WO1999060105A1 - Flat sheet cell encapsulation device - Google Patents

Flat sheet cell encapsulation device Download PDF

Info

Publication number
WO1999060105A1
WO1999060105A1 PCT/IB1999/000889 IB9900889W WO9960105A1 WO 1999060105 A1 WO1999060105 A1 WO 1999060105A1 IB 9900889 W IB9900889 W IB 9900889W WO 9960105 A1 WO9960105 A1 WO 9960105A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
cell matrix
semi
matrix system
Prior art date
Application number
PCT/IB1999/000889
Other languages
French (fr)
Inventor
Markus Borkenhagen
Original Assignee
Modex Therapeutiques, S.A.
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 Modex Therapeutiques, S.A. filed Critical Modex Therapeutiques, S.A.
Priority to AU36231/99A priority Critical patent/AU3623199A/en
Publication of WO1999060105A1 publication Critical patent/WO1999060105A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • 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
    • 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
    • A61K2035/126Immunoprotecting barriers, e.g. jackets, diffusion chambers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the present invention relates generally to cell encapsulation devices, and more particularly to devices for cell screening and implantation into a mammalian host.
  • xenogenic or allogenic cells are isolated from the host's immune system by surrounding the cells in a semi-permeable membrane prior to implantation within the host.
  • This semi -permeable membrane jacket is relatively impermeable to large molecules, such as components of the host's immune system, but is permeable to small molecules.
  • the semi-permeable membrane jacket has a molecular weight cutoff of between 50 and 2000 kilodaltons (kDa).
  • the semi-permeable membrane allows the implanted cells to receive nutrients necessary for viability and allows metabolic waste to be removed.
  • the membrane also allows therapeutic molecules produced by the implanted cells to diffuse to host cells.
  • endogenous proteins or those cloned into the cell are delivered to the host.
  • an immuno-protective, semi-permeable membrane now allows transplantation of encapsulated cells from one species into a host from a different species without the risk of immune rejection or use of immunosuppressive drugs.
  • Applications of encapsulated cell therapy include, for example, treatments for diabetes, hemophilia, anemia, ⁇ -thalassemia, Parkinson's disease, and amyotropic lateral sclerosis.
  • the use of biologically compatible polymeric materials in construction of an encapsulation device is critical to a successful cell encapsulation therapy.
  • Important components of the encapsulation device include the surrounding semi-permeable membrane and the internal cell-supporting matrix, or scaffold.
  • the scaffold defines the microenvironment for the encapsulated cells and keeps the cells well distributed within the intracapsular compartment.
  • the optimal internal scaffold for a particular cell encapsulation device is highly dependent on the cell type. For example, while adherent cells often prefer a solid surface on which to lie, suspension cells may prefer a hydrophilic lightly cross-linked hydrogel as a matrix material.
  • adherent cells In the absence of a scaffold, adherent cells aggregate to form clusters. When the clusters grow too large, they typically develop a central necrotic core. Dying cells accumulate around the core and, upon lysing, release factors detrimental to the health of neighboring cells. The lysed cell fragments are also transported to the host environment, there eliciting an antigenic response.
  • the prior art discloses that the position of the cells may be stabilized by an internal scaffold composed of an immobilizing matrix formed from a hydrogel.
  • a "hydrogel” is a 3 -dimensional network of cross-linked hydrophilic polymers, in the form of a gel and substantially (e.g., greater than 90%) composed of water.
  • cells are encapsulated within an immunoisolatory membrane and then allowed to continue to proliferate. Once the cells are placed inside the encapsulating membrane, it is extremely difficult to characterize their morphology. Moreover, it is nearly impossible to control cell proliferation and functionality. As a result, devices made using methods of the prior art exhibit wide ranges of total expression of the desirable secreted protein. Consequently, it is difficult to control the therapeutic dosage received by the host.
  • the invention provides a cell implantation device.
  • the device has a cell matrix, or scaffold, affixed to a supporting frame.
  • the supporting frame is preferably a flat, ring-shaped element.
  • Cells are inoculated onto the matrix. Once the cells exhibit the desired morphology, the cell-containing matrix is encapsulated in a semi-permeable immunoisolatory membrane.
  • the cell matrix, or scaffold, in the device of the invention advantageously provides cells with a template for cellular organization in a three-dimensional orientation resembling their typical physiological shape.
  • the cell matrix is particularly useful for the use of suspension cells for encapsulated cell therapy.
  • the matrix is composed of a hydrogel material selected from agarose, alginate, chitosan, collagen, Matrigel®, Vitrogen®, or PVA foam.
  • the hydrogel is agarose.
  • the selected cells are mixed with a hydrogel solution at a desired cell density and a desired concentration of gel polymer.
  • the cell/hydrogel suspension is cast into the structural support which serves as a mold while the suspension solidifies into the cell-supporting matrix.
  • the matrix solidifies, mechanical forces, surface tension, and capillary forces hold the matrix in the structural support.
  • the hydrogel solution becomes gelled and acts as a supporting matrix for the cells.
  • the matrix/support can then be encapsulated in the semi-permeable membrane.
  • the cell matrix is composed of fibers selected from the group consisting of silk, cotton, chitin, for example, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, or polybutester.
  • the matrix is polyethyleneterephthalate (PET) fiber.
  • PET polyethyleneterephthalate
  • the fibrous material can be coated with a protective coating of hydrogel prior to encapsulation in the membrane.
  • the matrix is a cell-supporting fiber wound or woven around the structural support. PET fibers can be wound around a ring shaped structural support.
  • the structural support is a polyvinyl alcohol sponge incorporated into the structural support.
  • a desired cell type is then inoculated onto the selected solid matrix and cultured in the appropriate medium.
  • FIG. 1 shows three top views of components of the device.
  • FIG. 1 A is a representation of two structural support components.
  • FIG. IB is a representation of two structural support components, each fabricated with a cell/agarose component.
  • FIG. 1C is a representation of two structural support components fabricated with PET fibers.
  • FIG. 2 is a schematic representation of two preferred device fabrication processes.
  • FIG. 3 is a graph showing mEPO output over time for devices with cell captured in an agarose matrix and cultured in various media.
  • FIG. 4 is a graph showing mEPO output over time for devices with cell captured in a PET fiber matrix and cultured in various media.
  • FIG. 5 is a graph showing mEPO output over time for pre- and post-transplant devices cultured in various media.
  • FIG. 6 is a graph showing the diffusive properties of a dip-coated membrane fabricated with 8 % PAN/PVC + 20 % PVP (10 6 daltons [Da]).
  • FIG. 7 is a graph showing hematocrit levels over time in rats which were implanted with the device subcutaneously.
  • FIG. 8 is a graph showing hematocrit levels over time in rats which were implanted with the device interperitoneally.
  • FIG. 9 shows the viability scores of devices which were implanted syngenically and allogenically.
  • the invention provides a cell encapsulation device configured as a flat sheet.
  • the device has (1) a structural support or supporting frame element (e.g., in the shape of a ring); (2) matrices to support the cells to be transplanted; and (3) a semi-permeable membrane that permits diffusion of nutrients, waste, and other small molecules to and from the transplanted cells, while isolating those cells from the host.
  • the invention contemplates the use of various matrices, or matrix systems, to support the cells to be encapsulated.
  • the selection of a matrix may depend on the cell type used, with a particular cell type preferentially adhering and growing onto one matrix over another.
  • the matrix is a hydrogel.
  • a "hydrogel” is a substance formed when a polymer is cross linked to create a 3-dimensional structure, entrapping water molecules to form a gel.
  • FIG. 1A shows two examples of a structural support, each having a tab for handling the device.
  • FIG. IB and FIG. 1C show the structural support fixedly coupled to the cell matrix system.
  • the cell matrix system is a cell/agarose mixture.
  • the cell matrix system is the fibrous material, preferably PET fibers, which is wound or otherwise fixedly coupled to the structural support.
  • the support can be dipped into a cell/agarose suspension and allowed to attain the desired conformation (see, FIG. 2, left side of the FIG. (A)).
  • a support containing the cell supporting fiber can be placed in a Petri dish on a non-adherent surface suitable to culture cells, such as, for example, non-adherent agarose
  • the cells to be implanted are then inoculated onto the fiber (see, FIG. 2, right side of the FIG. (B)).
  • the cells are then cultured until they attach to the fiber.
  • An extracellular matrix may be used to coat the fiber to aid cell attachment.
  • fibers suitable for use in the present invention include natural fibers such cotton, silk, chitin or carbon.
  • Non-naturally occurring fibers such as, for example, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, or polybutester are also suitable for use in the present invention.
  • the fiber is PET, which is typically comprised of bundles of approximately 80 PET monofilaments.
  • the diameter of the monofilaments selected can range between 1-30 ⁇ m. Monofilaments having a range of diameters between approximately 1-10 ⁇ m are preferred, with a diameter of 5 ⁇ m being most preferred.
  • the type of fiber selected and the dimensions of the fiber may depend on cell type as one cell type may prefer one type of fiber over another. Additionally, the fibrous matrix selected may be coated with a protective layer of hydrogel prior to encapsulation in the membrane.
  • the structural component to support the cell matrix system can be formed in many different dimensions and geometric shapes.
  • the supports may be circular, elliptical, square, rectangular, triangular, or star shaped. Since devices are fabricated as flat sheets, the thickness of the ring is much less than the diameter of the ring. TABLE 1 lists the dimensions of devices that have been fabricated.
  • the structural support can be made by methods well known to those skilled in the art.
  • the structural support can be made, for example, from a polymeric solution cast into a mold or by stamping, cutting, and extruding.
  • molds for casting polymeric solutions are fabricated from polytetrafluoroetheylene (PTFE), which is resistant to organic solvents.
  • the organic solvent is phase-separated from the polymer using an aqueous solution.
  • the completed structural support is removed from the mold after the polymeric solution solidifies.
  • the structural support may be fabricated from any biocompatible, substantially non-degradable polymer.
  • a "biocompatible" device causes substantially no adverse consequences to the host upon transplantation and thereafter.
  • biocompatible polymers suitable for use in the invention include acrylic, polyester, polyethylene, polypropylene, polyacetonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, polybutester, silk, cotton, chitin, carbon, as well as other biocompatible metals.
  • the polymer support is cast from a solution of 20 % weight to volume (w:v) PA /PVC in dimethyl sulfoxide (DMSO).
  • the present invention contemplates the use of several types cells to be inoculated on the matrices.
  • Examples of cells suitable for use in the present invention include C 2 C I2 mouse myoblast cells , LNS-1 cells, and ⁇ tet cells.
  • Also useful are human Hs683 glial-derived cells, human A172 glial-derived cells, porcine glioblasts, chondroblasts isolated from human long bone, mice 3T3 fibroblast cells, rabbit SIRC corneal-derived cells, Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK) cells, and CAC cells.
  • Cells are also useful when endogenously producing a protein of desired interest, such as when transfected to express the protein of interest.
  • Devices have been fabricated with C 2 C 12 myoblasts transfected with the murine EPO (mEPO) gene. Additionally, devices have been fabricated with C 2 C 12 cells transfected to express GLP-1.
  • mEPO murine EPO
  • myoblasts in culture grow in the form of clusters. These clusters have the capacity to fuse into larger clusters, referred to in the literature as "myoballs".
  • Myoblasts cultured in the present invention may be monitored, characterized, and controlled for the desired morphology prior to encapsulation. For instance, the myoblasts may be cultured in the present invention until all, or substantially all, of the myoblasts exist in the form of myoballs. Once the myoblasts have become myoballs, they no longer proliferate.
  • the number of cells that each device is able to contain is dictated by many factors, including the structural parameters of the device type (i.e. geometry and size).
  • the maximum number of cells per device is also a function of both the volume of the hydrogel and the cell density per unit volume of gel.
  • a ring-shaped device having a 9.7 mm thickness and an inner diameter of 12 mm has a volume of 80 ⁇ l.
  • the device therefore holds a maximum of approximately 10,000,000 cells.
  • the total length of PET fibers determines the maximum cell number for the PET fiber devices.
  • the corresponding length of PET fibers with a diameter of approximately 5 ⁇ m, which can be wound around the device is approximately 16 cm. Given that each 1 cm of fibers supports approximately 500,000 cells, such a device holds approximately 8,000,000 cells.
  • FIG. 2 shows two preferred encapsulation processes of the present invention.
  • a suspension of cells in agarose is polymerized in a PAN/PVC structural support.
  • the cells are cultured until they exhibit the desired morphology and are then dipped in a polymer solution and subsequently dipped in an aqueous solution, preferably a physiological buffer, which phase inverts the polymer solution and forms a semi-permeable membrane around the cells.
  • the finished devices are then placed in Petri dishes for in vitro storage.
  • FIG. 1 left side of the FIG. (A)
  • a suspension of cells in agarose is polymerized in a PAN/PVC structural support.
  • the cells are cultured until they exhibit the desired morphology and are then dipped in a polymer solution and subsequently dipped in an aqueous solution, preferably a physiological buffer, which phase inverts the polymer solution and forms a semi-permeable membrane around the cells.
  • the finished devices are then placed in Petri dishes
  • cell-supporting fibers preferably PET fibers
  • a cell solution is inoculated on the fibers and the cells are cultured until the exhibit the selected morphology.
  • the support containing the cells is then dipped in a polymer solution and then dipped in an aqueous solution, preferably a physiological buffer , which phase inverts the polymer solution and forms a semi-permeable membrane around the cells.
  • the finished devices are then placed in Petri dishes for in vitro storage.
  • membranes can be characterized by diffusion properties and by scanning electron microscopy (SEM).
  • SEM scanning electron microscopy
  • various molecular weight polydextrans are fluorescence labeled, incorporated into agarose, and encapsulated.
  • the membrane capsules are then placed into fluorophore-free water.
  • the -amount of fluorophore that has diffused into the water is measured periodically, over a 6-hr period. Fluorescence in aliquots of the water is detected with a fluorospectrometer.
  • SEM techniques common in the art. Individual membranes are glycerinated, dried, and gold-plated. The membranes are then introduced into the vacuum chamber of an electron microscope, so that their surface morphology can be viewed.
  • Example 1 Encapsulated mEPO Transfected Mouse C 2 C U Cells in an Agarose Matrix
  • the matrix formed as a semi-solid gel in a ring-shaped support.
  • the volume of the ring support totaled 80 ⁇ l.
  • the cell aggregates were formed under rotation at 60 rpm for three days in surface treated non-adherent Petri dishes in regular growth medium (DMEM, 10 % fetal calf serum [FCS], penicillin streptomycin, and MTX).
  • the cell containing matrix and the support were encapsulated in a 100 ⁇ m thick membrane by dip-coating in an 8 % (w:v) PAN/PVC solution in DMSO.
  • Group 1 was cultured in regular growth medium;
  • Group 2 was cultured in Prolifix, a culture medium containing vegetable proteins instead of animal serum proteins, and Group 3 was cultured in medium containing 2 % FCS instead of 10% FCS.
  • Cell viability was assayed by histology.
  • Cell functionality was assayed by measuring mEPO output in the media over time. The release of mEPO was determined by techniques common in the art, such as ELISA. EPO release was compared to EPO production under standard culture conditions. In culture, 1,000,000 mEPO transfected C 2 C 12 cells release approximately 50 IU mEPO in 24 hr.
  • FIG. 3 demonstrates that only single cell suspensions cultured in regular growth medium showed a significant increase in output of mEPO over time, evidence of continuous proliferation of cells within the device.
  • the remaining devices produced a relatively constant output of mEPO over time.
  • the overall output in these devices was relatively low. This finding was explained by poor viability of the cells in the form of cell clusters and the even worse viability of the single cells. Histological analysis revealed evenly distributed dead cells and dead cell clusters within each of the respective devices, suggesting that the dimensions of the device have no effect on viability.
  • devices cultured in Prolifix displayed the worst survival compared to devices cultured in 2 % FCS.
  • Two groups of devices were separately fabricated in agarose and encapsulated as described in the above examples.
  • One group contained ⁇ TC tet cells.
  • the second group contained INS-1 cell clusters. After encapsulation, the devices were maintained in culture. At various time points, the devices were harvested and examined by histology and by light microscope. The histological and light microscopy analysis revealed that the INS-1 cell clusters showed good viability and proliferation. The ⁇ TC tet cells, however, did not survive the cell culture period post encapsulation.
  • PET devices were inoculated with approximately 300,000 C 2 C 12 cells transfected with the mEPO gene and were cultured for four days in regular growth medium. After day four, the devices were split into three groups. The first group was encapsulated and put directly into medium containing 2% FCS. The second group was encapsulated and put back into regular growth medium for three days. After three days, the medium was changed to low serum growth medium to compensate for cell loss during the stress of encapsulation. The third group was not encapsulated and kept in 10% FCS growth medium. Samples were taken regularly from the culture medium and the amount of mEPO in the medium was measured by ELISA.
  • FIG. 4 shows the amount of mEPO released over time for the above-described three groups. These data establish that devices cultured in low serum containing media produced much less protein than devices cultured in regular growth medium. Moreover, as demonstrated by comparing TABLE 2 with FIG. 4, the amount of mEPO produced corresponds with the determined cell number at the end of the experiment. These data lead to the conclusion that culturing devices in low serum containing media significantly slowed the rate of proliferation in the cells. Additionally, the amount of mEPO in the media of non-encapsulated cells cultured in growth medium showed a plateau between days 8 and 10. This suggests that the maximum capacity of cells was reached for the given device size, geometry and type.
  • FIG. 5 compares cell number and amount of mEPO in the cell media for both pre- and post-encapsulation devices. Additionally, all unencapsulated devices were monitored by microscopy. At day -three post-encapsulation (day 10 overall) none of the devices showed any proliferation. This result is consistent with data shown in FIG. 5. The graph of proliferation as measured by mEPO release and cell number plateaus for devices of this size around day eight. This result was further confirmed by histological analysis. Prior to sacrificing devices for DNA analysis, unencapsulated devices were analyzed by light microscope at a 32x magnification. At this magnification, it was observed that most visible cells displayed myoball morphology.
  • murine C 2 C 12 cells transfected with the GP-1 gene were inoculated onto a PET fiber device at an initial loading density of 300,000 cells per device. After 3 days, the number of cells increased to 4,700,000 cells per device +/- 500,000 cells. After 7 days, the number of cells increased to 5,800,000 cells per device +/- 400,000 cells. Examination of the cells by light microscope revealed a very homogeneous, rapid colonization of fibers.
  • the apparent maximum number of cells that can be maintained within this given device configuration is between 5-6,000,000 cells, somewhat lower than the theoretical value of 8,000,000 cells. This finding emphasizes that in order to achieve the high cell densities described in TABLE 1, the cells must be loaded under compressive forces. One may obtain better seeding and survival of the cells within the devices by allowing the cells to attain a natural density upon and within the matrix by cell-matrix interaction, rather than by profusion,
  • Membranes were fabricated as described above and membrane mass transport properties were determined by measuring the diffusive flux of fluorescenfly labeled dextran molecules of different molecular weights. Mass transfer of the membrane was studied by immobilizing a membrane between two chambers of equal volume. One chamber was filled with the fluorescently labeled dextran solution and the other chamber was filled with the same volume of fluorophore-free solution. Over time, fluorescently labeled dextran molecules that are small enough to pass through the membrane will diffuse into the fluorophore-free chamber, changing the fluorescence of the recipient chamber. This change in fluorescence was measured by analyzing samples of the solution in a fluorimeter (Perkin Elmer). In addition to studying the diffusion properties, membranes were also characterized by scanning electron microscopy (SEM).
  • membrane thickness varied between about 100-300 ⁇ m. This variation was caused by the fabrication process in which the device is dip-coated to form the membrane. As the device is removed from the polymer solution, gravity causes the polymer solution to collect disproportionately on the bottom of the device. When the device is then phase inverted in the precipitation bath, the membrane forms with the thicker portion on the bottom and the thinner portion on the top.
  • TABLE 3 summarizes the diffusive properties and the physical properties of the membrane as determined by SEM. The diffusive properties of the dip-coated membrane fabricated with 8% PAN/PVC + 20% PVP (10 6 Da) are also illustrated in FIG. 6.
  • PAN/PVC 9° - 8 50 100-300 400 thick
  • Group 1 devices containing mouse C 2 C 12 cells transfected with GLP-1 were implanted syngenically into mice.
  • Group 2 devices containing mouse C 2 C, 2 cells transfected with GLP-1 were implanted allogenically into mice.
  • Group 3 devices containing mouse C 2 C 12 transfected with rat EPO were xenogenically transplanted into rats.
  • three devices were implanted subcutuaneously (SC) and three devices were transplanted intraperitoneally (IP).
  • rat EPO Before and after explant, the release of rat EPO was determined for xenogenically implanted devices. Devices which were syngenically and allogenically implanted into mice were evaluated for GLP-1 expression, both prior to implant and post explant. All devices were examined histologically for cell survival and biocompatibility. Additionally, the hematocrit of all EPO-device-implanted rats was monitored by a standard hematocrit method (Koepke, 1991).
  • the EPO output of devices pre-implant was quantified and is summarized in TABLE 6. As shown in the TABLE, the amount of EPO released from the capsules prior to implantation is relatively uniform. Additionally, the EPO of one capsule post explanation was determined to be undetectable.
  • FIG. 7 and FIG. 8 illustrate the hematocrit of individual animals over time for the subcutaneous and intraperitoneal implants, respectively.
  • the hematocrit rises dramatically between day 0 and day 10. Thereafter, the hematocrit level plateaus until approximately day fifteen. After approximately day fifteen, the hematocrit level for four of the six animals begins to decline. Two of 6 animals continued to show increased hematocrit for significantly longer as shown in the figures. Mouse Data.
  • FIG. 9 shows the viability scores of the syngenic and allogenic capsules.

Abstract

The present invention provides a cell implantation device comprised of a cell matrix system which is fixedly coupled to a flat sheet supporting structure, preferably ring-shaped. Cells are inoculated onto the matrix, which may be a cell supporting fiber or a hydrogel. The proliferation, viability, and functionality of the cells are characterized prior to encapsulation in an immunoisolatory semi-permeable membrane. Cell growth under controlled conditions allows the encapsulated device to provide a relatively constant dose of secreted protein.

Description

FLAT SHEET CELL ENCAPSULATIONDEVICE
RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application 60/085,648, filed May 15, 1998.
FIELD OF THE INVENTION
The present invention relates generally to cell encapsulation devices, and more particularly to devices for cell screening and implantation into a mammalian host.
BACKGROUND OF THE INVENTION
In encapsulated cell therapy, xenogenic or allogenic cells are isolated from the host's immune system by surrounding the cells in a semi-permeable membrane prior to implantation within the host. This semi -permeable membrane jacket is relatively impermeable to large molecules, such as components of the host's immune system, but is permeable to small molecules. In one embodiment, the semi-permeable membrane jacket has a molecular weight cutoff of between 50 and 2000 kilodaltons (kDa). Thus, the semi-permeable membrane allows the implanted cells to receive nutrients necessary for viability and allows metabolic waste to be removed. The membrane also allows therapeutic molecules produced by the implanted cells to diffuse to host cells. For example, endogenous proteins or those cloned into the cell are delivered to the host. The use of an immuno-protective, semi-permeable membrane now allows transplantation of encapsulated cells from one species into a host from a different species without the risk of immune rejection or use of immunosuppressive drugs. Applications of encapsulated cell therapy include, for example, treatments for diabetes, hemophilia, anemia, β-thalassemia, Parkinson's disease, and amyotropic lateral sclerosis.
The use of biologically compatible polymeric materials in construction of an encapsulation device is critical to a successful cell encapsulation therapy. Important components of the encapsulation device include the surrounding semi-permeable membrane and the internal cell-supporting matrix, or scaffold. The scaffold defines the microenvironment for the encapsulated cells and keeps the cells well distributed within the intracapsular compartment. The optimal internal scaffold for a particular cell encapsulation device is highly dependent on the cell type. For example, while adherent cells often prefer a solid surface on which to lie, suspension cells may prefer a hydrophilic lightly cross-linked hydrogel as a matrix material.
In the absence of a scaffold, adherent cells aggregate to form clusters. When the clusters grow too large, they typically develop a central necrotic core. Dying cells accumulate around the core and, upon lysing, release factors detrimental to the health of neighboring cells. The lysed cell fragments are also transported to the host environment, there eliciting an antigenic response.
The prior art discloses that the position of the cells may be stabilized by an internal scaffold composed of an immobilizing matrix formed from a hydrogel. A "hydrogel" is a 3 -dimensional network of cross-linked hydrophilic polymers, in the form of a gel and substantially (e.g., greater than 90%) composed of water. In typical prior art systems, however, cells are encapsulated within an immunoisolatory membrane and then allowed to continue to proliferate. Once the cells are placed inside the encapsulating membrane, it is extremely difficult to characterize their morphology. Moreover, it is nearly impossible to control cell proliferation and functionality. As a result, devices made using methods of the prior art exhibit wide ranges of total expression of the desirable secreted protein. Consequently, it is difficult to control the therapeutic dosage received by the host.
Thus a need remains to develop an encapsulation device in which the cells to be encapsulated are selected for a desired morphology prior to encapsulation, resulting in better control of expression of the secreted protein after encapsulation.
SUMMARY OF THE INVENTION
The invention provides a cell implantation device. The device has a cell matrix, or scaffold, affixed to a supporting frame. The supporting frame is preferably a flat, ring-shaped element. Cells are inoculated onto the matrix. Once the cells exhibit the desired morphology, the cell-containing matrix is encapsulated in a semi-permeable immunoisolatory membrane. The cell matrix, or scaffold, in the device of the invention advantageously provides cells with a template for cellular organization in a three-dimensional orientation resembling their typical physiological shape. The cell matrix is particularly useful for the use of suspension cells for encapsulated cell therapy. In one embodiment of the present invention, the matrix is composed of a hydrogel material selected from agarose, alginate, chitosan, collagen, Matrigel®, Vitrogen®, or PVA foam. In a preferred embodiment, the hydrogel is agarose. In this embodiment, the selected cells are mixed with a hydrogel solution at a desired cell density and a desired concentration of gel polymer. After mixing, the cell/hydrogel suspension is cast into the structural support which serves as a mold while the suspension solidifies into the cell-supporting matrix. Once the matrix solidifies, mechanical forces, surface tension, and capillary forces hold the matrix in the structural support. Upon curing, the hydrogel solution becomes gelled and acts as a supporting matrix for the cells. The matrix/support can then be encapsulated in the semi-permeable membrane.
In another embodiment, the cell matrix is composed of fibers selected from the group consisting of silk, cotton, chitin, for example, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, or polybutester. In a preferred embodiment, the matrix is polyethyleneterephthalate (PET) fiber. The fibrous material can be coated with a protective coating of hydrogel prior to encapsulation in the membrane. In this embodiment, the matrix is a cell-supporting fiber wound or woven around the structural support. PET fibers can be wound around a ring shaped structural support. Alternatively, the structural support is a polyvinyl alcohol sponge incorporated into the structural support. A desired cell type is then inoculated onto the selected solid matrix and cultured in the appropriate medium.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows three top views of components of the device. FIG. 1 A is a representation of two structural support components. FIG. IB is a representation of two structural support components, each fabricated with a cell/agarose component. FIG. 1C is a representation of two structural support components fabricated with PET fibers.
FIG. 2 is a schematic representation of two preferred device fabrication processes.
FIG. 3 is a graph showing mEPO output over time for devices with cell captured in an agarose matrix and cultured in various media. FIG. 4 is a graph showing mEPO output over time for devices with cell captured in a PET fiber matrix and cultured in various media.
FIG. 5 is a graph showing mEPO output over time for pre- and post-transplant devices cultured in various media. FIG. 6 is a graph showing the diffusive properties of a dip-coated membrane fabricated with 8 % PAN/PVC + 20 % PVP (106 daltons [Da]).
FIG. 7 is a graph showing hematocrit levels over time in rats which were implanted with the device subcutaneously.
FIG. 8 is a graph showing hematocrit levels over time in rats which were implanted with the device interperitoneally.
FIG. 9 shows the viability scores of devices which were implanted syngenically and allogenically.
DETAILED DESCRIPTION OF THE INVENTION
The invention provides a cell encapsulation device configured as a flat sheet. The device has (1) a structural support or supporting frame element (e.g., in the shape of a ring); (2) matrices to support the cells to be transplanted; and (3) a semi-permeable membrane that permits diffusion of nutrients, waste, and other small molecules to and from the transplanted cells, while isolating those cells from the host.
The details of one or more embodiments of the invention are set forth in the accompanying description below. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated by reference. Matrices
The invention contemplates the use of various matrices, or matrix systems, to support the cells to be encapsulated. The selection of a matrix may depend on the cell type used, with a particular cell type preferentially adhering and growing onto one matrix over another. In one embodiment, the matrix is a hydrogel. A "hydrogel" is a substance formed when a polymer is cross linked to create a 3-dimensional structure, entrapping water molecules to form a gel.
In another embodiment solid matrices, which may be more appropriate for certain types of cells, are used. This embodiment is shown in FIG 1. FIG. 1A shows two examples of a structural support, each having a tab for handling the device. FIG. IB and FIG. 1C show the structural support fixedly coupled to the cell matrix system. In FIG. 1C, the cell matrix system is a cell/agarose mixture. In FIG. 1C, the cell matrix system is the fibrous material, preferably PET fibers, which is wound or otherwise fixedly coupled to the structural support.
To inoculate and culture the cells, the support can be dipped into a cell/agarose suspension and allowed to attain the desired conformation (see, FIG. 2, left side of the FIG. (A)). Alternatively, a support containing the cell supporting fiber can be placed in a Petri dish on a non-adherent surface suitable to culture cells, such as, for example, non-adherent agarose The cells to be implanted are then inoculated onto the fiber (see, FIG. 2, right side of the FIG. (B)). The cells are then cultured until they attach to the fiber. An extracellular matrix may be used to coat the fiber to aid cell attachment. Examples of fibers suitable for use in the present invention include natural fibers such cotton, silk, chitin or carbon. Non-naturally occurring fibers, such as, for example, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, or polybutester are also suitable for use in the present invention. Preferably, the fiber is PET, which is typically comprised of bundles of approximately 80 PET monofilaments. The diameter of the monofilaments selected can range between 1-30 μm. Monofilaments having a range of diameters between approximately 1-10 μm are preferred, with a diameter of 5 μm being most preferred. The type of fiber selected and the dimensions of the fiber may depend on cell type as one cell type may prefer one type of fiber over another. Additionally, the fibrous matrix selected may be coated with a protective layer of hydrogel prior to encapsulation in the membrane. Structural Support
The structural component to support the cell matrix system can be formed in many different dimensions and geometric shapes. For example, the supports may be circular, elliptical, square, rectangular, triangular, or star shaped. Since devices are fabricated as flat sheets, the thickness of the ring is much less than the diameter of the ring. TABLE 1 lists the dimensions of devices that have been fabricated.
TABLE 1
Ring Inner Outer Volume Cell Number Corresponding
Thickness Diameter Diameter Hollow Fiber
[μl] (in millions)
[mm] [mm] [mm] Length [cm]
0.4 12 14 45 6 12
0.7 12 14 80 10 20
0.4 20 22 125 15 30
0.7 20 22 219 27 54
(PMMA PET) 10 13 31 8 16 0.4
(PMMA/PET) 20 23 125 32 64 0.4
The structural support can be made by methods well known to those skilled in the art. The structural support can be made, for example, from a polymeric solution cast into a mold or by stamping, cutting, and extruding. Typically, molds for casting polymeric solutions are fabricated from polytetrafluoroetheylene (PTFE), which is resistant to organic solvents.
Alternatively, other organic solvent-resistant materials can be used. After the polymeric solution is cast into the mold, the organic solvent is phase-separated from the polymer using an aqueous solution. The completed structural support is removed from the mold after the polymeric solution solidifies. The structural support may be fabricated from any biocompatible, substantially non-degradable polymer. A "biocompatible" device causes substantially no adverse consequences to the host upon transplantation and thereafter. Examples of biocompatible polymers suitable for use in the invention include acrylic, polyester, polyethylene, polypropylene, polyacetonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, polybutester, silk, cotton, chitin, carbon, as well as other biocompatible metals. Preferably, the polymer support is cast from a solution of 20 % weight to volume (w:v) PA /PVC in dimethyl sulfoxide (DMSO).
Cells
The present invention contemplates the use of several types cells to be inoculated on the matrices. Examples of cells suitable for use in the present invention include C2CI2 mouse myoblast cells , LNS-1 cells, and β tet cells. Also useful are human Hs683 glial-derived cells, human A172 glial-derived cells, porcine glioblasts, chondroblasts isolated from human long bone, mice 3T3 fibroblast cells, rabbit SIRC corneal-derived cells, Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK) cells, and CAC cells. Cells are also useful when endogenously producing a protein of desired interest, such as when transfected to express the protein of interest. Devices have been fabricated with C2C12 myoblasts transfected with the murine EPO (mEPO) gene. Additionally, devices have been fabricated with C2C12 cells transfected to express GLP-1.
In culture, cellular behavior is often related to cell morphology (i.e. differentiation, growth arrested). Accordingly, cells can be cultured under selected conditions until the cells exhibit a particular morphology corresponding with the desired phenotype. For example, myoblasts in culture grow in the form of clusters. These clusters have the capacity to fuse into larger clusters, referred to in the literature as "myoballs". Myoblasts cultured in the present invention may be monitored, characterized, and controlled for the desired morphology prior to encapsulation. For instance, the myoblasts may be cultured in the present invention until all, or substantially all, of the myoblasts exist in the form of myoballs. Once the myoblasts have become myoballs, they no longer proliferate. Once the cells stop proliferating, the number of cells, and therefore the amount of protein they produce, becomes relatively constant. In this manner the dose of protein delivered to the patient can be regulated. The number of cells that each device is able to contain is dictated by many factors, including the structural parameters of the device type (i.e. geometry and size). For devices in which a hydrogel is used, the maximum number of cells per device is also a function of both the volume of the hydrogel and the cell density per unit volume of gel. For example, a ring-shaped device having a 9.7 mm thickness and an inner diameter of 12 mm has a volume of 80 μl. Assuming that in order to maintain sufficient structural integrity, 50% of the volume is occupied by the hydrogel, the remaining volume available to be occupied by the cells is 40 μl. Given that 4 μl holds approximately 1 ,000,000 cells, the device therefore holds a maximum of approximately 10,000,000 cells.
In addition to device parameters, the total length of PET fibers determines the maximum cell number for the PET fiber devices. For example, in a ring-shaped PET fiber device having a ring thickness of 0.4 mm, an inner diameter of 12 mm and an outer diameter of 13 mm, the corresponding length of PET fibers, with a diameter of approximately 5 μm, which can be wound around the device is approximately 16 cm. Given that each 1 cm of fibers supports approximately 500,000 cells, such a device holds approximately 8,000,000 cells.
Encapsulation
Once the cultured cells have reached their desired morphology on the cell matrix system/structural support, the cells on the matrix/structural support can then be encapsulated. Encapsulation involves a 2-step process. FIG. 2 shows two preferred encapsulation processes of the present invention. In FIG. 2, left side of the FIG. (A), a suspension of cells in agarose is polymerized in a PAN/PVC structural support. The cells are cultured until they exhibit the desired morphology and are then dipped in a polymer solution and subsequently dipped in an aqueous solution, preferably a physiological buffer, which phase inverts the polymer solution and forms a semi-permeable membrane around the cells. The finished devices are then placed in Petri dishes for in vitro storage. In FIG. 2, right side of the FIG. (B), cell-supporting fibers, preferably PET fibers, are fixedly coupled to a PAN/PVC structural support. A cell solution is inoculated on the fibers and the cells are cultured until the exhibit the selected morphology. The support containing the cells is then dipped in a polymer solution and then dipped in an aqueous solution, preferably a physiological buffer , which phase inverts the polymer solution and forms a semi-permeable membrane around the cells. The finished devices are then placed in Petri dishes for in vitro storage.
In both encapsulation procedures, percentages of the polymer solutions may be varied to produce membranes with different thickness. Membranes can be characterized by diffusion properties and by scanning electron microscopy (SEM). To measure diffusion properties, various molecular weight polydextrans are fluorescence labeled, incorporated into agarose, and encapsulated. The membrane capsules are then placed into fluorophore-free water. The -amount of fluorophore that has diffused into the water is measured periodically, over a 6-hr period. Fluorescence in aliquots of the water is detected with a fluorospectrometer. To study the morphological aspects of membranes made from the various polymeric solutions, the membranes are viewed using SEM techniques common in the art. Individual membranes are glycerinated, dried, and gold-plated. The membranes are then introduced into the vacuum chamber of an electron microscope, so that their surface morphology can be viewed.
The following examples are presented in order to more fully illustrate the preferred embodiments of the invention. These examples should in no way be construed as limiting the scope of the invention, as defined by the appended claims.
Example 1 Encapsulated mEPO Transfected Mouse C2CU Cells in an Agarose Matrix
Approximately 1,000,000, murine C2C12 cells transfected with the murine EPO gene, either as single cells or cell aggregates, were trapped within a 1% low-melting agarose matrix. The matrix formed as a semi-solid gel in a ring-shaped support. The volume of the ring support totaled 80 μl. The cell aggregates were formed under rotation at 60 rpm for three days in surface treated non-adherent Petri dishes in regular growth medium (DMEM, 10 % fetal calf serum [FCS], penicillin streptomycin, and MTX). The cell containing matrix and the support were encapsulated in a 100 μm thick membrane by dip-coating in an 8 % (w:v) PAN/PVC solution in DMSO.
To evaluate the possibility of using low serum media to slow or halt proliferation, three groups of devices containing either single cells or cell clusters were grown in different medium conditions. Group 1 was cultured in regular growth medium; Group 2 was cultured in Prolifix, a culture medium containing vegetable proteins instead of animal serum proteins, and Group 3 was cultured in medium containing 2 % FCS instead of 10% FCS. Cell viability was assayed by histology. Cell functionality was assayed by measuring mEPO output in the media over time. The release of mEPO was determined by techniques common in the art, such as ELISA. EPO release was compared to EPO production under standard culture conditions. In culture, 1,000,000 mEPO transfected C2C12 cells release approximately 50 IU mEPO in 24 hr.
FIG. 3 demonstrates that only single cell suspensions cultured in regular growth medium showed a significant increase in output of mEPO over time, evidence of continuous proliferation of cells within the device. The remaining devices produced a relatively constant output of mEPO over time. However, the overall output in these devices was relatively low. This finding was explained by poor viability of the cells in the form of cell clusters and the even worse viability of the single cells. Histological analysis revealed evenly distributed dead cells and dead cell clusters within each of the respective devices, suggesting that the dimensions of the device have no effect on viability. Finally, devices cultured in Prolifix displayed the worst survival compared to devices cultured in 2 % FCS.
Example 2 β TC tet Cells and INS-1 Cells in Agarose
Two groups of devices were separately fabricated in agarose and encapsulated as described in the above examples. One group contained β TC tet cells. The second group contained INS-1 cell clusters. After encapsulation, the devices were maintained in culture. At various time points, the devices were harvested and examined by histology and by light microscope. The histological and light microscopy analysis revealed that the INS-1 cell clusters showed good viability and proliferation. The β TC tet cells, however, did not survive the cell culture period post encapsulation.
Example 3 Encapsulated mEPO Transfected C2C12 Cells on PET Matrix
An analysis was performed to determine whether culturing devices in low serum containing medium slowed mitosis or altogether arrested proliferation. PET devices were inoculated with approximately 300,000 C2C12 cells transfected with the mEPO gene and were cultured for four days in regular growth medium. After day four, the devices were split into three groups. The first group was encapsulated and put directly into medium containing 2% FCS. The second group was encapsulated and put back into regular growth medium for three days. After three days, the medium was changed to low serum growth medium to compensate for cell loss during the stress of encapsulation. The third group was not encapsulated and kept in 10% FCS growth medium. Samples were taken regularly from the culture medium and the amount of mEPO in the medium was measured by ELISA. The quantity of DNA present was measured to determine the number of cells. The devices were also evaluated by histology. FIG. 4 shows the amount of mEPO released over time for the above-described three groups. These data establish that devices cultured in low serum containing media produced much less protein than devices cultured in regular growth medium. Moreover, as demonstrated by comparing TABLE 2 with FIG. 4, the amount of mEPO produced corresponds with the determined cell number at the end of the experiment. These data lead to the conclusion that culturing devices in low serum containing media significantly slowed the rate of proliferation in the cells. Additionally, the amount of mEPO in the media of non-encapsulated cells cultured in growth medium showed a plateau between days 8 and 10. This suggests that the maximum capacity of cells was reached for the given device size, geometry and type.
TABLE 2
Number of cells cultured in: Days Growth medium 2% FCS post-enc. 2% FCS directly
0 300,000 300,000 300,000
13 6.51 x lO6 3.2 x lO6 3.0 x lO6
In a second study, 18 devices were inoculated with 300,000 C2C12 cells transfected with the mEPO gene. The cell-seeded devices were kept in regular growth medium for seven days before encapsulation. During the course of the experiment, the number of cells present in each device was monitored. On the day of encapsulation and on day four after encapsulation, both the amount of mEPO and the number of cells were quantified. At the day of encapsulation, the devices were divided into three groups. The first group was left unencapsulated and continued to be cultured in regular growth media. The second group of devices was encapsulated and immediately put into culture medium containing 2% FCS. The third group was left unencapsulated and was also transferred into medium containing 2% FCS.
FIG. 5 compares cell number and amount of mEPO in the cell media for both pre- and post-encapsulation devices. Additionally, all unencapsulated devices were monitored by microscopy. At day -three post-encapsulation (day 10 overall) none of the devices showed any proliferation. This result is consistent with data shown in FIG. 5. The graph of proliferation as measured by mEPO release and cell number plateaus for devices of this size around day eight. This result was further confirmed by histological analysis. Prior to sacrificing devices for DNA analysis, unencapsulated devices were analyzed by light microscope at a 32x magnification. At this magnification, it was observed that most visible cells displayed myoball morphology. Because myoblasts stop proliferating when they are formed in the myoball morphology, this result is consistent the plateau of cell growth in FIG. 6. It should be noted, however, that some cells in the device were not visible because they had colonized the interior of the PET bundles.
Example 4 Rate of Colonization of Device with C2C12 Cells Transfected with GLP-1
To determine whether the rate of colonization of C2C12 cells vary with different clones, murine C2C12 cells transfected with the GP-1 gene were inoculated onto a PET fiber device at an initial loading density of 300,000 cells per device. After 3 days, the number of cells increased to 4,700,000 cells per device +/- 500,000 cells. After 7 days, the number of cells increased to 5,800,000 cells per device +/- 400,000 cells. Examination of the cells by light microscope revealed a very homogeneous, rapid colonization of fibers.
The apparent maximum number of cells that can be maintained within this given device configuration is between 5-6,000,000 cells, somewhat lower than the theoretical value of 8,000,000 cells. This finding emphasizes that in order to achieve the high cell densities described in TABLE 1, the cells must be loaded under compressive forces. One may obtain better seeding and survival of the cells within the devices by allowing the cells to attain a natural density upon and within the matrix by cell-matrix interaction, rather than by profusion,
Example 5 Membrane Characterization
Membranes were fabricated as described above and membrane mass transport properties were determined by measuring the diffusive flux of fluorescenfly labeled dextran molecules of different molecular weights. Mass transfer of the membrane was studied by immobilizing a membrane between two chambers of equal volume. One chamber was filled with the fluorescently labeled dextran solution and the other chamber was filled with the same volume of fluorophore-free solution. Over time, fluorescently labeled dextran molecules that are small enough to pass through the membrane will diffuse into the fluorophore-free chamber, changing the fluorescence of the recipient chamber. This change in fluorescence was measured by analyzing samples of the solution in a fluorimeter (Perkin Elmer). In addition to studying the diffusion properties, membranes were also characterized by scanning electron microscopy (SEM).
By SEM analysis, membrane thickness varied between about 100-300 μm. This variation was caused by the fabrication process in which the device is dip-coated to form the membrane. As the device is removed from the polymer solution, gravity causes the polymer solution to collect disproportionately on the bottom of the device. When the device is then phase inverted in the precipitation bath, the membrane forms with the thicker portion on the bottom and the thinner portion on the top. TABLE 3 summarizes the diffusive properties and the physical properties of the membrane as determined by SEM. The diffusive properties of the dip-coated membrane fabricated with 8% PAN/PVC + 20% PVP (106 Da) are also illustrated in FIG. 6.
TABLE 3
Composition of Polymer MW Cut-off Diffusion Coefficient Surface Membrane
Solution [w/v] [daltons (Da)] [cmVsec] Smoothness Thickness [μm]
8% PAN/PVC 12.00 1 0 + 80-10
6% PAN/PVC +30% w/w ++ 100-200 PVP 106Da
6% PAN/PVC +50% w/w +++ 200-300 PVP 106 Da
Physical properties of the membrane and a control PES-5 membrane are summarized in TABLE 4. The molecular weight cut-off for the 8% PAN/PVC + 20% PVP flat sheet membrane was determined to be approximately 50,000 Da. This value is significantly lower than the molecular weight cut-off determined for the PES-5 membrane, which was approximately 240,000 Da. In terms of overall transport properties, however, the absolute values for the diffusion coefficients of the individual dextran molecules (TABLE 4, Column 2) do not differ significantly. In order to provide sufficient immunoisolation from the host, the diffusion coefficients may need to be at least two orders of magnitude lower.
TABLE 4
Membrane Type Diff. Coeff. Cut-off [kDa] Wall thickness [μm] ID [μm]
[cm2/s] T70
PES 5 0.5 - le - 7 240 100 500
PAN/PVC 9° - 8 50 100-300 400 thick
Example 6 In Vivo Data in Mice and Rats
In vivo functionality and biocompatibility of the devices were studied in mice and rats. Eighteen devices were prepared and divided into three groups of six as summarized in TABLE 5. For each device, 40 cm of PET yarn was wound onto PMMA rings. The devices were inoculated with approximately 300,000 cells (day zero) and incubated for seven days in culture. After seven days, devices were first coated in a layer of agarose and then encapsulated by dip-coating into a polymer solution of 8% weight to volume of PAN/PVC + 20% weight to weight PVP (106 Da) followed by phase inversion in a physiological solution. Immediately after encapsulation, all implants were directly transferred into cell culture medium containing 2% serum.
Group 1 devices containing mouse C2C12 cells transfected with GLP-1 were implanted syngenically into mice. Group 2 devices containing mouse C2C,2 cells transfected with GLP-1 were implanted allogenically into mice. Lastly, Group 3 devices containing mouse C2C12 transfected with rat EPO were xenogenically transplanted into rats. In all three groups, three devices were implanted subcutuaneously (SC) and three devices were transplanted intraperitoneally (IP). TABLE 5 summarizes the experimental methodology.
TABLE 5
Group 1 Group 2 Group 3 mouse C2C12 #149 GLP1 mouse C2C12 #149 GLP1 mouse C2C12rat EPO (pool) n=6 n=6 n=6 syngenic allogenic xenogenic (rat)
SC: n=3 SC: n=3 SC: n=3 IP: n=3 IP: n=3 IP: n=3
Before and after explant, the release of rat EPO was determined for xenogenically implanted devices. Devices which were syngenically and allogenically implanted into mice were evaluated for GLP-1 expression, both prior to implant and post explant. All devices were examined histologically for cell survival and biocompatibility. Additionally, the hematocrit of all EPO-device-implanted rats was monitored by a standard hematocrit method (Koepke, 1991).
The EPO output of devices pre-implant was quantified and is summarized in TABLE 6. As shown in the TABLE, the amount of EPO released from the capsules prior to implantation is relatively uniform. Additionally, the EPO of one capsule post explanation was determined to be undetectable.
TABLE 6
Capsule number pre-implant post-explant UI/24 hrs/caps UI/24 hrs/caps
1 SC 223.5 -
2 SC 230 0
3 SC 265 -
4 IP 310 -
5 IP 237.5 -
6 IP 260 -
Rat Data.
FIG. 7 and FIG. 8 illustrate the hematocrit of individual animals over time for the subcutaneous and intraperitoneal implants, respectively. As shown in FIG. 7 and FIG. 8, the hematocrit rises dramatically between day 0 and day 10. Thereafter, the hematocrit level plateaus until approximately day fifteen. After approximately day fifteen, the hematocrit level for four of the six animals begins to decline. Two of 6 animals continued to show increased hematocrit for significantly longer as shown in the figures. Mouse Data.
In order to obtain a two week time point of cell viability within the capsule, two devices were explanted from mice after two weeks in vivo. The remaining capsules were explanted after three weeks. All capsules were analyzed for GLP-1 output and by histology for cell survival. TABLE 7 illustrates the output GLP-1 from the capsules post-explantation.
TABLE 7
Capsule # syngenic allogenic GLP-1 [ng] gLP-1 [ng]
1 0 2 weeks: 229
2 7070 7650
3 0 7830
4 2 weeks: 217 8045
5 7485 7750
6 7885 7770
As TABLE 7 illustrates, there was no significant difference in GLP-1 output between the devices explanted at two weeks. With the exception of two capsules in which GLP-1 production was undetectable, the remaining capsules all produced the same relative amount of GLP-1 output. Note, however, that the values of GLP-1 on the three-week time point were very high compared to the two week time point. This difference was a result of an error in the cAMP assay for the three-week time point. Thus, only relative comparisons may be made.
Histology of the individual devices revealed excellent viability of cells inside capsules that had been implanted into an allogenic environment, with approximately 67% viability. Capsules that had been syngenically implanted fared less well, but still had a viability of approximately 41%. FIG. 9 shows the viability scores of the syngenic and allogenic capsules.
EQUIVALENTS
From the foregoing detailed description of the specific embodiments of the invention, it should be apparent that a novel flat sheet cell implantation device and method has been described. Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims which follow. In particular, it is contemplated by the inventor that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims.

Claims

What is claimed is:
1. A method of making a cell implantation device comprising the steps of:
(a) affixing a supporting frame element to a cell matrix system;
(b) inoculating cells onto the cell matrix system;
(c) culturing the cells until the cells exhibit a selected morphology having a corresponding selected phenotype; and
(d) encapsulating at least the cell matrix and cells within a semi-permeable immunoisolatory membrane.
2. The method of claim 1 , wherein the semi-permeable immunoisolatory membrane is formed from a polymer solution capable of forming a semi-permeable immunoisolatory membrane upon phase inversion in an aqueous medium; and
3. The method of claim 2, wherein said polymer solution is selected from the group consisting essentially of PAN, PVC, PVP, and combinations thereof.
4. The method of claim 1 wherein the cells are myoblasts, the selected morphology is cells in the form of myoballs, and the selected phenotype is the substantial halting of proliferation of the cells upon forming into myoballs.
5. The method of claim 1 wherein the cell matrix system is a network of fibers selected from the group consisting of silk, cotton, chitin, silk, cotton, chitin, carbon, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, polybutester, and polyethyleneterephthalate (PET).
7. A cell implantation device, comprising: a cell matrix system affixed to a supporting frame element, wherein the cell matrix system can be inoculated with cells having a selected moφhology in response to the cell matrix system, and wherein the frame element and cell matrix are encapsulated in a semi-permeable membrane jacket.
8. The device of claim 7, wherein the cells are myoblasts.
9. The device of claim 7, wherein semi-permeable membrane jacket is formed from a polymer solution selected from the group consisting essentially of PAN, PVC, PVP, and combinations thereof.
10. The device of claim 7, wherein the cell matrix system is a network of fibers selected from the group consisting of silk, cotton, chitin, silk, cotton, chitin, carbon, acrylic, polyester, polyethylene, polypropylene, polyacrylonitrile, polyethylene terephthalate, nylon, polyamides, polyurethanes, polybutester, and PET.
12. The device of claim 7, wherein the cell matrix and cells are coated with a hydrogel prior to encapsulation.
13. The device of claim 7, wherein the device is configured as a flat sheet.
14. The device of claim 13, wherein the device contains between 5 million and 500 million cells.
15. The device of claim 7, wherein the semi-permeable membrane jacket has a molecular weight cutoff of between 50 and 2000 kilodaltons (kDa).
16. A method for treating a subject, comprising: implanting into the subject a cell matrix system affixed to a supporting frame element, wherein the cell matrix system can be inoculated with cells having a selected moφhology in response to the cell matrix system, and wherein the frame element and cell matrix are encapsulated in a semi-permeable immunoisolatory membrane.
PCT/IB1999/000889 1998-05-15 1999-05-14 Flat sheet cell encapsulation device WO1999060105A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU36231/99A AU3623199A (en) 1998-05-15 1999-05-14 Flat sheet cell encapsulation device

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US8564898P 1998-05-15 1998-05-15
US60/085,648 1998-05-15

Publications (1)

Publication Number Publication Date
WO1999060105A1 true WO1999060105A1 (en) 1999-11-25

Family

ID=22193037

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1999/000889 WO1999060105A1 (en) 1998-05-15 1999-05-14 Flat sheet cell encapsulation device

Country Status (2)

Country Link
AU (1) AU3623199A (en)
WO (1) WO1999060105A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992019195A1 (en) * 1991-04-25 1992-11-12 Brown University Research Foundation Implantable biocompatible immunoisolatory vehicle for delivery of selected therapeutic products
WO1994007999A1 (en) * 1992-09-28 1994-04-14 Brown University Research Foundation Chitosan matrices for encapsulated cells
WO1997020569A2 (en) * 1995-12-07 1997-06-12 Encelle, Inc. Bioartificial devices and cellular matrices therefor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992019195A1 (en) * 1991-04-25 1992-11-12 Brown University Research Foundation Implantable biocompatible immunoisolatory vehicle for delivery of selected therapeutic products
WO1994007999A1 (en) * 1992-09-28 1994-04-14 Brown University Research Foundation Chitosan matrices for encapsulated cells
WO1997020569A2 (en) * 1995-12-07 1997-06-12 Encelle, Inc. Bioartificial devices and cellular matrices therefor

Also Published As

Publication number Publication date
AU3623199A (en) 1999-12-06

Similar Documents

Publication Publication Date Title
US6054142A (en) Biocompatible devices with foam scaffolds
US6303136B1 (en) Cells or tissue attached to a non-degradable filamentous matrix encapsulated by a semi-permeable membrane
AU698624B2 (en) Growth control for cells encapsulated within bioartificial organs
RU2177503C2 (en) Macrogranule with secretory cells, method of its preparing and method of treatment of patients with diseases caused by secretory cell function disorder
US5843431A (en) Controlling proliferation of cells before and after encapsulation in a bioartificial organ by gene transformation
EP3071248B1 (en) Tissue scaffold materials for tissue regeneration and methods of making
Mayer et al. Matrices for tissue engineering-scaffold structure for a bioartificial liver support system
US8778673B2 (en) Seeding cells on porous supports
JP4575152B2 (en) Porous gelatin material, gelatin structure, preparation method thereof and use thereof
US20230094662A1 (en) Method and apparatus for spatial control of cellular growth
Zondervan et al. Design of a polyurethane membrane for the encapsulation of islets of Langerhans
CN111467565B (en) Microtubule scaffold, preparation method and application thereof
US6495364B2 (en) Mx-1 conditionally immortalized cells
Sefton et al. Hydrophilic polyacrylates for the microencapsulation of fibroblasts or pancreatic islets
WO1999060105A1 (en) Flat sheet cell encapsulation device
Li et al. Encapsulation matrices for neurotrophic factor-secreting myoblast cells
CN111035809B (en) Double-layer composite nanofiber membrane with three-dimensional deformation structure and preparation method and application thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

NENP Non-entry into the national phase

Ref country code: KR

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase