional ways. Numerous vectors are now available which provide the desired features, such as long terminal repeats, marker genes, and restriction sites, which may be further modified by techniques known in the art.
  • the desired features such as long terminal repeats, marker genes, and restriction sites, which may be further modified by techniques known in the art.
  • constructs may encode a signal peptide sequence to ensure that genes encoding cell surface or secreted proteins are properly processed post-translationally and expressed on the cell surface if appropriate.
  • the foreign gene(s) is under the control of a cell specific promoter as discussed below.
  • a particular polymorphic region of a polymorphic protein such as a T-cell receptor, major histocompatibility complex antigen, or immunoglobulin subunit is involved with susceptibility to a particular disease, for example an autoimmune disease
  • the particular exon may be "knocked out” by homologous recombination, so as to provide hematopoietic cells which will not be responsive to the disease.
  • the introduced gene may be put under the control of a promoter that will cause the gene to be expressed constitutively, only under specific physiologic conditions, or in particular cell types.
  • promoters that may be used to cause expression of the introduced sequence in specific cell types include
  • Granzyme A for expression in T-cells and NK cells the CD34 promoter for expression in stem and progenitor cells, the CD8 promoter for expression in cytotoxic T-cells, and the CDllb promoter for expression in myeloid cells. Inducible
  • promoters may be used for gene expression under certain physiologic conditions.
  • an electrophile for example, an electrophile
  • response element may be used to induce expression of a chemoresistance gene in response to electrophilic molecules.
  • the therapeutic benefit may be further increased by targeting the gene product to the appropriate cellular location, for example the nucleus, by attaching the appropriate localizing sequences.
  • inducible promoters expression of various protein products can be achieved at selected levels of differentiation or in selected cell lineages, or even in response to particular chemicals, such as chemoattractants, particular ligands, and the like.
  • particular lineages such as megakaryocytes, subsets of T cells, monocytes, and the like can be produced in culture.
  • Possible methods of transduction include, but are not limited to, direct co-culture of stem cells with producer cells e.g. by the method of Bregni et al. (1992) Blood
  • cell immunotherapy involves removal of bone marrow or other source of stem cells from a human host, isolating the stem cells from the source and optionally expanding the stem cells. Meanwhile, the host may be treated to partially, substantially or completely ablate native hematopoietic capability. The isolated stem cells may be modified during this period of time, so as to provide for stem cells having the desired genetic modification. After completion of the treatment of the host, the modified stem cells may then be restored to the host to provide for
  • stem cell removal The methods of stem cell removal, host ablation and stem cell repopulation are known in the art. If necessary, the process may be repeated to ensure substantial repopulation of the modified stem cells.
  • a vector-specific probe may be used to verify the presence of the vector in the transduced stem cells or their progeny.
  • the cells may be grown under various conditions to ensure that they are capable of maturation to all of the hematopoietic lineages while maintaining the capability, as appropriate, of the introduced DNA.
  • Various tests in vi tro and in vivo may be employed to ensure that the pluripotent capability of the stem cells has been maintained.
  • compositions comprising stem cells provide for production of myeloid cells and lymphoid cells in appropriate cultures, cultures providing hydrocortisone for production of myeloid cells (associated with Dexter-type cultures) and B lymphocytes in cultures lacking hydrocortisone, (associated with Whitlock-Witte type cultures).
  • mouse or human stromal cells are provided, which may come from various sources, including, but not limited to, AC3, AC6 or stromal cells derived from mouse or human FBM by selection for the ability to maintain stem cells, and the like.
  • the stromal cells are AC6.21 and the ability to produce B lymphocytes and myeloid cells is determined in cultures supplied with LIF and IL-6.
  • the stem cells give rise to B cells, T cells and myelomonocytic cells in the in vivo assays described below.
  • fetal thymus is isolated and cultured from 4-7 days at about 25°C, so as to deplete substantially the lymphoid population.
  • the cells to be tested for T cell activity are then microinjected into the thymus tissue, where the HLA of the population which is injected is mismatched with the HLA of the thymus cells.
  • the thymus tissue may then be transplanted into a scid/scid mouse as described in US Patent No. 5,147,784, particularly transplanting under the kidney capsule.
  • the population of stem cells can be microinjected into HLA mismatched thymus fragments. After 6- 10 weeks, assays of the thymus fragments injected with stem cells can be performed and assessed for donor- derived T cells. Injected thymus fragments injected with cells having T cell differentiative capacity will generate and sustain CD3 + , CD4 + , and CD8 + T cells along with their progenitors.
  • BFU-E units for example methylcellulose culture demonstrating that the cells are capable of developing the erythroid lineage. Metcalf (1977) In: Recent Results in Cancer Research 61. Springer-Verlag, Berlin, pp. 1-227.
  • a pluripotent stem cell may be defined as follows: (1) gives rise to progeny in all defined hematolymphoid lineages; and (2) limiting numbers of cells are capable of fully reconstituting a seriously immunocompromised human host in all blood cell types and their progenitors, including the pluripotent hematopoietic stem cell by self renewal.
  • GM-CSF granulocyte macrophage colony stimulating factor
  • Antibodies to CD2, CD14, CD15, CD16 and CD19 were obtained as FITC conjugates from Becton-Dickinson.
  • Antibody to Thy-1 (GM201) was obtained from Dr. Wolfgang Rettig
  • the antibody to glycophorin A was obtained as a FITC conjugate from AMAC.
  • the lineage cocktail was a combination of antibodies to CD2, CD14, CD15, CD16, CD19 and glycophorin A.
  • Fractions 2 and 3 were pooled and incubated with 1 mg/ml heat-inactivated human gamma-globulin to block non-specific Fc binding.
  • Granulocytes were further depleted by incubation with CD15 conjugated to magnetic beads (Dynal M450, Oslo, Norway) followed by magnetic selection.
  • Anti-CD34 antibody or an IgG3 isotype matched control were added to cells in staining buffer (HBSS, 2% FCS, 10 mM HEPES) for 20 minutes on ice, together with anti-Thy-1 antibody at 5 ⁇ g/ml.
  • Cells were washed with a FCS underlay, and then incubated with Texas Red conjugated goat anti-mouse IgG3 antibody and phycoerythrin-conjugated goat anti-mouse IgGl antibody for 20 minutes on ice. Blocking IgGl was then added for 10 minutes. After blocking, the FITC-conjugated lineage antibody panel was added, and incubated for another 20 minutes on ice. After a final washing, cells were resuspended in staining buffer containing propidium iodide
  • CD34 + cells were positively selected from cadaveric adult bone marrow (ABM) using a biotinylated anti-CD34 antibody (K6.1) and a biotin competition release according to the method described in PCT patent application no.
  • amphotropic virus used was MFG-lacZ.
  • MFG vectors are described in Dranoff et al. (1993) Proc. Natl. Acad, Sci, USA 90 : 3539-3543.
  • Amphotropic MFG-lacZ was obtained as viral supernatant from Somatix Therapy
  • VSV-G pseudotype virus was prepared using a cDNA clone of the New Jersey isolate of the VSV-G envelope protein as the source of the VSV-G gene. Rose and Bergmann (1982) Cell 30:753-762.
  • the VSV-G gene was subcloned by polymerase chain reaction (PCR) on a Cetus GeneAmp 9600 machine using 100 ng plasmid; 10 ng each primer and 10 cycles of the following: 30 sec 92°C, 30 sec 55°C, and 1 min 72°C.
  • PCR polymerase chain reaction
  • Primer No. 1 was the 5' primer and generated an EcoRI site
  • Primer No. 2 was the 3' primer and generated an Xbal site.
  • the resulting fragment was digested with EcoRI and Xbal and cloned into plasmid pME-18S digested with EcoRI /Xbal to generate plasmid pME-VSV-G.
  • pME-18S was obtained from DNAX; plasmid pME-VSV-G is depicted in Figure 1. Plasmid MFG-lacZ is depicted in Figure 2.
  • the pseudotyped virus was prepared by transducing the ANJOU cell line according to the method described by Pear et al. (1993) Proc. Natl. Acad. Sci. USA 90: 8392-8396.
  • ANJOU cells used in transfection were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% (v/v) FCS, 2 mM 1-glutamine, penicillin (100 U/ml), and
  • NIH3T3 cells were maintained in similar media but containing 10% (v/v) calf serum instead of FCS.
  • ANJOU cells were plated at 10 7 /15 cm plate 18 hours prior to transfection.
  • transfection solution (Pear et al. (1993)) was prepared with 30 ⁇ g MFG-LacZ and 30 ⁇ g pME-VSV-G and added dropwise to cells in media containing 25 ⁇ g/ml chloroquine to increase transfection efficiency. Six to 8 hours later the media was changed to 10 ml fresh DMEM, cells were grown an additional 10 hours and the media changed again. Viral supernatants were collected 48 hours after transfection and either (1) used to infect target cells, (2) frozen down at -20°C, or (3) concentrated as discussed below. ANJOU cells were fixed in a 2% paraformaldehyde/0.2% glutaraldehyde solution and stained with X-Gal to determine the transfection efficiency. Pear et al. (1993). The virus obtained was designated VSV-G- MFG-lacZ.
  • the viruses were titered on target cells plated in 3 ml of appropriate media in 6 cm plates at 2x10 5 cells per plate to achieve 60-73% confluence at the time of infection. Eighteen to twenty-four hours after seeding, the media was supplemented with 5 ⁇ g/ml polybrene and virus was added immediately afterward. Media was changed to 6 ml of fresh media 8 hours after addition of virus to avoid polybrene toxicity. Forty-eight hours after infection, the media was removed and cells were assayed for ⁇ -galactosidase activity by X-gal or FACS-Gal according to the method described by Nolan et al. (1988) Proc. Natl. Acad. Sci. USA 85: 2603-260 for determination of viral titers. The titration results are shown in column 3 of Table 2.
  • Example 3 using concentrated, frozen, virus preparations.
  • Example 3 using concentrated, frozen, virus preparations.
  • CD34 + Thy-1 + Lin- (MPB) cells obtained as described in Example 1 were transduced as described above except that the cells were incubated with the retroviral supernatant for 16 hours without change of supernatant. The results of the 16 hour transduction are shown in Tables 2 and 3.
  • the lysates were amplified by PCR to determine the presence of the vector in the transduced cells.
  • the PCR assay amplified a 539 bp fragment from MFG-lacZ.
  • a 40 cycle amplification (50 ⁇ l total volume) in Perkin-Elmer 480 Cycler using 25 ⁇ l lysate and 100 ng each LZ 4927 and LZ 5466
  • a second, nested amplification was performed using a 30 cycle amplification in PE cycler using 5 ⁇ l of primary PCR product and 100 ng each of LZ4953 and LZ5425 (Keystone) Econopure primers.
  • transduction frequency represented in the final column represents a minimum transduction frequency since it assumes that, in a positive sample, only one colony of each pooled sample was positive. At higher transduction
  • amphotropic vector The observed differences are not due to patient variability or variation in the stem cell preparation since, in each case, the MPB samples from a single

Abstract

L'invention concerne des procédés permettant de modifier génétiquement des cellules souches hématopoïétiques au moyen de vecteurs viraux portant la glycoprotéine G du virus de la stomatite vésiculaire.
PCT/US1995/011892 1994-09-19 1995-09-18 Procedes permettant de modifier genetiquement des cellules souches hematopoietiques WO1996009400A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU36356/95A AU3635695A (en) 1994-09-19 1995-09-18 Methods for genetically modifying hematopoietic stem cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30902694A 1994-09-19 1994-09-19
US08/309,026 1994-09-19

Publications (1)

Publication Number Publication Date
WO1996009400A1 true WO1996009400A1 (fr) 1996-03-28

Family

ID=23196354

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/011892 WO1996009400A1 (fr) 1994-09-19 1995-09-18 Procedes permettant de modifier genetiquement des cellules souches hematopoietiques

Country Status (2)

Country Link
AU (1) AU3635695A (fr)
WO (1) WO1996009400A1 (fr)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048815A2 (fr) * 1996-06-17 1997-12-24 Novartis Ag Methode pour ameliorer le transfert de genes
WO1998012306A1 (fr) * 1996-09-23 1998-03-26 Ontogeny, Inc. Cellules souches hematopoietiques et procedes relatifs a leur production
WO1998053063A2 (fr) * 1997-05-16 1998-11-26 Leuven Research & Development Vzw Transduction de cellules de mammiferes utilisee en therapie genique
WO1998055607A2 (fr) 1997-06-04 1998-12-10 Oxford Biomedica (Uk) Limited Vecteur
EP0938904A1 (fr) * 1998-02-09 1999-09-01 Leuven Research & Development vzw Transduction des cellules mammifères pour utilisation dans la thérapie génique
WO1999061639A2 (fr) * 1998-05-22 1999-12-02 Oxford Biomedica (Uk) Limited Systeme d'apport retroviral
WO1999061644A1 (fr) * 1998-05-29 1999-12-02 Case Western Reserve University Transduction genique des cellules progenitrices hematopoietiques
WO2003064665A2 (fr) 2002-02-01 2003-08-07 Oxford Biomedica (Uk) Limited Vecteur viral
US6818209B1 (en) 1998-05-22 2004-11-16 Oxford Biomedica (Uk) Limited Retroviral delivery system
US6969598B2 (en) 2001-04-30 2005-11-29 Oxford Biomedica (Uk) Limited Methods for producing high titre vectors and compositions used in such methods
EP1624899A2 (fr) * 2003-05-05 2006-02-15 VIRxSYS Corporation Amelioration de la transduction au moyen de substrats et/ou d'inhibiteurs du transporteur abc
EP2045268A1 (fr) 2005-05-13 2009-04-08 Oxford BioMedica (UK) Limited Antigène peptide mhc classe I et II dérivé d'une tumeur antigène 5t4
US7541343B2 (en) 2001-05-22 2009-06-02 Newsouth Innovations Pty Limited Inhibiting cellular proliferation by expressing yin yang-1
EP2180057A1 (fr) 2000-10-06 2010-04-28 Oxford Biomedica (UK) Limited Système de vecteur retroviral
EP2194137A2 (fr) 2000-04-19 2010-06-09 Oxford BioMedica (UK) Limited Cellules comprenant des particules rétrovirales avec des codons optimisés
WO2010085660A2 (fr) 2009-01-23 2010-07-29 Roger Williams Hospital Vecteurs viraux codant pour de multiples polypeptides non viraux très homologues et leur utilisation
WO2012075337A2 (fr) 2010-12-01 2012-06-07 Spinal Modulation, Inc. Administration dirigée d'agents à une anatomie neuronale
EP3587582A1 (fr) 2013-10-24 2020-01-01 Adaptimmune Limited Vecteurs d'expression transgénique
DE102020111571A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren
DE102020106710A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988008450A1 (fr) * 1987-05-01 1988-11-03 Birdwell Finlayson Therapie genetique pour troubles du metabolisme
WO1992014829A1 (fr) * 1991-02-19 1992-09-03 The Regents Of The University Of California Particules virales agissant sur une gamme d'hotes modifiee
WO1993014188A1 (fr) * 1992-01-17 1993-07-22 The Regents Of The University Of Michigan Virus cible
WO1993018137A1 (fr) * 1992-03-04 1993-09-16 Systemix, Inc. Culture de cellules souches hematopoietiques et leur preparation par genie genetique
WO1994029438A1 (fr) * 1993-06-11 1994-12-22 Cell Genesys, Inc. Procede de production d'un virus a titre eleve et transduction a haut rendement de cellules mammiferes induite par retrovirus

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988008450A1 (fr) * 1987-05-01 1988-11-03 Birdwell Finlayson Therapie genetique pour troubles du metabolisme
WO1992014829A1 (fr) * 1991-02-19 1992-09-03 The Regents Of The University Of California Particules virales agissant sur une gamme d'hotes modifiee
WO1993014188A1 (fr) * 1992-01-17 1993-07-22 The Regents Of The University Of Michigan Virus cible
WO1993018137A1 (fr) * 1992-03-04 1993-09-16 Systemix, Inc. Culture de cellules souches hematopoietiques et leur preparation par genie genetique
WO1994029438A1 (fr) * 1993-06-11 1994-12-22 Cell Genesys, Inc. Procede de production d'un virus a titre eleve et transduction a haut rendement de cellules mammiferes induite par retrovirus

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5928638A (en) * 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
WO1997048815A3 (fr) * 1996-06-17 1998-03-26 Ciba Geigy Ag Methode pour ameliorer le transfert de genes
WO1997048815A2 (fr) * 1996-06-17 1997-12-24 Novartis Ag Methode pour ameliorer le transfert de genes
WO1998012306A1 (fr) * 1996-09-23 1998-03-26 Ontogeny, Inc. Cellules souches hematopoietiques et procedes relatifs a leur production
WO1998053063A2 (fr) * 1997-05-16 1998-11-26 Leuven Research & Development Vzw Transduction de cellules de mammiferes utilisee en therapie genique
WO1998053063A3 (fr) * 1997-05-16 1999-03-18 Leuven Res & Dev Vzw Transduction de cellules de mammiferes utilisee en therapie genique
WO1998055607A2 (fr) 1997-06-04 1998-12-10 Oxford Biomedica (Uk) Limited Vecteur
EP0938904A1 (fr) * 1998-02-09 1999-09-01 Leuven Research & Development vzw Transduction des cellules mammifères pour utilisation dans la thérapie génique
WO1999061639A2 (fr) * 1998-05-22 1999-12-02 Oxford Biomedica (Uk) Limited Systeme d'apport retroviral
WO1999061639A3 (fr) * 1998-05-22 2000-01-27 Oxford Biomedica Ltd Systeme d'apport retroviral
GB2351290A (en) * 1998-05-22 2000-12-27 Oxford Biomedica Ltd Retroviral delivery sytem
US6818209B1 (en) 1998-05-22 2004-11-16 Oxford Biomedica (Uk) Limited Retroviral delivery system
WO1999061644A1 (fr) * 1998-05-29 1999-12-02 Case Western Reserve University Transduction genique des cellules progenitrices hematopoietiques
EP2194137A2 (fr) 2000-04-19 2010-06-09 Oxford BioMedica (UK) Limited Cellules comprenant des particules rétrovirales avec des codons optimisés
EP2180057A1 (fr) 2000-10-06 2010-04-28 Oxford Biomedica (UK) Limited Système de vecteur retroviral
US6969598B2 (en) 2001-04-30 2005-11-29 Oxford Biomedica (Uk) Limited Methods for producing high titre vectors and compositions used in such methods
US7541343B2 (en) 2001-05-22 2009-06-02 Newsouth Innovations Pty Limited Inhibiting cellular proliferation by expressing yin yang-1
EP2348119A2 (fr) 2002-02-01 2011-07-27 Oxford BioMedica (UK) Limited Vecteur de lentivirus multicistronique
WO2003064665A2 (fr) 2002-02-01 2003-08-07 Oxford Biomedica (Uk) Limited Vecteur viral
EP1624899A4 (fr) * 2003-05-05 2006-05-10 Virxsys Corp Amelioration de la transduction au moyen de substrats et/ou d'inhibiteurs du transporteur abc
EP1624899A2 (fr) * 2003-05-05 2006-02-15 VIRxSYS Corporation Amelioration de la transduction au moyen de substrats et/ou d'inhibiteurs du transporteur abc
EP2277918A2 (fr) 2005-05-13 2011-01-26 Oxford BioMedica (UK) Limited Antigène peptide mhc classe I et II dérivé d'une tumeur antigène 5t4
EP2045268A1 (fr) 2005-05-13 2009-04-08 Oxford BioMedica (UK) Limited Antigène peptide mhc classe I et II dérivé d'une tumeur antigène 5t4
WO2010085660A2 (fr) 2009-01-23 2010-07-29 Roger Williams Hospital Vecteurs viraux codant pour de multiples polypeptides non viraux très homologues et leur utilisation
WO2012075337A2 (fr) 2010-12-01 2012-06-07 Spinal Modulation, Inc. Administration dirigée d'agents à une anatomie neuronale
EP3587582A1 (fr) 2013-10-24 2020-01-01 Adaptimmune Limited Vecteurs d'expression transgénique
DE102020111571A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren
DE102020106710A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren
WO2021183643A1 (fr) 2020-03-11 2021-09-16 Immatics US, Inc. Constructions mutantes wpre, compositions et procédés associés

Also Published As

Publication number Publication date
AU3635695A (en) 1996-04-09

Similar Documents

Publication Publication Date Title
WO1996009400A1 (fr) Procedes permettant de modifier genetiquement des cellules souches hematopoietiques
US7416887B2 (en) Methods for use of MPL ligands with primitive human stem cells
US5928638A (en) Methods for gene transfer
Cavazzana-Calvo et al. Role of interleukin-2 (IL-2), IL-7, and IL-15 in natural killer cell differentiation from cord blood hematopoietic progenitor cells and from gamma c transduced severe combined immunodeficiency X1 bone marrow cells
AU679120B2 (en) Genetically modified human hematopoietic stem cells and their progeny
KR20160075676A (ko) 방법
EP1053302B1 (fr) Populations multipliees et genetiquement modifiees de cellules souches hematopoietiques humaines
US20030044978A1 (en) Expanded and genetically modified populations of human hematopoietic stem cells
WO1996033281A1 (fr) Transduction ex vivo a productivite elevee de cellules souches hematopoietiques au moyen de preparations retrovirales xenotropes recombinees
WO1997012052A1 (fr) Transduction de cellules hematopoietiques par vecteur viral et lipide cationique
AU2002301459B2 (en) Expanded and genetically modified populations of human hematopoietic stem cells
AU2003224985A1 (en) Enhancement of hematopoietic stem cell survival
Bierhuizen et al. Efficient detection and selection of immature rhesus monkey and human CD34+ hematopoietic cells expressing the enhanced green fluorescent protein (EGFP)
Jurecic et al. Human Hematopoietic Cells for Gene Therapy
Horwitz et al. Gene therapy for hematopoietic disorders
Bank et al. Transfer of the MDR-1 gene into hematopoietic cells
Mostoslavsky et al. 274. Lentiviral Gene Transfer into Hematopoietic Stem Cells: Getting the Best out of It

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TT UA UG UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase