US20060093581A1 - Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses - Google Patents

Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses Download PDF

Info

Publication number
US20060093581A1
US20060093581A1 US11/295,596 US29559605A US2006093581A1 US 20060093581 A1 US20060093581 A1 US 20060093581A1 US 29559605 A US29559605 A US 29559605A US 2006093581 A1 US2006093581 A1 US 2006093581A1
Authority
US
United States
Prior art keywords
cells
dendritic cells
human
lineage committed
patient
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/295,596
Inventor
Alan Smith
Douglas Smith
Ramkumar Mandalam
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vericel Corp
Original Assignee
Aastrom Biosciences Inc
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 Aastrom Biosciences Inc filed Critical Aastrom Biosciences Inc
Priority to US11/295,596 priority Critical patent/US20060093581A1/en
Publication of US20060093581A1 publication Critical patent/US20060093581A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0655Chondrocytes; Cartilage
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464838Viral antigens
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/25Tumour necrosing factors [TNF]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/59Lectins

Definitions

  • the present invention relates to methods for culturing lineage committed human cells, the cells-thus obtained, and their uses.
  • tissue engineering the goal is to reconstitute fully or partially functioning human tissue in vitro to enable a variety of clinical and other applications.
  • Several studies have been carried out that are aimed at reconstituting functioning human tissues in vitro. Illustratively, the cultivation of human skin has been successful.
  • the hematopoietic system exemplifies the broad range of cells involved in protection of mammalian hosts against pathogens toxins, neoplastic cells, and other diseases.
  • the hematopoietic system is believed to evolve from a single stem cell, from which all the lineages of the hematopoietic system derive.
  • Hematopoietic cells have been used in human therapy. Methods and apperati for culturing precursor hematopoietic cells to obtain desired mature hematopoietic cells have been described. See, U.S. Pat. Nos. 5,605,822, 5,399,493; 5,437,994; 5,459,069; 5,635,386, 5,670,147 and 5,670,351.
  • Adoptive cell therapy is the ex vivo expansion and re-infusion of immune effector cells into human recipients for the treatment or prevention of disease (Rosenberg et al., Science 233, 1318 (1986)).
  • Developments in T-lymphocyte-based adoptive immunotherapy have lead to significant advances in the treatment of metastatic cancer (Rosenberg et al., Science 233, 1318 (1986); Rosenberg et al., N. Eng. J. Med. 319, 1676 (1988); Rosenberg et al., N. Eng. J. Med. 323, (1990)) and viral diseases including Epstein-Barr Virus (EBV) (Heslop et al.
  • EBV Epstein-Barr Virus
  • T-cell therapy has achieved a response rate of approximately 34% after administration of tumor infiltrating lymphocytes (TILs) to metastatic melanoma patients (Rosenberg et al., J. Natl. Cancer Inst. 86, 1159 (1994)).
  • TILs tumor infiltrating lymphocytes
  • TILs A target human therapeutic dose of 10 10 -10 11 TILs is suggested based on extrapolation from studies in mouse tumor models and clinical experience (Topalian et al., J. Immunol. Meth. 102, 107 (1987)). Clinical responses to melanoma tumors have been obtained after expansion ex vivo of TILs in medium containing interleukin-2 (IL-2) and re-infusion of greater than 10 11 T-cells per treatment cycle together with high doses of exogenous IL-2 (Rosenberg et al., N. Eng. J. Med. 319, 1676 (1988); Rosenberg et al. 1994).
  • IL-2 interleukin-2
  • the enhanced anti-tumor activity of TILs in vivo compared to lymphokine activated killer (LAK) cells or NK cells may be a function of several complex processes including the ability of these T-cells to proliferate and release an array of lymphokines, to recirculate and accumulate at sites of tumor growth, and to specifically recognize and lyse autologous tumor cells (Pockaj et al. 1994; Rosenberg et al. 1994).
  • Dendritic cells are highly specialized bone-marrow derived antigen presenting cells (APCs) which function as potent stimulating cells for primary T-lymphocyte-mediated immune response.
  • APCs bone-marrow derived antigen presenting cells
  • Dendritic cell based immunotherapy is an emerging treatment strategy involving ex vivo expansion and reinfusion of antigen-pulsed or genetically modified DCs into human patients to vaccinate against cancer or infectious diseases. Clinical implementation of these therapies requires the capability of product sufficient quantities of functional DCs for effective patient treatment.
  • cultured human cells in human therapy has required that a quantity of active cells sufficient to provide a therapeutic effect upon infusion into the patient be used. This has required using culture systems providing large numbers of cells. There is therefore a need for methods for obtaining lineage committed cells with augmented proliferative potential, biological function, or both since such methods would provide more potent cell compositions, capable of being used in smaller amounts in therapy.
  • lineage committed human cells having enhanced proliferative potential, biological function, or both.
  • lineage committed human cells are cultured in any physiologically acceptable liquid culture medium conditions with the liquid culture medium being replaced.
  • FIG. 1 shows the amount of cytokine produced after stimulation of T-cells cultured using continuous medium exchange conditions according to the present invention and cells cultured under standard static low density medium conditions.
  • AIM V represents a control with no added stimulus, all experiments were cultured with AIM V culture medium.
  • OKT3 represents anti-CD3 mAb.
  • D IL-10 production.
  • FIG. 2 shows (A) the lactate concentration during a dendritic cell culture according to the present invention (100% exchange) and in a static culture (0% medium exchange) at low inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • A the lactate concentration during a dendritic cell culture according to the present invention (100% exchange) and in a static culture (0% medium exchange) at low inoculum density
  • MLR mixed leucocyte response
  • FIG. 3 shows (A) the lactate concentration during a dendritic cell culture according to the present invention (150% exchange) and in a static culture (0% medium exchange) at intermediate inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • FIG. 4 shows (A) the lactate concentration during a dendritic cell culture according to the present invention at 100% exchange and 300% medium exchange at high inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • MLR mixed leucocyte response
  • FIG. 5 shows the effect of medium exchange rate of canine chondrocyte production at different inoculum densitites.
  • the present invention applies to the culture of lineage committed human cells using any known culture techniques.
  • the invention is based on the discovery that by replacing the liquid culture media in any lineage committed human cell culture without substantially changing the cell density, it is possible to obtain cells having an enhanced proliferative potential, an enhanced biological function, or both.
  • the cells obtained in accordance with the invention have enhanced proliferative potential as compared to the cells prior to culture and/or as compared to cells one obtains using otherwise identical culture conditions except that the liquid culture media is replaced by diluting the culture to achieve a lower cell density.
  • the biological function of the cells obtained in accordance with the invention is enhanced as compared with the biological function of these same cells prior to culture and/or as compared to the biological function of the same type of cells cultured under otherwise identical culture conditions except that the liquid medium is not replaced.
  • the cells one obtains in accordance with the invention are, by virtue of their enhanced proliferative potential and/or biological effector function, more potent.
  • the present invention permits using a smaller number of lineage committed human cells to obtain similar or better results than one obtains using lineage committed human cells cultured in accordance with techniques existing prior to the present invention.
  • the lineage committed human cells one obtains in accordance with the invention can be used in any application in which lineage committed human cells are used, including, but not limited to, therapy treatments including adoptive immunotherapy with effector cells, tumor specific cytotoxic T-cells, infectious disease specific cytotoxic T lymphocytes, cytokine induced killer cell therapy, antigen presenting cells to either tumor or infectious diseases, dendritic cells, antigen primed, dendritic cells, tumor vaccines, genetically modified attenuated tumor cells, genetically modified antigen presenting cells, structural repair procedures such as cartilage defect repair, bone defect repair, tissue repair, would healing, burn care, solid organ repair, neurological defect repair, etc. Derivation and expansion of antigen specific T-cell populations including, but not limited to, viral (e.g.
  • TILs human tumor infiltrating lymphocytes
  • CIK cytokine induced killer cells
  • immunotherapy after autologous or allogeneic bone marrow transplantation.
  • dendritic cells antigen presenting cell (APC)-based vaccines to stimulate T-cell responses in vitro or in vivo against simple or complex antigens including, but not limited to, tumor, viral, fungal and bacterial antigens.
  • Therapy may include treatment or prevention of disease in normal individuals or human cancer patients.
  • the lineage committed human cell used in accordance with the present invention are cells which are differentiated to at least a point where they are programmed to develop into a specific type of cell. These cells are not necessarily terminally differentiated. For example, CFU-GEMM cells are committed to develop, ultimately, into mature myeloid cells. Similarly, CFU-L cells are committed to develop into lymphoid cells. Accordingly, the lineage committed cells may be multipotential stem cells or committed progenitor cells. Of course, the lineage committed cells may be mature cells derived from these precursors. As used herein, the term “mature cells” refers to-cells which are terminally differentiated; In one embodiment, the lineage committed cells are more differentiated than human stem cells.
  • the lineage committed cells are more differentiated than progenitor cells. In yet another embodiment, the lineage committed cells are mature cells.
  • a discussion of cell development for blood cells and lymphocytes is provided by S. McKenzie, Hematology, Second Edition, William and Wilkins, 1996, pp. 9-30 and 55-89.
  • lineage committed cells used in the present method may vary widely. Any type of lineage committed cell which may benefit from medium replacement may be used in the present invention.
  • Preferred cells which can be used include human hematopoietic cells, mesenchymal cells, dendritic cells, fibroblasts, hepatocytes, neural cells, epithelial cells, lymphocytes, keratinocytes, osteoblasts or osteoclasts.
  • suitable human hematopoietic cells include megakaryocytes, monocytes, neutrophils, basophils, eosinophils, tumor specific cytotoxic T lymphocytes, cytokine induced killer cells, antigen presenting cells to either tumor or infectious diseases, dendritic cells, antigen primed dendritic cells, leukocyte precursor, and neutrophils.
  • Suitable examples of mesenchymal cells include, chondrocytes, osteoblasts, myeoblasts, fibroblasts, tenoblasts, stromal cells (e.g., from bone marrow), tenocytes, adipocytes, osteocytes and myocytes.
  • Suitable examples of lymphocytes include T-cells and B-cells. Pre-T and pre-B cells are also suitable.
  • the T-cells (CD3 + ) may be CD8 + or CD4 + cells, or cells derived from said populations.
  • the lineage committed cells may be stem cells (e.g., hematopoietic or stromal stem cells), progenitor cells (e.g., hematopoietic progenitors), mature myeloid cells, or stromal cells (e.g., from bone marrow).
  • the cells may be dendritic cells (e.g., myeloid- or lymphoid-derived) or non-myeloid mature cells which are other than stromal cells (e.g., the mature cells described above, especially T-cells or chondrocytes).
  • the cells used in the present method may be obtained from a variety of sources using well-known techniques, see Heslop H E, Ng Cyc, Li C, Smith C A, Loftin S K, Krance Ra, Brenner M K, Rooney C M: Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nature Med 2:551, 1996; Lu P, Negrin RS: A novel population of expanded human CD3+CD56+ cells derived from T-cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency.
  • a human hematopoietic cell composition enriched in T-cells may be used.
  • Such a composition may be enriched in T-cell content by any desired amount, such as by up to 10 3 fold or more.
  • Different, known methods may be used to achieve this enrichment, corresponding either to a negative selection method or a positive selection method.
  • mature cells are isolated using immunological techniques, e.g.,. labeling non-progenitor, non-stem cells with a panel of mouse anti-human monoclonal antibodies, then removing the mouse antibody-coated cells by adherence to rabbit-anti-mouse Ig-coated plastic dishes. See, for example, Emerson et al, J. Clin. Invest. (1985) 76:1286-1290.
  • the lineage committed cells used in the present invention may be substantially free of stem and progenitor cells.
  • the lineage committed cells are other than human bone marrow stromal cells.
  • the lineage committed cells may be other than mesenchymal cells.
  • the term “proliferative potential” refers to the ability of a cell population to divide and thereby produce more cells of the same or more differentiated type. Accordingly, the proliferative potential of a cell population may be determined by culturing the cells and then determining the degree of expansion of the original cell population. By culturing the lineage committed cells in a medium that is replaced product cells are obtained which have enhanced proliferative potential. This means that the product cells have a greater ability to produce more cells as compared to the cells that were used at the beginning of the culturing. In a particularly preferred embodiment, the product cells of the present invention have a greater ability to replicate or further differentiate to the desired cell type as compared to the same cells which have been cultured at low densities in a static culture.
  • Static culture conditions for the comparative purposes are conditions employing the same medium, same starting cell inoculum source except grown without frequent medium exchange.
  • the T-cell concentration in a conventional low density culture is allowed to reach a maximum of 2-4 ⁇ 10 6 cells/ml. during cell growth with time. After growth to this maximum density, the cell concentration is reduced to 5 ⁇ 10 5 cells/ml and the cells are allowed to grow again to 2-4 ⁇ 10 6 cells/ml. This cycle of growth to maximum cell density followed by a reduction of 5 ⁇ 10 5 cells/ml is repeated throughout the entire culture period.
  • the cell density is not substantially reduced or adjusted at any time during the culture period.
  • T-cells grow to maximum cell densities of 12-40 ⁇ 10 6 cells/ml under conditions of culture medium replacement.
  • the cells having enhanced proliferative potential expand at least 5-fold expansion when cultured in a static culture using a T-flask (see Example 2 below). More preferably, the T-cells of the present invention expand at least 10-fold, even more preferably at least 25-fold.
  • T-cells which expand at least 40, 75, 100, and 1 50-fold in the secondary static culture may be obtained. Even higher proliferative potentials, e.g., at least 200, 500, 1000 or 2000-fold expansion in the secondary culture may be observed using the method of the present invention.
  • biological function refers to the ability of a cell population to carry out its biological mission, i.e., to perform its recognized biological purpose in vivo.
  • biological function of a cell population is determined by the nature of the lineage committed cells that are being cultured. Therefore, the biological function of one cell population may be quite different from another.
  • biological function examples include secretion of substances (such as cytokines, hormones, antibodies, etc.), cell-cell communication, receptor expression on the cell surface, cytolysis, antigen presentation, antigen processing, ability to home in vivo to sites for function, ability to proliferate leading to development/regeneration of tissue similar to naturally occurring structure/function.
  • substances such as cytokines, hormones, antibodies, etc.
  • cell-cell communication such as cytokines, hormones, antibodies, etc.
  • receptor expression on the cell surface such as cytolysis, antigen presentation, antigen processing, ability to home in vivo to sites for function, ability to proliferate leading to development/regeneration of tissue similar to naturally occurring structure/function.
  • the biological function of T-cells are cytolysis and secretion of cytokines such as IFN- ⁇ , IL-10, TNF- ⁇ and GM-CSF.
  • cytokines such as IFN- ⁇ , IL-10, TNF- ⁇ and GM-CSF.
  • the cell products of the present invention have a greater biological function as compared to the cells used at the beginning of the culture.
  • the product cells have a greater biological function as compared to the same cells cultured under static medium culture conditions.
  • the biological function of a cell population may be determined by measuring the amount of a particular response, e.g., cytokine secretion, and quantifying the amount of response on a per cell basis.
  • the cells cultured according to the present invention may have a biological function which is enhanced at least 1.2-, 1.5-, 2-, 3-, 4-, 5-, 10-, 20-, 50-, 75-, 100-, 150-, 200-, 250-, 300-, 350-, 400-, 450- or 500-fold, as compared to cells cultured without medium replacement (i.e., cells cultured under static medium culture conditions).
  • the replicative potential or biological function of a cell population is a property of the population considered as a whole. For example, even in the cells produced by the present invention it is possible that some of the cells in the population may not divide or effect a biological function. However, on average, the cells produced according to the present invention have greater replicative potential, biological function, or both, as compared to cells produced according to low density static culture procedures.
  • cells may have a variety of biological functions. For example, a cell may secrete multiple cytokines. In the present invention, the cells may have one enhanced biological function, e.g., the secretion of one cytokine is enhanced. Alternatively, multiple biological functions may be enhanced.
  • the cells produced according to the present invention have enhanced replicative potential. In another embodiment, the cells have enhanced biological function. In still another, and preferred embodiment, the cells obtained by the present method have both enhanced replicative potential and enhanced biological function. As compared to cells cultured in a static medium, the cells produced according to the present invention preferably have at least a two-fold greater replicative potential and/or biological function. More preferably at least five-fold, and, most preferably at least ten-fold greater replicative potential and/or biological function as compared to cells cultured in static culture. This enhanced replicative potential and/or biological function may be determined after the cells are isolated.
  • the inventors have discovered that after isolating the cells produced under conditions of medium replacement the cells can be recultured (i.e., a secondary culture) in, for example, a static medium and the enhanced replicative potential and/or biological function may be observed. Accordingly, the cells produced according to the present method are expected to demonstrate enhanced replicative potential and/or biological function after infusion into a patient.
  • the lineage committed cells may be cultured in any known physiologically acceptable liquid culture medium, i.e., a medium which supports the cell viability and proliferation, using any conditions as long as media replacement conditions are used.
  • a physiologically acceptable liquid culture medium i.e., a medium which supports the cell viability and proliferation
  • the composition of the media may vary with the cell type being cultured.
  • Media suitable for culturing specific cells are well-known, for example, see Schmidt-Wolf IGH, Negrin R S, Kiem H, Blume K G, Weissman I L: Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity.
  • the culture medium contains organic and inorganic components required for cell proliferation and may contain standard known medium components such as, for example, AIM V, IMDM, MEM, DMEM, RPMI 1640, Alpha Medium or McCoy's Medium, which can use combinations of serum albumin, cholesterol and/or lecithin selenium and inorganic salts.
  • these cultures may be supplemented with corticosteroids, such as hydrocortisone at a concentration of 10 ⁇ 4 to 10 ⁇ 7 M, or other corticosteroids at equal potent dose, such as cortisone, dexamethasome or solumedrol.
  • corticosteroids such as hydrocortisone at a concentration of 10 ⁇ 4 to 10 ⁇ 7 M
  • corticosteroids such as hydrocortisone at a concentration of 10 ⁇ 4 to 10 ⁇ 7 M
  • other corticosteroids at equal potent dose, such as cortisone, dexamethasome or solumedrol.
  • the cultures are
  • the medium used in accordance with the invention may comprise one or more basic components.
  • the first component is a media component comprised of AIM V, IMDM, MEM, DMEM, RPMI 1640, Alpha Medium or McCoy's Medium, or an equivalent known culture medium component.
  • the second is a serum component which comprises at least horse serum or human serum and may optionally further comprise fetal calf serum, newborn calf serum, and/or calf serum.
  • the third component is a corticosteroid, such as hydrocortisone, cortisone, dexamethasome, solumedrol, or a combination of these, preferably hydrocortisone.
  • compositional make up of various media which can be used in the present invention are well-known, see U.S. Pat. No. 5,635,386, columns 11-30; Lewko W M, Good R W, Bowman D, Smith T L, Oldham R K: Growth of tumor derived activated T-cells for the treatment of cancer. Cancer Biotherapy, vol 9, No. 3, pp 221, 1994; Freedman R S, Tomasovic B, Templin S, Atkinson E N, Kudelka A, Edwards C L, Platsoucas C D: Large-scle expansion in interleukin-2 of tumor-infiltrating lymphocytes from patients with ovarian carcinoma.for adoptive immunotherapy: J Immunol Methods 167:145-160, 1994.
  • the serum component may be present in the culture in an amount of at least 1% (v/v) to 50% (v/v).
  • the serum concentration may be preferably in the neighborhood of 15 to 30% (v/v).
  • the exchange rate is increased proportionately.
  • the third component may be present in an amount of from 10 ⁇ 7 M to 10 ⁇ 4 M, and is preferably present in an amount of from 5 ⁇ 10 ⁇ 6 to 5 ⁇ 10 ⁇ 5 M.
  • the media component represents the balance such that all three components add up to 100%.
  • the serum component can be replaced by any of several standard serum replacement mixtures which typically include insulin, albumin, and lecithin or cholesterol. See, Migliaccio, et al, Exp. Hematol. (1990) 18:1049-1055, Iscove et al, Exp. Cell Res. (1980) 126:121-126, and Dainiak et al, J. Clin..Invest. (1985) 76:1237-1242.
  • the cell density in the present method may vary widely.
  • the CD34+ selected inoculum density for derivation of dendritic cells is 3.33-33.3 cells/cm 2 or 10 4 -10 5 cells/ml.
  • growth factors e.g., hematopoietic growth factors
  • synthetic hematopoietic growth factors are selected according to the nature of the lineage committed cells being cultured.
  • the particular growth factors which stimulate a given cell type in cell culture are well-known, see for example, Romani N. Gruner S, Brang D, Kampgen E, Lenz A,rbacher B, Konwalinka G, Fritsch P O, Steinman R M, Schuler G: Proliferating dendritic cell progenitors in human blood. J Exp Med 180:83,1994; Maraskovsky E, Brasel K.
  • IL-2 is preferably used in the culturing of T-cells.
  • the amount of IL-2 in the culture medium may vary widely.
  • the IL-2 concentration is 25 to 1,000 IU/ml.
  • GM-CSF, IL-4, TNF- ⁇ , Flt3-L, or combinations thereof, are preferably added to the culturing medium when expanding dendritic cells.
  • GM-CSF 50 ng/ml (1 to 500 ng/ml); IL-4: 25 ng/ml (1 to 500 ng/ml); TNF ⁇ : 25 ng/ml (1 to 500 ng/ml); Flt3L: 25 ng/ml (1 to 500 ng/ml).
  • growth factors which may be added to the culture medium include the cytokines IL-3 and GM-CSF alone or together at a rate of from 0.1 to 100 ng/ml/day, preferably about 0.5 to 10 ng/ml/day, most preferably 1 to 2 ng/ml/day.
  • Epo may be added to the nutrient medium in an amount of from 0.001 to 10 U/miday, preferably 0.05 to 0.15 U/ml/day.
  • Mast cell growth factor (MCGF, c-kit ligand, Steel factor)
  • IL-1 ( ⁇ or ⁇ ) may also be added in an amount of from 10 to 100 units/ml per 3 to 5 day period. Additionally, IL-6, G-CSF, basic fibroblast growth factor, IL-7, IL-8, IL-9, IL-10, IL-11, PDGF, or EGF to be added, at a rate of from 1 to 100 ng/ml/day.
  • the present invention relies on first culturing the lineage committed cells in a liquid culture medium which is replaced, preferably perfused, either continuously or periodically.
  • the rate of medium replacement may be at least 25% daily replacement, preferably at least 50% daily replacement, and may be 25% to 100% daily replacement for a cell density of from 1 ⁇ 10 4 to 1 ⁇ 10 7 cells per ml of culture.
  • the medium exchange rate may be increased proportionally to achieve a constant medium and serum flux per cell per unit time.
  • the medium exchange rate may likewise be decreased proportionately.
  • the medium replacement rate may vary during the culturing. In one embodiment, the replacement rate is relatively low at the beginning of the culturing and then increased as the cell density in the culture increases.
  • Replacement of the nutrient medium in accordance with the invention may be carried out in any manner which will achieve the result of replacing the medium, e.g., as described in U.S. Pat. No. 5,646,043.
  • the flow of the aliquot being added may be by gravity, by pump, or by any other suitable means.
  • the flow may be in any direction or multiplicity of directions, depending upon the configuration and packing of the culture.
  • the new medium is added to the culture in a manner such that it contacts the cell mass. Most preferably, it is added the culture in a manner mimicking in vivo perfusion, i.e., it is perfused through at least part of the cell mass and up to the whole cell mass.
  • the metabolic product level in the medium is normally maintained within a particular range.
  • Glucose concentration is usually maintained in the range of about 5 to 20 mM.
  • Lactate concentration is usually maintained below 35 mM, preferably below 0.5 mg/ml.
  • Glutamine concentration is generally maintained in the range of from about 1 to 3 mM.
  • Ammonium concentration is usually maintained below about 2.4 mM.
  • the culture medium is continuously perfused to provide fresh medium at a rate which is proportional to the lactate concentration and/or the cell density in the culture.
  • this medium replacement is accomplished without diluting the cell density of the culture.
  • the culture time may vary widely.
  • the cells are preferably cultured for at least the minimum amount of time required to produce cells with enhanced replicative potential, biological function, or both. This time may vary with cell type, depending on the cell doubling time.
  • the cells are cultured according to the invention for at least 2 days, more preferably, at least 4 days.
  • the maximum culture time is not particularly limited.
  • the cells may be cultured for up to 10 days, up to 25 days, up to 50 days, up to 75 days, up to 100 days, or longer, if desired (>100 days [e.g., T-cells, Rosenberg]).
  • An obstacle with ex vivo expansion has been the ability to reliably generate large quantities of ex vivo expanded lineage committed cells in a controlled process for effective therapy.
  • the traditional cell expansion methods in bags, flasks or roller bottles involves multi-step manual cell culture processes. Due to the lack of continuous medium perfusion features, the cell concentrations are maintained at a low level ( ⁇ 1 ⁇ 10 6 cells/ml) by the addition of fresh medium to the culture container or by transferring part of the culture to additional culture containers. Such procedures require numerous aseptic transfers resulting in less reproducible and reliable performance and increased risk of contamination or critical operator error.
  • an automated clinical-scale cell production system which has been developed to process and expand cells in a closed and sterile environment and which is described in allowed U.S. application Ser. No. 08/478,622 (attorney reference: 4292-022-55), incorporated herein by reference, is used. This is capable of exchanging medium at a continuous rate without removal of cells.
  • the use of the CPS requires a single aseptic transfer and is microprocessor controlled, resulting in significant savings in labor and space.
  • the CPS has been automated by incorporating microprocessor controlled hardware and the development of software to run the processes.
  • The. CPS embodies a modular, closed-system process comprised of a pre-sterilized, single-use disposable cell cassette operated by automated instruments.
  • the instrumentation components of the system include an incubator unit and processor unit, along with a computer based system manager.
  • the cell cassette provides a closed, sterile environment in which cell production can occur.
  • the single-use cartridge is provided fully assembled in a sterile package, opened just prior to use.
  • the sterile pathway contained in the cell cassette includes: cell growth chamber, medium supply container, a pump for delivery of components, and sterile barrier elements throughout.
  • a dedicated, small incubator controls the biological and physical environment and operations of each cell cassette necessary to support the cell growth process.
  • the incubator receives and self-engages the disposable cell cassette, much like a VCR and videocassette.
  • the incubator controls: the flow of medium to the growth chamber, the temperature (4° C.) of the growth medium supply compartment, the temperature (37° C.) of the growth chamber compartment, and the concentration and flow rate of gases delivered to the gas compartment of the culture chamber.
  • the incubator also monitors various safety-alarm parameters to assure that the cell production process is proceeding as expected. In the unlikely event of failure of an incubator, the cell cassette, is readily transferred to another incubator with minimal interruption of the culture process.
  • the processor performs the initial priming of the cell cassette with growth medium and, through instructions to the operator, the controlled inoculation of cells.
  • the same unit also performs the removal (harvest) of the cells from the growth chamber at the completion of the cell production process, sterilely transferring the cells to a pre-attached harvest container (analogous to a blood transfusion bag).
  • the system manager employs a user-friendly graphical interface.
  • the system manager provides a convenient central user interface and provides for redundant monitoring of each incubator in the network.
  • the system manager can perform procedure scheduling (up to 50 incubators) tasks for the operators and provide a daily or weekly printed record of alarm events for quality control and record keeping purposes.
  • the ID key contains a semiconductor memory device and clock and is affixed to each cell cassette at the beginning of a cell production procedure.
  • the ID key provides reliable identification of the cell product, instructs the instruments for the cell production process, prevents mix-ups and operator error, and stores the primary data for complete process history record (effectively a “manufacturing batch record” for the cell product).
  • the lineage committed human cells After culturing according to the invention, the lineage committed human cells have enhanced replicative and/or biological function.
  • the cultured cells may be isolated by harvesting them from the culture apparatus.
  • the cells may be harvested by, for example, withdrawing the cells by syringe, or by continuously allowing the cells to flow out of the culture reactor, by the pressure produced by replacing the culture medium, through an exit tube. After harvesting, the cells may be infused in a patient to obtain the therapeutic benefits of the cultured cells.
  • lineage committed cells cultured under conditions of medium exchange have an enhanced ability to replicatate and/or have enhanced biological function after the cells are isolated. Since the cells of the present invention have replicative and/or biological function, fewer cells may be required to achieve a given level of effectiveness as compared to cells that were cultured under static conditions.
  • CD8+ cells were inoculated into the CPS at a density of 20-80 million cells (0.08 ⁇ 10 6 -0.32 ⁇ 10 6 cells/ml) in AIM-V medium containing 600 IU/ml of rIL-2.
  • a yield of 3-8 billion cells (12 ⁇ 10 6 -32 ⁇ 10 6 cells/ml) (>90% viability) was obtained at harvest on day 10 (n 12). Continuous one pass perfusion of fresh medium enabled the CPS to maintain high cell densities and low lactate concentrations ( ⁇ 0.5 mg/ml).
  • TCR T-cell receptor
  • the growth of PHA-activated human CD8 + T-cells was used as a model system for evaluating the effects of continuous perfusion on T-cell expansion.
  • Human CD8 + T-cells were expanded using continuous one-pass perfusion with medium containing IL-2 in the CPS for ten days. The perfusion rates were increased with the increase in cell number such that lactate concentration in the spent medium was maintained at 0.5 mg/ml.
  • the continuous perfusion of medium resulted in high density cultures of concentrations ranging from 12 to 32 million cells/ml starting from a population of 0.16-0.32 million cells/ml.
  • Parallel T-flask cultures were set up at the same culture conditions (vis. inoculum density, culture depth, oxygenation, and medium composition).
  • T-cell harvested from the CPS or parallel T-flask cultures was assessed by analysis of cytokine release (see FIG. 1 ).
  • T-cells harvested from the CPS or T-flasks produced similar concentrations of IFN- ⁇ in response to anti-CD3 mAb.
  • T-cells derived from the CPS produced higher concentrations of TNF- ⁇ and GM-CSF than T-cells derived from T-flasks.
  • the data indicates that T-cells derived in the CPS are fully responsive to stimulation through the TCR-CD3 complex and may produce higher levels of particular cytokines on a per cell basis than T-cells produced in T-flasks.
  • T-cells derived in the CPS using continuous perfusion have important applications for the therapeutic efficacy of these T-cells after reinfusion.
  • Effective adoptive immunotherapy for viral diseases and cancer requires the reliable generation of large quantities of functional cells capable of continued cell expansion in vivo together with recirculation and localization of effector T-cells at sites of various infection or tumor cell growth.
  • Expansion of T-cells using continuous perfusion in the CPS results in large numbers of high quality effector T-cells with enhanced replication potential and effector function.
  • PB MNCs Peripheral blood mononuclear cells
  • the cultures either were not fed, or received 50% fresh serum-free AIM V medium containing GM-CSF and IL-4 on days 2, 4 and 6. TNF ⁇ was added on day 6 for the final 24 hours of culture.
  • Cells from these cultures expressed the characteristic “veiled” morphology and surface phenotype of dendritic cells as determined by the cytometry.
  • DCs demonstrated at least 50-fold greater stimulating activity in the alloMLR compared to PB MNCs.
  • frequent medium exchange particularly at high inoculum density, significantly enhanced the stimulating activity of harvested DCs (>5-fold compared to static culture conditions.
  • Total cell recovery correlated well with lactate produced per culture supporting the utility of lactate measurement as a non-invasive means to improve medium exchange protocols and monitor DC culture productivity.
  • FIGS. 2-4 Results obtained with such small-scale cultures are shown in FIGS. 2-4 .
  • the cultures represented in FIGS. 2, 3 , and 4 were conducted at low, intermediate, and high inoculum density, respectively.
  • Panel (A) of each Figure shows the lactate concentration as a function of time during the seven day culture period.
  • Panel (B) of each Figure shows the mixed leucocyte response (MLR) observed with the harvested DCs. In these assays, the harvested DCs, which were derived from one individual, were contacted with the T-cells of another individual.
  • MLR mixed leucocyte response
  • the MLR response was determined by measuring the uptake of 3 -H-thymidine in the stimulated T-cells (a measure of DNA synthesis in these T-cells) and is shown as “CPM” plotted against the number of harvested DCs in contact with the T-cells.
  • the data in FIG. 2 shows the results obtained with 100% medium exchange and 0% medium exchange. In the medium-exchanged culture, 50% medium replacement was performed on day 4 and 7 (indicated by arrows in the Figure). The data in Figure. 3 shows the results observed with 0% medium exchange and 150% medium exchange. In the medium-exchanged culture, 50% medium replacement was performed on day 3, 5, and 7 (indicated by arrows).
  • FIG. 4 shows the results of culturing using 100% and 300% medium exchange.

Abstract

A method for obtaining lineage committed human cells imbued with enhanced proliferative potential, biological function, or both, comprising culturing lineage committed human cells under physiologically acceptable liquid culture conditions, where the liquid culture medium is replaced at a rate and for a time sufficient to obtain the human lineage committed cells imbued with enhanced proliferative potential, biological function, or both; and isolating the cultured cells.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention relates to methods for culturing lineage committed human cells, the cells-thus obtained, and their uses.
  • 2. Description of the Background
  • There is significant interest using both early and lineage committed cells for a variety of therapeutic purposes.
  • In tissue engineering, the goal is to reconstitute fully or partially functioning human tissue in vitro to enable a variety of clinical and other applications. Several studies have been carried out that are aimed at reconstituting functioning human tissues in vitro. Illustratively, the cultivation of human skin has been successful.
  • The hematopoietic system exemplifies the broad range of cells involved in protection of mammalian hosts against pathogens toxins, neoplastic cells, and other diseases. The hematopoietic system is believed to evolve from a single stem cell, from which all the lineages of the hematopoietic system derive. Hematopoietic cells have been used in human therapy. Methods and apperati for culturing precursor hematopoietic cells to obtain desired mature hematopoietic cells have been described. See, U.S. Pat. Nos. 5,605,822, 5,399,493; 5,437,994; 5,459,069; 5,635,386, 5,670,147 and 5,670,351.
  • Adoptive cell therapy is the ex vivo expansion and re-infusion of immune effector cells into human recipients for the treatment or prevention of disease (Rosenberg et al., Science 233, 1318 (1986)). Developments in T-lymphocyte-based adoptive immunotherapy have lead to significant advances in the treatment of metastatic cancer (Rosenberg et al., Science 233, 1318 (1986); Rosenberg et al., N. Eng. J. Med. 319, 1676 (1988); Rosenberg et al., N. Eng. J. Med. 323, (1990)) and viral diseases including Epstein-Barr Virus (EBV) (Heslop et al. 1996), cytomegalovirus (CMV) (Riddell et al., Science 257, 238 (1992)) and human immunodeficiency virus (HIV) (Whiteside et al., Blood 81, 2085 (1993); Ridell et al., Hum. Gen. Ther. 3, 319 (1992)) in humans. Multiple patient populations have been identified in which T-cell therapy has achieved a response rate of approximately 34% after administration of tumor infiltrating lymphocytes (TILs) to metastatic melanoma patients (Rosenberg et al., J. Natl. Cancer Inst. 86, 1159 (1994)). A target human therapeutic dose of 1010-1011 TILs is suggested based on extrapolation from studies in mouse tumor models and clinical experience (Topalian et al., J. Immunol. Meth. 102, 107 (1987)). Clinical responses to melanoma tumors have been obtained after expansion ex vivo of TILs in medium containing interleukin-2 (IL-2) and re-infusion of greater than 1011 T-cells per treatment cycle together with high doses of exogenous IL-2 (Rosenberg et al., N. Eng. J. Med. 319, 1676 (1988); Rosenberg et al. 1994). The enhanced anti-tumor activity of TILs in vivo compared to lymphokine activated killer (LAK) cells or NK cells may be a function of several complex processes including the ability of these T-cells to proliferate and release an array of lymphokines, to recirculate and accumulate at sites of tumor growth, and to specifically recognize and lyse autologous tumor cells (Pockaj et al. 1994; Rosenberg et al. 1994).
  • Dendritic cells (DCs) are highly specialized bone-marrow derived antigen presenting cells (APCs) which function as potent stimulating cells for primary T-lymphocyte-mediated immune response. Dendritic cell based immunotherapy is an emerging treatment strategy involving ex vivo expansion and reinfusion of antigen-pulsed or genetically modified DCs into human patients to vaccinate against cancer or infectious diseases. Clinical implementation of these therapies requires the capability of product sufficient quantities of functional DCs for effective patient treatment.
  • The use of cultured human cells in human therapy has required that a quantity of active cells sufficient to provide a therapeutic effect upon infusion into the patient be used. This has required using culture systems providing large numbers of cells. There is therefore a need for methods for obtaining lineage committed cells with augmented proliferative potential, biological function, or both since such methods would provide more potent cell compositions, capable of being used in smaller amounts in therapy.
  • SUMMARY OF THE INVENTION
  • Accordingly, it is an object of this invention to provide novel methods, including culture media conditions, for obtaining human lineage committed cells with enhanced proliferative potential, biological function, or both.
  • It is another object of this invention to provide cultured human lineage committed cells having enhanced proliferative potential, biological function, or both.
  • These objects and others may be accomplished by a method for obtaining lineage committed human cells having enhanced proliferative potential, biological function, or both. In this method, lineage committed human cells are cultured in any physiologically acceptable liquid culture medium conditions with the liquid culture medium being replaced.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the amount of cytokine produced after stimulation of T-cells cultured using continuous medium exchange conditions according to the present invention and cells cultured under standard static low density medium conditions. AIM V represents a control with no added stimulus, all experiments were cultured with AIM V culture medium. OKT3 represents anti-CD3 mAb. (A) IFN-γ production; (B) TNF-α production; (C) GM-CSF production; (D) IL-10 production.
  • FIG. 2 shows (A) the lactate concentration during a dendritic cell culture according to the present invention (100% exchange) and in a static culture (0% medium exchange) at low inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • FIG. 3 shows (A) the lactate concentration during a dendritic cell culture according to the present invention (150% exchange) and in a static culture (0% medium exchange) at intermediate inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • FIG. 4 shows (A) the lactate concentration during a dendritic cell culture according to the present invention at 100% exchange and 300% medium exchange at high inoculum density, and (B) the mixed leucocyte response (MLR) of the isolated dendritic cells.
  • FIG. 5 shows the effect of medium exchange rate of canine chondrocyte production at different inoculum densitites.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The present invention applies to the culture of lineage committed human cells using any known culture techniques. The invention is based on the discovery that by replacing the liquid culture media in any lineage committed human cell culture without substantially changing the cell density, it is possible to obtain cells having an enhanced proliferative potential, an enhanced biological function, or both. The cells obtained in accordance with the invention have enhanced proliferative potential as compared to the cells prior to culture and/or as compared to cells one obtains using otherwise identical culture conditions except that the liquid culture media is replaced by diluting the culture to achieve a lower cell density. Similarly, the biological function of the cells obtained in accordance with the invention is enhanced as compared with the biological function of these same cells prior to culture and/or as compared to the biological function of the same type of cells cultured under otherwise identical culture conditions except that the liquid medium is not replaced.
  • The cells one obtains in accordance with the invention are, by virtue of their enhanced proliferative potential and/or biological effector function, more potent. The present invention permits using a smaller number of lineage committed human cells to obtain similar or better results than one obtains using lineage committed human cells cultured in accordance with techniques existing prior to the present invention. The lineage committed human cells one obtains in accordance with the invention can be used in any application in which lineage committed human cells are used, including, but not limited to, therapy treatments including adoptive immunotherapy with effector cells, tumor specific cytotoxic T-cells, infectious disease specific cytotoxic T lymphocytes, cytokine induced killer cell therapy, antigen presenting cells to either tumor or infectious diseases, dendritic cells, antigen primed, dendritic cells, tumor vaccines, genetically modified attenuated tumor cells, genetically modified antigen presenting cells, structural repair procedures such as cartilage defect repair, bone defect repair, tissue repair, would healing, burn care, solid organ repair, neurological defect repair, etc. Derivation and expansion of antigen specific T-cell populations including, but not limited to, viral (e.g. EBV, HPV, CMV, HIV, influenza) and tumor reactive T-cells, ex vivo expansion of human tumor infiltrating lymphocytes (TILs) for adoptice cancer immunotherapy, derivation and ex vivo expansion of cytokine induced killer (CIK) cells for rejection of human tumors including leukemia, lymphoma, breast and other cancers. Also, immunotherapy after autologous or allogeneic bone marrow transplantation. With dendritic cells: antigen presenting cell (APC)-based vaccines to stimulate T-cell responses in vitro or in vivo against simple or complex antigens including, but not limited to, tumor, viral, fungal and bacterial antigens. Therapy may include treatment or prevention of disease in normal individuals or human cancer patients.
  • Lineage Committed Cells
  • The lineage committed human cell used in accordance with the present invention are cells which are differentiated to at least a point where they are programmed to develop into a specific type of cell. These cells are not necessarily terminally differentiated. For example, CFU-GEMM cells are committed to develop, ultimately, into mature myeloid cells. Similarly, CFU-L cells are committed to develop into lymphoid cells. Accordingly, the lineage committed cells may be multipotential stem cells or committed progenitor cells. Of course, the lineage committed cells may be mature cells derived from these precursors. As used herein, the term “mature cells” refers to-cells which are terminally differentiated; In one embodiment, the lineage committed cells are more differentiated than human stem cells. In another embodiment, the lineage committed cells are more differentiated than progenitor cells. In yet another embodiment, the lineage committed cells are mature cells. A discussion of cell development for blood cells and lymphocytes is provided by S. McKenzie, Hematology, Second Edition, William and Wilkins, 1996, pp. 9-30 and 55-89.
  • The types of lineage committed cells used in the present method may vary widely. Any type of lineage committed cell which may benefit from medium replacement may be used in the present invention. Preferred cells which can be used include human hematopoietic cells, mesenchymal cells, dendritic cells, fibroblasts, hepatocytes, neural cells, epithelial cells, lymphocytes, keratinocytes, osteoblasts or osteoclasts. Specific examples of suitable human hematopoietic cells include megakaryocytes, monocytes, neutrophils, basophils, eosinophils, tumor specific cytotoxic T lymphocytes, cytokine induced killer cells, antigen presenting cells to either tumor or infectious diseases, dendritic cells, antigen primed dendritic cells, leukocyte precursor, and neutrophils. Suitable examples of mesenchymal cells include, chondrocytes, osteoblasts, myeoblasts, fibroblasts, tenoblasts, stromal cells (e.g., from bone marrow), tenocytes, adipocytes, osteocytes and myocytes. Suitable examples of lymphocytes include T-cells and B-cells. Pre-T and pre-B cells are also suitable. The T-cells (CD3+) may be CD8+ or CD4+ cells, or cells derived from said populations.
  • In one embodiment of the present invention, the lineage committed cells may be stem cells (e.g., hematopoietic or stromal stem cells), progenitor cells (e.g., hematopoietic progenitors), mature myeloid cells, or stromal cells (e.g., from bone marrow). In another embodiment, the cells may be dendritic cells (e.g., myeloid- or lymphoid-derived) or non-myeloid mature cells which are other than stromal cells (e.g., the mature cells described above, especially T-cells or chondrocytes).
  • The cells used in the present method may be obtained from a variety of sources using well-known techniques, see Heslop H E, Ng Cyc, Li C, Smith C A, Loftin S K, Krance Ra, Brenner M K, Rooney C M: Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nature Med 2:551, 1996; Lu P, Negrin RS: A novel population of expanded human CD3+CD56+ cells derived from T-cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency. J Immunol 153:1687, 1994; Romani N, Gruner S, Brang D, Kampgen E, Lenz A, Trockenbacher B, Konwalinka G, Fritsch P O, Steinman R M, Schuler G: Proliferating dendritic cell progenitors in human blood. J Exp Med 180:83, 1994; Rosenberg S A, Spiess P, Lafreniere R: A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233:1318, 1986.
  • When culturing T-cells, for example, a human hematopoietic cell composition enriched in T-cells may be used. Such a composition may be enriched in T-cell content by any desired amount, such as by up to 103 fold or more. Different, known methods may be used to achieve this enrichment, corresponding either to a negative selection method or a positive selection method. For example, in accordance with the negative selection method, mature cells are isolated using immunological techniques, e.g.,. labeling non-progenitor, non-stem cells with a panel of mouse anti-human monoclonal antibodies, then removing the mouse antibody-coated cells by adherence to rabbit-anti-mouse Ig-coated plastic dishes. See, for example, Emerson et al, J. Clin. Invest. (1985) 76:1286-1290.
  • In one embodiment of the present invention, the lineage committed cells used in the present invention may be substantially free of stem and progenitor cells. As discussed above, the lineage committed cells are other than human bone marrow stromal cells. In another embodiment, the lineage committed cells may be other than mesenchymal cells.
  • Enhanced Proliferative and Biological Function
  • As used herein, the term “proliferative potential” refers to the ability of a cell population to divide and thereby produce more cells of the same or more differentiated type. Accordingly, the proliferative potential of a cell population may be determined by culturing the cells and then determining the degree of expansion of the original cell population. By culturing the lineage committed cells in a medium that is replaced product cells are obtained which have enhanced proliferative potential. This means that the product cells have a greater ability to produce more cells as compared to the cells that were used at the beginning of the culturing. In a particularly preferred embodiment, the product cells of the present invention have a greater ability to replicate or further differentiate to the desired cell type as compared to the same cells which have been cultured at low densities in a static culture.
  • Static culture conditions for the comparative purposes are conditions employing the same medium, same starting cell inoculum source except grown without frequent medium exchange. For example, the T-cell concentration in a conventional low density culture is allowed to reach a maximum of 2-4×106 cells/ml. during cell growth with time. After growth to this maximum density, the cell concentration is reduced to 5×105 cells/ml and the cells are allowed to grow again to 2-4×106 cells/ml. This cycle of growth to maximum cell density followed by a reduction of 5×105 cells/ml is repeated throughout the entire culture period.
  • In contrast, in the method of the present invention for culturing T-cells, the cell density is not substantially reduced or adjusted at any time during the culture period. Thus, T-cells grow to maximum cell densities of 12-40×106 cells/ml under conditions of culture medium replacement.
  • For the cultured cells of the present invention (e.g., T-cells) the cells having enhanced proliferative potential expand at least 5-fold expansion when cultured in a static culture using a T-flask (see Example 2 below). More preferably, the T-cells of the present invention expand at least 10-fold, even more preferably at least 25-fold. Using the method of the present invention T-cells which expand at least 40, 75, 100, and 1 50-fold in the secondary static culture may be obtained. Even higher proliferative potentials, e.g., at least 200, 500, 1000 or 2000-fold expansion in the secondary culture may be observed using the method of the present invention.
  • The term “biological function” refers to the ability of a cell population to carry out its biological mission, i.e., to perform its recognized biological purpose in vivo. As one skilled in the art will readily appreciate, the biological function of a cell population is determined by the nature of the lineage committed cells that are being cultured. Therefore, the biological function of one cell population may be quite different from another.
  • Examples of biological function include secretion of substances (such as cytokines, hormones, antibodies, etc.), cell-cell communication, receptor expression on the cell surface, cytolysis, antigen presentation, antigen processing, ability to home in vivo to sites for function, ability to proliferate leading to development/regeneration of tissue similar to naturally occurring structure/function.
  • For example, the biological function of T-cells are cytolysis and secretion of cytokines such as IFN-γ, IL-10, TNF-α and GM-CSF. By replacing the medium during culturing, cells having enhanced biological function may be obtained. Thus, the cell products of the present invention have a greater biological function as compared to the cells used at the beginning of the culture. In a particularly preferred embodiment, the product cells have a greater biological function as compared to the same cells cultured under static medium culture conditions. The biological function of a cell population may be determined by measuring the amount of a particular response, e.g., cytokine secretion, and quantifying the amount of response on a per cell basis. The cells cultured according to the present invention may have a biological function which is enhanced at least 1.2-, 1.5-, 2-, 3-, 4-, 5-, 10-, 20-, 50-, 75-, 100-, 150-, 200-, 250-, 300-, 350-, 400-, 450- or 500-fold, as compared to cells cultured without medium replacement (i.e., cells cultured under static medium culture conditions).
  • It is important to note that the replicative potential or biological function of a cell population is a property of the population considered as a whole. For example, even in the cells produced by the present invention it is possible that some of the cells in the population may not divide or effect a biological function. However, on average, the cells produced according to the present invention have greater replicative potential, biological function, or both, as compared to cells produced according to low density static culture procedures. In addition, cells may have a variety of biological functions. For example, a cell may secrete multiple cytokines. In the present invention, the cells may have one enhanced biological function, e.g., the secretion of one cytokine is enhanced. Alternatively, multiple biological functions may be enhanced.
  • In one embodiment, the cells produced according to the present invention have enhanced replicative potential. In another embodiment, the cells have enhanced biological function. In still another, and preferred embodiment, the cells obtained by the present method have both enhanced replicative potential and enhanced biological function. As compared to cells cultured in a static medium, the cells produced according to the present invention preferably have at least a two-fold greater replicative potential and/or biological function. More preferably at least five-fold, and, most preferably at least ten-fold greater replicative potential and/or biological function as compared to cells cultured in static culture. This enhanced replicative potential and/or biological function may be determined after the cells are isolated. The inventors have discovered that after isolating the cells produced under conditions of medium replacement the cells can be recultured (i.e., a secondary culture) in, for example, a static medium and the enhanced replicative potential and/or biological function may be observed. Accordingly, the cells produced according to the present method are expected to demonstrate enhanced replicative potential and/or biological function after infusion into a patient.
  • Culture Medium and Environment
  • The lineage committed cells may be cultured in any known physiologically acceptable liquid culture medium, i.e., a medium which supports the cell viability and proliferation, using any conditions as long as media replacement conditions are used. Of course, the composition of the media may vary with the cell type being cultured. Media suitable for culturing specific cells are well-known, for example, see Schmidt-Wolf IGH, Negrin R S, Kiem H, Blume K G, Weissman I L: Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med 174:139, 1991; Morse M A, Zhou L J, Tedder T F, Lyerly H K, Smith C: Generation of dendritic cells in vitro form peripheral blood mononuclear cells with granulocyte-macrophage-colony-stimulating factor, interleukin-4, and tumor necrosis factor-alpha for use in cancer immunotherapy. Ann Surg. July 1; 226:16, 1997; Romani N, Grunner S, Brang D, Kampgen E, Lenz A. Trockenbacher B, Konwalinka G, Fritsch P O, Steinman R M, Schuler G: Proliferating dendritic cell progenitors in human blood. J Exp Med 180:83, 1994.
  • The culture medium contains organic and inorganic components required for cell proliferation and may contain standard known medium components such as, for example, AIM V, IMDM, MEM, DMEM, RPMI 1640, Alpha Medium or McCoy's Medium, which can use combinations of serum albumin, cholesterol and/or lecithin selenium and inorganic salts. As known, these cultures may be supplemented with corticosteroids, such as hydrocortisone at a concentration of 10−4 to 10−7 M, or other corticosteroids at equal potent dose, such as cortisone, dexamethasome or solumedrol. The cultures are typically carried out at a pH which is roughly physiologic, i.e., 6.9 to 7.4. The medium is typically exposed to an oxygen-containing atmosphere which contains from 4 to 20 vol. percent oxygen, preferably 6 to 8 vol. percent oxygen.
  • Illustratively, the medium used in accordance with the invention may comprise one or more basic components. The first component is a media component comprised of AIM V, IMDM, MEM, DMEM, RPMI 1640, Alpha Medium or McCoy's Medium, or an equivalent known culture medium component. The second is a serum component which comprises at least horse serum or human serum and may optionally further comprise fetal calf serum, newborn calf serum, and/or calf serum. The third component is a corticosteroid, such as hydrocortisone, cortisone, dexamethasome, solumedrol, or a combination of these, preferably hydrocortisone.
  • The compositional make up of various media which can be used in the present invention are well-known, see U.S. Pat. No. 5,635,386, columns 11-30; Lewko W M, Good R W, Bowman D, Smith T L, Oldham R K: Growth of tumor derived activated T-cells for the treatment of cancer. Cancer Biotherapy, vol 9, No. 3, pp 221, 1994; Freedman R S, Tomasovic B, Templin S, Atkinson E N, Kudelka A, Edwards C L, Platsoucas C D: Large-scle expansion in interleukin-2 of tumor-infiltrating lymphocytes from patients with ovarian carcinoma.for adoptive immunotherapy: J Immunol Methods 167:145-160, 1994.
  • The serum component may be present in the culture in an amount of at least 1% (v/v) to 50% (v/v). The serum concentration may be preferably in the neighborhood of 15 to 30% (v/v). For higher serum concentrations, the exchange rate is increased proportionately. The third component may be present in an amount of from 10−7 M to 10−4 M, and is preferably present in an amount of from 5×10−6 to 5×10−5 M. The media component represents the balance such that all three components add up to 100%. Alternatively the serum component can be replaced by any of several standard serum replacement mixtures which typically include insulin, albumin, and lecithin or cholesterol. See, Migliaccio, et al, Exp. Hematol. (1990) 18:1049-1055, Iscove et al, Exp. Cell Res. (1980) 126:121-126, and Dainiak et al, J. Clin..Invest. (1985) 76:1237-1242.
  • Cell Density
  • The cell density in the present method may vary widely. Preferably, the cell density is 104 to 108 cells per ml of culture. More preferably, the cell density is T-cells inoculum: 5×104/ml to 2×106/ml; T-cells final density: 5×106 to 5×107/ml; dendritic cells: 1×106 cells/cm2 (3.33×10-3.33×107 cells/ml) (inoculum density); T-cells inoculum: starting population of 40-80 million cells (0.16-0.32×106 cells/ml; harvest: 12-32×106 cells/ml, 10-108 cells/ml; CIK: inoculum density=1×106 cells/ml; density at harvest: 12-42×106 cells/ml. The CD34+ selected inoculum density for derivation of dendritic cells is 3.33-33.3 cells/cm2 or 104-105 cells/ml.
  • Growth Factors
  • Another, optional but important, embodiment of the present invention, resides in the addition of growth factors (e.g., hematopoietic growth factors), including synthetic hematopoietic growth factors, to the medium-exchanged cultures. Of course, the growth factors are selected according to the nature of the lineage committed cells being cultured. The particular growth factors which stimulate a given cell type in cell culture are well-known, see for example, Romani N. Gruner S, Brang D, Kampgen E, Lenz A, Trockenbacher B, Konwalinka G, Fritsch P O, Steinman R M, Schuler G: Proliferating dendritic cell progenitors in human blood. J Exp Med 180:83,1994; Maraskovsky E, Brasel K. Teepe M, Roux E R, Lyman S D, Shortman K, McKenna H J: Dramatic increase in the number of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med 184:1953, 1996; Sallusto F, Lanzavecchia A: Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med 179:1109, 1994; Santiago-Schwarz F, Divaris N, Kay C, Carsons S E: Mechanisms of tumor necrosis factor-granulocyte-macrophage colony-stimulating factor-induced dendritic cell development. Blood 82:3019, 1993; Siena S, Di Nicola M, Bregni M. Mortarini R, Anichini A, Lombardi L, Ravagnani F, Parmiani G, Gianni Am: Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy. Exp Hematol 23:1463, 1995; Rosenberg S A, Spiess P, Lafreniere R: A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233:1318,-1986; Rosenberg S A, Packard B S, Aebersold P M, Solomon D, Topalian S L, Toy S T, Simon P, Lotze M T, Yang J C, Seipp C A, Simpson C, Carter C, Bock S, Schwartzentruber D, Wei J P, White D E: Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. New Engl J Med 319:1676, 1988; Schmidt-Wolf IGH, Negrin R S, Kiem H, Blume K G, Weissman I L: Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med 174:139, 1991.
  • It is widely known that IL-2 is preferably used in the culturing of T-cells. The amount of IL-2 in the culture medium may vary widely. Preferably, the IL-2 concentration is 25 to 1,000 IU/ml. GM-CSF, IL-4, TNF-α, Flt3-L, or combinations thereof, are preferably added to the culturing medium when expanding dendritic cells. General concentration ranges for cytokines in dendritic cell culture media are: GM-CSF: 50 ng/ml (1 to 500 ng/ml); IL-4: 25 ng/ml (1 to 500 ng/ml); TNFα: 25 ng/ml (1 to 500 ng/ml); Flt3L: 25 ng/ml (1 to 500 ng/ml). These ranges include all specific values and subranges.
  • Other growth factors which may be added to the culture medium include the cytokines IL-3 and GM-CSF alone or together at a rate of from 0.1 to 100 ng/ml/day, preferably about 0.5 to 10 ng/ml/day, most preferably 1 to 2 ng/ml/day. Epo may be added to the nutrient medium in an amount of from 0.001 to 10 U/miday, preferably 0.05 to 0.15 U/ml/day. Mast cell growth factor (MCGF, c-kit ligand, Steel factor), may be added to the medium in an amount of from 1 to 100 ng/ml/day, preferably 10 to 50 ng/ml/day. IL-1 (α or β) may also be added in an amount of from 10 to 100 units/ml per 3 to 5 day period. Additionally, IL-6, G-CSF, basic fibroblast growth factor, IL-7, IL-8, IL-9, IL-10, IL-11, PDGF, or EGF to be added, at a rate of from 1 to 100 ng/ml/day.
  • Medium Replacement
  • The present invention relies on first culturing the lineage committed cells in a liquid culture medium which is replaced, preferably perfused, either continuously or periodically. The rate of medium replacement may be at least 25% daily replacement, preferably at least 50% daily replacement, and may be 25% to 100% daily replacement for a cell density of from 1×104 to 1×107 cells per ml of culture. For cell densities higher than 107 cells per ml, the medium exchange rate may be increased proportionally to achieve a constant medium and serum flux per cell per unit time. For cell densities lower than 104 cells per ml, the medium exchange rate may likewise be decreased proportionately. The medium replacement rate may vary during the culturing. In one embodiment, the replacement rate is relatively low at the beginning of the culturing and then increased as the cell density in the culture increases.
  • Replacement of the nutrient medium in accordance with the invention may be carried out in any manner which will achieve the result of replacing the medium, e.g., as described in U.S. Pat. No. 5,646,043. The flow of the aliquot being added may be by gravity, by pump, or by any other suitable means. The flow may be in any direction or multiplicity of directions, depending upon the configuration and packing of the culture. Preferably, the new medium is added to the culture in a manner such that it contacts the cell mass. Most preferably, it is added the culture in a manner mimicking in vivo perfusion, i.e., it is perfused through at least part of the cell mass and up to the whole cell mass.
  • The metabolic product level in the medium is normally maintained within a particular range. Glucose concentration is usually maintained in the range of about 5 to 20 mM. Lactate concentration is usually maintained below 35 mM, preferably below 0.5 mg/ml. Glutamine concentration is generally maintained in the range of from about 1 to 3 mM. Ammonium concentration is usually maintained below about 2.4 mM. These concentrations may be monitored by either periodic or on-line continuous measurements using known methods. See e.g., Caldwell et al, J. Cell. Physiol. (1991) 147:344-353.
  • In a preferred embodiment of the invention, the culture medium is continuously perfused to provide fresh medium at a rate which is proportional to the lactate concentration and/or the cell density in the culture. Preferably, this medium replacement is accomplished without diluting the cell density of the culture.
  • Culture Times
  • The culture time may vary widely. The cells are preferably cultured for at least the minimum amount of time required to produce cells with enhanced replicative potential, biological function, or both. This time may vary with cell type, depending on the cell doubling time. In preferred embodiment, the cells are cultured according to the invention for at least 2 days, more preferably, at least 4 days. The maximum culture time is not particularly limited. For example, the cells may be cultured for up to 10 days, up to 25 days, up to 50 days, up to 75 days, up to 100 days, or longer, if desired (>100 days [e.g., T-cells, Rosenberg]).
  • Cell Culture Apparatus
  • An obstacle with ex vivo expansion has been the ability to reliably generate large quantities of ex vivo expanded lineage committed cells in a controlled process for effective therapy. The traditional cell expansion methods in bags, flasks or roller bottles involves multi-step manual cell culture processes. Due to the lack of continuous medium perfusion features, the cell concentrations are maintained at a low level (˜1×106 cells/ml) by the addition of fresh medium to the culture container or by transferring part of the culture to additional culture containers. Such procedures require numerous aseptic transfers resulting in less reproducible and reliable performance and increased risk of contamination or critical operator error.
  • In a preferred embodiment, an automated clinical-scale cell production system (CPS), which has been developed to process and expand cells in a closed and sterile environment and which is described in allowed U.S. application Ser. No. 08/478,622 (attorney reference: 4292-022-55), incorporated herein by reference, is used. This is capable of exchanging medium at a continuous rate without removal of cells. The use of the CPS requires a single aseptic transfer and is microprocessor controlled, resulting in significant savings in labor and space.
  • The CPS has been automated by incorporating microprocessor controlled hardware and the development of software to run the processes. The. CPS embodies a modular, closed-system process comprised of a pre-sterilized, single-use disposable cell cassette operated by automated instruments. The instrumentation components of the system include an incubator unit and processor unit, along with a computer based system manager.
  • The cell cassette provides a closed, sterile environment in which cell production can occur. The single-use cartridge is provided fully assembled in a sterile package, opened just prior to use. The sterile pathway contained in the cell cassette includes: cell growth chamber, medium supply container, a pump for delivery of components, and sterile barrier elements throughout.
  • A dedicated, small incubator controls the biological and physical environment and operations of each cell cassette necessary to support the cell growth process. The incubator receives and self-engages the disposable cell cassette, much like a VCR and videocassette. The incubator controls: the flow of medium to the growth chamber, the temperature (4° C.) of the growth medium supply compartment, the temperature (37° C.) of the growth chamber compartment, and the concentration and flow rate of gases delivered to the gas compartment of the culture chamber. The incubator also monitors various safety-alarm parameters to assure that the cell production process is proceeding as expected. In the unlikely event of failure of an incubator, the cell cassette, is readily transferred to another incubator with minimal interruption of the culture process.
  • The processor performs the initial priming of the cell cassette with growth medium and, through instructions to the operator, the controlled inoculation of cells. The same unit also performs the removal (harvest) of the cells from the growth chamber at the completion of the cell production process, sterilely transferring the cells to a pre-attached harvest container (analogous to a blood transfusion bag).
  • The system manager employs a user-friendly graphical interface. The system manager provides a convenient central user interface and provides for redundant monitoring of each incubator in the network. The system manager can perform procedure scheduling (up to 50 incubators) tasks for the operators and provide a daily or weekly printed record of alarm events for quality control and record keeping purposes.
  • The ID key contains a semiconductor memory device and clock and is affixed to each cell cassette at the beginning of a cell production procedure. The ID key provides reliable identification of the cell product, instructs the instruments for the cell production process, prevents mix-ups and operator error, and stores the primary data for complete process history record (effectively a “manufacturing batch record” for the cell product).
  • Therapeutic Applications
  • After culturing according to the invention, the lineage committed human cells have enhanced replicative and/or biological function. The cultured cells may be isolated by harvesting them from the culture apparatus. The cells may be harvested by, for example, withdrawing the cells by syringe, or by continuously allowing the cells to flow out of the culture reactor, by the pressure produced by replacing the culture medium, through an exit tube. After harvesting, the cells may be infused in a patient to obtain the therapeutic benefits of the cultured cells.
  • The inventors have discovered that lineage committed cells cultured under conditions of medium exchange according to the present invention have an enhanced ability to replicatate and/or have enhanced biological function after the cells are isolated. Since the cells of the present invention have replicative and/or biological function, fewer cells may be required to achieve a given level of effectiveness as compared to cells that were cultured under static conditions.
  • The procedures for infusing the cells of the present invention for human therapy are well-known, see:
    • Anti-EBV Specific CTLs:
    • Heslop et al. New England Journal of Medicine 331:679-680 (1994),
    • Heslop et al. Nature Medicine 2:551-555 (1996), and
    • Heslop et al. Immunology Reviews 157:217-222 (1997);
    • Adoptive Immunotherapy in Viral Diseases
    • Riddell et al. Annual Reviews in Immunology 13:545-3159 (1991),
    • Anti-CMVCTLs, and
    • Walter et al. New England Journal of Medicine 333:1038-1044 (1995);
    • Anti-HIV CTLs
    • Levine et al. Science 272:1939-1943 (1996);
    • Anti-Tumor CTLs
    • Anichini et al. Journal of Immunology 156:206-217 (1996),
    • Cardoso et al. Blood 90:549-561 (1997), and
    • Schultze et al. Blood 89:3806-3816 (1997); and
    • Dendritic Cells
    • Choudhury A, Gajewski, J L, Liang, J C, Popat, U, Claxton, D F, Kliche K O, Andreef M, Champlin, R E, Blood 89:1133 (1997),
    • Girolomoni G, Ricciardi-Castagnoli, P: Immunol. Today 18, 102 (1997),
    • Hsu, F J, Benike, C, Fagnoni F, Liles T M, Czerwinski D, Taidi B, Engleman, E G, Levy R, Nature Med. 2: 52 (1996), and
    • Mayordomo J I, Zorina T, Storkus W J, Zitvogel, L, Garcia-Prats M D, DeLeo A B, Lotze M T: Stem Cells 15: 94 (1997).
  • Having generally described this invention, a further understanding can be obtained by reference to certain specific examples which are provided herein for purposes of illustration only and are not intended to be limiting unless otherwise specified.
  • EXAMPLES Examlple 1 Large Scale Ex Vivo Expansion of Human T-Lymphocytes
  • The growth of human T-lymphocytes in a CPS was demonstrated using a model system in which peripheral blood CD8+ T-cells from normal donors were activated using PHA. CD8+ cells were inoculated into the CPS at a density of 20-80 million cells (0.08×106-0.32×106 cells/ml) in AIM-V medium containing 600 IU/ml of rIL-2. A yield of 3-8 billion cells (12×106-32×106 cells/ml) (>90% viability) was obtained at harvest on day 10 (n=12). Continuous one pass perfusion of fresh medium enabled the CPS to maintain high cell densities and low lactate concentrations (<0.5 mg/ml). Under these conditions the total medium exchanged was 4-6 liters. Polyclonal T-cell expansion in CPS was demonstrated by flow cytometric analysis using a panel of 10 different T-cell receptor (TCR) Vα and Vβ-subfamily-specific mAbs. Each TCR family was represented at similar levels in the CPS cultures compared to conventional control T-flask cultures. Furthermore, harvested T-lymphocytes secreted IFN-γ (490-2300 pg/ml), TNF-α (190-880 pg/ml), GM-CSF (1900-5100 pg/ml) and IL-10 (5-35 pg/ml) in response to anti-CD3 mAb. These-findings indicate that human T-cells expanded in the CPS are fully responsive to stimulation through the TCR. In conclusion, these studies demonstrate the potential of the CPS for large scale expansion of functional effector T-cells for use in human T-cell therapies.
  • Example 2 Enhancing the Proliferative Potential and Biological Effector Function of Human T-Lymphocytes
  • The growth of PHA-activated human CD8+ T-cells was used as a model system for evaluating the effects of continuous perfusion on T-cell expansion. Human CD8+ T-cells were expanded using continuous one-pass perfusion with medium containing IL-2 in the CPS for ten days. The perfusion rates were increased with the increase in cell number such that lactate concentration in the spent medium was maintained at 0.5 mg/ml. The continuous perfusion of medium resulted in high density cultures of concentrations ranging from 12 to 32 million cells/ml starting from a population of 0.16-0.32 million cells/ml. Parallel T-flask cultures were set up at the same culture conditions (vis. inoculum density, culture depth, oxygenation, and medium composition). These cultures were hemidepleted to 0.5×109 cells/ml when the cell densities reached 1.0×109 cells/ml by manual removal of spent medium and cells and replenishing with fresh medium. The ex vivo expanded T-cells harvested from the CPS and T-flasks on day 10 (also called primary cultures) were further expanded in presence of IL-2 for four days (secondary cultures) in a static culture medium. The results of the expansions from three experiments are shown in Table 1.
  • The expansion in the CPS during the ten days of primary culture was lower by 1.85-4.63 fold compared to T-flasks. However, subsequent evaluation (secondary culture) of the replicative potential of CPS and T-flask expanded cells revealed 12.7-42.2 fold higher expansion with the CPS derived cells in the secondary culture. In this secondary culture both the CPS-expanded and the T-flask-expanded cells were cultured under static medium conditions in a hemidepleted T-flask. Thus, the overall expansion, i.e., after primary and secondary culturing, of CPS derived cells was 3.8-10.3 fold higher than in hemidepleted T-flask cultures. These results demonstrate the benefit of continuous perfusion in enhancing the replicative potential of human CD8+ cells.
  • The functionality of the T-cell harvested from the CPS or parallel T-flask cultures was assessed by analysis of cytokine release (see FIG. 1). T-cells harvested from the CPS or T-flasks produced similar concentrations of IFN-γ in response to anti-CD3 mAb. In contrast, T-cells derived from the CPS produced higher concentrations of TNF-α and GM-CSF than T-cells derived from T-flasks. The data indicates that T-cells derived in the CPS are fully responsive to stimulation through the TCR-CD3 complex and may produce higher levels of particular cytokines on a per cell basis than T-cells produced in T-flasks.
  • Significance
  • The enhanced proliferative potential of T-cells derived in the CPS using continuous perfusion has important applications for the therapeutic efficacy of these T-cells after reinfusion. Effective adoptive immunotherapy for viral diseases and cancer requires the reliable generation of large quantities of functional cells capable of continued cell expansion in vivo together with recirculation and localization of effector T-cells at sites of various infection or tumor cell growth. Expansion of T-cells using continuous perfusion in the CPS results in large numbers of high quality effector T-cells with enhanced replication potential and effector function.
    TABLE 1
    Post-Expansion Replicative Potential
    Secondary Increased
    Primary Culture Culture2 Fold Relative
    Fold Expansion Fold Expansion Expansion
    Exp. Device1 (10 days) Expansion Overall3 in CPS
    #
    1 CPS 65 40 2600 10.3
    T-flasks 120 2.1 252
    #2 CPS 93 152 14136 9.1
    T-flasks 431 3.6 1552
    #3 CPS 57 33 1881 3.8
    T-flasks 190 2.6 494

    1T-25 flasks; hemidepletion.

    2Secondary culturing was conducted in T-25 flasks, hemidepletion, for both CPS- and T-flask expanded cells. The values indicate fold expansion after 4 days.

    3Represents (fold expansion in primary culture) ×(fold expansion in secondary culture)
  • Example 3 Clinical Scale Expansion of Human Dendritic Cells in a Continuously Perfused Bioreactor System
  • The purpose of the present studies was to develop and implement a process for expansion of human dendritic cells (DCs) in a continuously perfused clinical scale bioreactor system. Experiments in small scale cultures were conducted to evaluate the effect of frequent medium exchange on dendritic cell expansion and function. Peripheral blood mononuclear cells (PB MNCs) from normal donors were inoculated at low (1×106 cells/cm2) an high (5×106 cells/cm2) inoculum densities in T25 cm2 flasks. Non-plastic adherent cells were gently removed after 2 hours at 37° C., and adherent cells comprising approximately 50% of the total inoculated cell population were cultured for 7 days. The cultures either were not fed, or received 50% fresh serum-free AIM V medium containing GM-CSF and IL-4 on days 2, 4 and 6. TNFα was added on day 6 for the final 24 hours of culture. Cells from these cultures expressed the characteristic “veiled” morphology and surface phenotype of dendritic cells as determined by the cytometry. Furthermore, DCs demonstrated at least 50-fold greater stimulating activity in the alloMLR compared to PB MNCs. Interestingly, frequent medium exchange, particularly at high inoculum density, significantly enhanced the stimulating activity of harvested DCs (>5-fold compared to static culture conditions. Total cell recovery correlated well with lactate produced per culture supporting the utility of lactate measurement as a non-invasive means to improve medium exchange protocols and monitor DC culture productivity. Initial experiments in a CPS at an inoculum density of 1.6×106 cells/cm2 produced 295.3×106 dendritic cells at harvest on Day 7. Experiments are in progress to optimize the expansion and function of dendritic cells at high density in the CPS under condition of continuous medium perfusion. These studies demonstrate that continuous medium perfusion significantly improves the quantity and biological function of harvested DCs.
  • Results obtained with such small-scale cultures are shown in FIGS. 2-4. The cultures represented in FIGS. 2, 3, and 4 were conducted at low, intermediate, and high inoculum density, respectively. Panel (A) of each Figure shows the lactate concentration as a function of time during the seven day culture period. Panel (B) of each Figure shows the mixed leucocyte response (MLR) observed with the harvested DCs. In these assays, the harvested DCs, which were derived from one individual, were contacted with the T-cells of another individual. The MLR response was determined by measuring the uptake of 3-H-thymidine in the stimulated T-cells (a measure of DNA synthesis in these T-cells) and is shown as “CPM” plotted against the number of harvested DCs in contact with the T-cells. The data in FIG. 2 shows the results obtained with 100% medium exchange and 0% medium exchange. In the medium-exchanged culture, 50% medium replacement was performed on day 4 and 7 (indicated by arrows in the Figure). The data in Figure. 3 shows the results observed with 0% medium exchange and 150% medium exchange. In the medium-exchanged culture, 50% medium replacement was performed on day 3, 5, and 7 (indicated by arrows). FIG. 4 shows the results of culturing using 100% and 300% medium exchange. In the 100% exchange culture, the 50% of the medium was replaced at day 4 and 7 (indicated by the arrows in the Figure). For 300% medium exchange, 50% of the culture medium was replaced on day 2, 3, 4, 5, 6, and 7 (represented as a solid line in FIG. 4). These results show that increased rates of culture medium exchange produces harvested DCs with enhanced an enhanced ability to stimulate T-cells.
  • Example 4 Preliminary Optimization for Expansion of Chondrocytes
  • To explore the benefits of medium exchange rate/perfusion with chondrocytes preliminary experiments were conducted with canine chondrocytes in a manual small scale system. Cells expanding in monolayer cultures were trypsinized and inoculated at different cell densities per cm2 of the culture vessel (T-flask). The cultures were then expanded for several days at two rates of medium exchange (i) 50% medium exchange on day 3 and 5, and (ii) 50% medium exchange on days 2, 4, and 6. As is evident from FIG. 5, medium exchange rate can have a pronounced effect on the cell production. By increasing the medium exchange rate, a lower inoculum density can result in similar yield as two-fold higher inoculum density at lower medium exchange rate. The effects of perfusion rates becomes particular significant when the number of cells available for expansion is limited, resulting in more reliable generation of a target cell dose.
  • All publications and patent applications cited in this disclosure are incorporated herein by reference in their entirety.
  • Obviously, additional modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein.

Claims (33)

1-45. (canceled)
46. A composition comprising lineage committed dendritic cells exhibiting enhanced biological function as compared to the biological function of the lineage committed dendritic cells cultured ex vivo under conditions which do not include replacement of the liquid culture medium during the culturing.
47. The composition of claim 46, wherein the biological function enhanced in the isolated lineage committed dendritic cells comprises increased release of cytokines.
48. The composition of claim 46, the lineage committed dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of at least 25% daily replacement for more than one day.
49. The composition of claim 46, the lineage committed dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of at least 50% daily replacement for more than one day.
50. The composition of claim 46, wherein the lineage committed dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of from 25% to 100% daily replacement for about 1×104 to about 1×107 cells/ml culture for more than one day.
51. The composition of claim 46, wherein the dendritic cells are antigen primed dendritic cells.
52. The composition of claim 46, wherein the dendritic cells are myeloid derived dendritic cells.
53. The composition of claim 46, wherein the dendritic cells are non-myeloid derived dendritic cells.
54. A composition comprising lineage committed human dendritic cells exhibiting enhanced biological function as compared to the biological function of the lineage committed human dendritic cells cultured ex vivo under conditions which do not include replacement of the liquid culture medium during the culturing.
55. The composition of claim 54, wherein the biological function enhanced in the isolated lineage committed human dendritic cells comprises increased release of cytokines.
56. The composition of claim 54, the lineage committed human dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of at least 25% daily replacement for more than one day.
57. The composition of claim 54, the lineage committed human dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of at least 50% daily replacement for more than one day.
58. The composition of claim 54, wherein the lineage committed human dendritic cells are cultured under conditions where the liquid culture medium is replaced at a rate of from 25% to 100% daily replacement for about 1×104 to about 1×107 cells/ml culture for more than one day.
59. The composition of claim 54, wherein the human dendritic cells are antigen primed dendritic cells.
60. The composition of claim 54, wherein the human dendritic cells are myeloid derived dendritic cells.
61. The composition of claim 54, wherein the human dendritic cells are non-myeloid derived dendritic cells.
62. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 46.
63. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 47.
64. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 48.
65. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 49.
66. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 50.
67. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 51.
68. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 52.
69. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 53.
70. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 54.
71. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 55.
72. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 56.
73. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 57.
74. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 58.
75. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 59.
76. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 60.
77. A method of treating a human patient in need of an infusion of lineage committed human dendritic cells, comprising administering to said patient a composition according to claim 61.
US11/295,596 1998-02-23 2005-12-07 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses Abandoned US20060093581A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/295,596 US20060093581A1 (en) 1998-02-23 2005-12-07 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US09/027,671 US20020034819A1 (en) 1998-02-23 1998-02-23 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US09/893,470 US6835566B2 (en) 1998-02-23 2001-06-29 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US10/815,707 US20040185557A1 (en) 1998-02-23 2004-04-02 Human linkeage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US11/295,596 US20060093581A1 (en) 1998-02-23 2005-12-07 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/815,707 Continuation US20040185557A1 (en) 1998-02-23 2004-04-02 Human linkeage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses

Publications (1)

Publication Number Publication Date
US20060093581A1 true US20060093581A1 (en) 2006-05-04

Family

ID=21839121

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/027,671 Abandoned US20020034819A1 (en) 1998-02-23 1998-02-23 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US09/893,470 Expired - Lifetime US6835566B2 (en) 1998-02-23 2001-06-29 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US10/668,214 Abandoned US20040062755A1 (en) 1998-02-23 2003-09-24 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US10/815,707 Abandoned US20040185557A1 (en) 1998-02-23 2004-04-02 Human linkeage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US11/295,596 Abandoned US20060093581A1 (en) 1998-02-23 2005-12-07 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both: methods for obtaining same; and their uses

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US09/027,671 Abandoned US20020034819A1 (en) 1998-02-23 1998-02-23 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US09/893,470 Expired - Lifetime US6835566B2 (en) 1998-02-23 2001-06-29 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US10/668,214 Abandoned US20040062755A1 (en) 1998-02-23 2003-09-24 Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US10/815,707 Abandoned US20040185557A1 (en) 1998-02-23 2004-04-02 Human linkeage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses

Country Status (1)

Country Link
US (5) US20020034819A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2314300A1 (en) * 2006-11-03 2011-04-27 Aastrom Biosciences, Inc. Mixed cell populations for tissue repair and separation technique for cell processing

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020034819A1 (en) * 1998-02-23 2002-03-21 Alan K. Smith Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
US20040076620A1 (en) * 2002-09-03 2004-04-22 Donnie Rudd Method of repairing primate mammalian tissue
US20040043009A1 (en) * 2002-09-03 2004-03-04 Donnie Rudd Method of repairing primate mammalian tissue
EP1670513B1 (en) * 2003-10-06 2013-01-23 Cedars-Sinai Medical Center Cox-2 inhibitors and dendritic cells for use in the treatment of cancer.
EP1676132B1 (en) * 2003-10-21 2014-01-22 Cedars-Sinai Medical Center Combination of chemotherapy and administration of glioma-antigen-pulsed dendritic cells in the treatment of glioma
EP1773981A1 (en) * 2004-07-12 2007-04-18 Sorin Group Italia S.R.L. Device and method for growing human cells
US20070077231A1 (en) * 2005-09-30 2007-04-05 Contag Christopher H Immune effector cells pre-infected with oncolytic virus
US8129184B2 (en) * 2006-09-26 2012-03-06 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
US8871211B2 (en) 2006-09-28 2014-10-28 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
EP2115116B1 (en) * 2007-03-05 2018-01-24 Terumo BCT, Inc. Methods to control cell movement in hollow fiber bioreactors
JP2010519937A (en) 2007-03-05 2010-06-10 カリディアンビーシーティー、インコーポレーテッド Cell proliferation system and method of use
EP2152851B1 (en) * 2007-04-06 2018-04-25 Terumo BCT, Inc. Improved bioreactor surfaces
EP2148922B1 (en) * 2007-04-13 2020-03-04 Terumo BCT, Inc. Cell expansion system and methods of use
AU2009203893B2 (en) * 2008-01-08 2014-10-02 The University Of Queensland Method of producing a population of cells
DK2328923T3 (en) * 2008-09-02 2016-03-21 Cedars Sinai Medical Center CD133 epitopes
WO2010129895A2 (en) * 2009-05-07 2010-11-11 Immunocellular Therapeutics, Ltd. Cd133 epitopes
EP2566951B1 (en) 2010-05-05 2020-03-04 Terumo BCT, Inc. Method of reseeding adherent cells grown in a hollow fiber bioreactor system
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
CN104540933A (en) 2012-08-20 2015-04-22 泰尔茂比司特公司 Method of loading and distributing cells in a bioreactor of a cell expansion system
US20140234351A1 (en) 2013-02-14 2014-08-21 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods
JP2016523125A (en) 2013-05-30 2016-08-08 グラハム エイチ. クリーシー Local nervous stimulation
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
WO2015164808A1 (en) 2014-04-24 2015-10-29 Terumo Bct, Inc. Measuring flow rate
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
US9550972B2 (en) 2014-09-29 2017-01-24 General Electric Company Devices, systems and methods for automated cell culturing
US11077301B2 (en) 2015-02-21 2021-08-03 NeurostimOAB, Inc. Topical nerve stimulator and sensor for bladder control
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2019094365A1 (en) 2017-11-07 2019-05-16 Neurostim Oab, Inc. Non-invasive nerve activator with adaptive circuit
CN110438076A (en) * 2018-05-02 2019-11-12 强普生技股份有限公司 T cell culture medium and cultural method
EP3990100A4 (en) 2019-06-26 2023-07-19 Neurostim Technologies LLC Non-invasive nerve activator with adaptive circuit
JP2023506713A (en) 2019-12-16 2023-02-20 ニューロスティム テクノロジーズ エルエルシー Noninvasive nerve activator using booster charge delivery

Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4135975A (en) * 1977-12-14 1979-01-23 Lichtman Marshall A Obtaining human cell lines that elaborate colony stimulating activity for marrow cells of man and other species and methods of preparing same
US4481946A (en) * 1980-08-14 1984-11-13 Altshuler John H Bone marrow transplant method and apparatus
US4486188A (en) * 1980-08-14 1984-12-04 Applied Medical Devices, Inc. Bone marrow transplant method and apparatus
US4514499A (en) * 1983-02-04 1985-04-30 Corning Glass Works Cell culture using a monolithic support
US4714680A (en) * 1984-02-06 1987-12-22 The Johns Hopkins University Human stem cells
US4808611A (en) * 1986-07-30 1989-02-28 Immunex Corporation Use of interleukin-1 to induce development of multipotent hemopoietic cell populations
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4847201A (en) * 1985-02-05 1989-07-11 Cetus Corporation DNA encoding for CSF-1 and accompanying recombinant systems
US4861714A (en) * 1985-04-04 1989-08-29 Verax Corporation Weighted collagen microsponge for immobilizing bioactive material
US4889812A (en) * 1986-05-12 1989-12-26 C. D. Medical, Inc. Bioreactor apparatus
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US4965204A (en) * 1984-02-06 1990-10-23 The Johns Hopkins University Human stem cells and monoclonal antibodies
US5004681A (en) * 1987-11-12 1991-04-02 Biocyte Corporation Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5032407A (en) * 1987-01-16 1991-07-16 Ohio University Edison Animal Biotechnology Center Gene transfer using transformed, neodetermined, embryonic cells
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5399493A (en) * 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
US5605822A (en) * 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5635386A (en) * 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
US5646043A (en) * 1989-06-15 1997-07-08 Regents Of The University Of Michigan Methods for the ex vivo replication of human stem cells and/or expansion of human progenitor cells
US5674750A (en) * 1995-05-19 1997-10-07 T. Breeders Continuous selective clonogenic expansion of relatively undifferentiated cells
US5728581A (en) * 1995-06-07 1998-03-17 Systemix, Inc. Method of expanding hematopoietic stem cells, reagents and bioreactors for use therein
US5811094A (en) * 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5851832A (en) * 1991-07-08 1998-12-22 Neurospheres, Ltd. In vitro growth and proliferation of multipotent neural stem cells and their progeny
US5922597A (en) * 1995-11-14 1999-07-13 Regents Of The University Of Minnesota Ex vivo culture of stem cells
US5925567A (en) * 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US5945225A (en) * 1996-12-18 1999-08-31 Hoechst Trespaphan Gmbh Oriented multilayer polyolefin film which is heat-sealable on both sides, its use, and process for its production
US5994126A (en) * 1992-04-01 1999-11-30 The Rockefeller University Method for in vitro proliferation of dendritic cell precursors and their use to produce immunogens
US6299650B1 (en) * 1996-03-01 2001-10-09 Isotis N.V. Method for in vitro production of bone
US6326198B1 (en) * 1990-06-14 2001-12-04 Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US20040062755A1 (en) * 1998-02-23 2004-04-01 Aastrom Biosciences, Inc. Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US481946A (en) * 1892-09-06 Spring-tooth attachment for cultivators
JPS58208046A (en) * 1982-05-28 1983-12-03 東洋製罐株式会社 Plastic vessel with oriented coating and its manufacture
US5180662A (en) * 1988-01-05 1993-01-19 The United States Of America As Represented By The Department Of Health And Human Services Cytotoxic T lymphocyte activation assay
US5858358A (en) * 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
JPH04506153A (en) 1989-06-15 1992-10-29 ザ・リージエンツ・オブ・ザ・ユニバーシテイ・オブ・ミシガン Cell proliferation methods, compositions and devices for cell proliferation
FR2660323B1 (en) * 1990-03-30 1992-07-24 Bertin & Cie CELL CULTURE DEVICE.
US5622853A (en) 1990-05-01 1997-04-22 Becton Dickinson And Company T lymphocyte precursor
US5401651A (en) * 1991-10-16 1995-03-28 Walz; Alfred DNA encoding ENA-78, a neutrophil activating factor
EP0631618A4 (en) 1992-03-04 1995-09-27 Systemix Inc Culturing of hematopoietic stem cells and their genetic engineering.
WO1993018136A1 (en) 1992-03-05 1993-09-16 Cytomed, Inc. Process for supporting hematopoietic progenitor cells
EP0716570A4 (en) 1993-09-03 1997-12-17 Systemix Inc Genetically modified human hematopoietic stem cells and their progeny
SE508468C2 (en) * 1996-10-08 1998-10-12 Sandvik Ab Rock drill bit for rotating, crushing machining of rock and sealing means for such rock drill bit
US6306576B1 (en) * 1999-02-19 2001-10-23 Cleveland Clinic Foundation Diagnostic methods for asthma

Patent Citations (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4135975A (en) * 1977-12-14 1979-01-23 Lichtman Marshall A Obtaining human cell lines that elaborate colony stimulating activity for marrow cells of man and other species and methods of preparing same
US4481946A (en) * 1980-08-14 1984-11-13 Altshuler John H Bone marrow transplant method and apparatus
US4486188A (en) * 1980-08-14 1984-12-04 Applied Medical Devices, Inc. Bone marrow transplant method and apparatus
US4514499A (en) * 1983-02-04 1985-04-30 Corning Glass Works Cell culture using a monolithic support
US4965204A (en) * 1984-02-06 1990-10-23 The Johns Hopkins University Human stem cells and monoclonal antibodies
US4714680A (en) * 1984-02-06 1987-12-22 The Johns Hopkins University Human stem cells
US4714680B1 (en) * 1984-02-06 1995-06-27 Univ Johns Hopkins Human stem cells
US4847201A (en) * 1985-02-05 1989-07-11 Cetus Corporation DNA encoding for CSF-1 and accompanying recombinant systems
US4861714A (en) * 1985-04-04 1989-08-29 Verax Corporation Weighted collagen microsponge for immobilizing bioactive material
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4889812A (en) * 1986-05-12 1989-12-26 C. D. Medical, Inc. Bioreactor apparatus
US4808611A (en) * 1986-07-30 1989-02-28 Immunex Corporation Use of interleukin-1 to induce development of multipotent hemopoietic cell populations
US5032407A (en) * 1987-01-16 1991-07-16 Ohio University Edison Animal Biotechnology Center Gene transfer using transformed, neodetermined, embryonic cells
US4963489A (en) * 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5004681A (en) * 1987-11-12 1991-04-02 Biocyte Corporation Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5004681B1 (en) * 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5032508A (en) * 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5670147A (en) * 1989-06-15 1997-09-23 Regents Of The University Of Michigan Compositions containing cultured mitotic human stem cells
US5399493A (en) * 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
US5605822A (en) * 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5635386A (en) * 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
US5646043A (en) * 1989-06-15 1997-07-08 Regents Of The University Of Michigan Methods for the ex vivo replication of human stem cells and/or expansion of human progenitor cells
US5670351A (en) * 1989-06-15 1997-09-23 The Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of human hematopoietic stem cells
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US6326198B1 (en) * 1990-06-14 2001-12-04 Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5811094A (en) * 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5851832A (en) * 1991-07-08 1998-12-22 Neurospheres, Ltd. In vitro growth and proliferation of multipotent neural stem cells and their progeny
US5994126A (en) * 1992-04-01 1999-11-30 The Rockefeller University Method for in vitro proliferation of dendritic cell precursors and their use to produce immunogens
US5925567A (en) * 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US5674750A (en) * 1995-05-19 1997-10-07 T. Breeders Continuous selective clonogenic expansion of relatively undifferentiated cells
US5728581A (en) * 1995-06-07 1998-03-17 Systemix, Inc. Method of expanding hematopoietic stem cells, reagents and bioreactors for use therein
US5922597A (en) * 1995-11-14 1999-07-13 Regents Of The University Of Minnesota Ex vivo culture of stem cells
US6299650B1 (en) * 1996-03-01 2001-10-09 Isotis N.V. Method for in vitro production of bone
US5945225A (en) * 1996-12-18 1999-08-31 Hoechst Trespaphan Gmbh Oriented multilayer polyolefin film which is heat-sealable on both sides, its use, and process for its production
US20040062755A1 (en) * 1998-02-23 2004-04-01 Aastrom Biosciences, Inc. Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2314300A1 (en) * 2006-11-03 2011-04-27 Aastrom Biosciences, Inc. Mixed cell populations for tissue repair and separation technique for cell processing

Also Published As

Publication number Publication date
US20040062755A1 (en) 2004-04-01
US20040185557A1 (en) 2004-09-23
US20020031498A1 (en) 2002-03-14
US20020034819A1 (en) 2002-03-21
US6835566B2 (en) 2004-12-28

Similar Documents

Publication Publication Date Title
US6835566B2 (en) Human lineage committed cell composition with enhanced proliferative potential, biological effector function, or both; methods for obtaining same; and their uses
Brasel et al. Generation of murine dendritic cells from flt3-ligand–supplemented bone marrow cultures
CN107922925B (en) Method for natural killer cell expansion
Son et al. A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells
Paquette et al. Interferon‐α and granulocyte‐macrophage colony‐stimulating factor differentiate peripheral blood monocytes into potent antigen‐presenting cells
Desmedt et al. Macrophages induce cellular immunity by activating Th1 cell responses and suppressing Th2 cell responses
US7198948B2 (en) Methods and compositions for obtaining mature dendritic cells
US6458585B1 (en) Cytokine-free culture of dendritic cells
Skalova et al. Human myeloid dendritic cells for cancer therapy: does maturation matter?
CN107475196A (en) The amplification cultivation method of NK culture matrix and NK
WO1997029182A9 (en) Method and compositions for obtaining mature dendritic cells
JP2001520509A (en) Autoimmune cell therapy: cell compositions, methods and applications in the treatment of human diseases
CN107460168A (en) The amplification cultivation method of NK culture matrix and NK
Pullarkat et al. Large-scale monocyte enrichment coupled with a closed culture system for the generation of human dendritic cells
CN109825473A (en) A kind of cultural method of the autologous peripheral blood lymphocyte using the stimulation of TLR7 agonist
CN107446888B (en) The application of NK cell culture mediums, cultural method and the two
Pierson et al. Production of human natural killer cells for adoptive immunotherapy using a computer-controlled stirred-tank bioreactor
CN112608896A (en) NK cell culture method and application thereof
Helinski et al. Long-term cultivation of functional human macrophages in Teflon dishes with serum-free media
Syme et al. Generation of dendritic cells: role of cytokines and potential clinical applications
EP0205387B1 (en) Human monocytes cultured in suspension in serum-free medium
Jackson et al. Characterization of an early dendritic cell precursor derived from murine lineage-negative hematopoietic progenitor cells
Syme et al. Effects of cytokines on the culture and differentiation of dendritic cells in vitro
CA2066645A1 (en) Method for the production of in vitro expanded lymphoid cells for use in adoptive immunotherapy
AU4052197A (en) Cytokine-free culture of dendritic cells

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION