US20030087427A1 - Method and apparatus for delivering oxygen to cells - Google Patents

Method and apparatus for delivering oxygen to cells Download PDF

Info

Publication number
US20030087427A1
US20030087427A1 US10/040,278 US4027801A US2003087427A1 US 20030087427 A1 US20030087427 A1 US 20030087427A1 US 4027801 A US4027801 A US 4027801A US 2003087427 A1 US2003087427 A1 US 2003087427A1
Authority
US
United States
Prior art keywords
cells
oxygen
oxygen generator
cell encapsulating
cell
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
US10/040,278
Inventor
Clark Colton
Larry Swette
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US10/040,278 priority Critical patent/US20030087427A1/en
Publication of US20030087427A1 publication Critical patent/US20030087427A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/022Artificial gland structures using bioreactors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/08Flask, bottle or test tube
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/12Well or multiwell plates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/24Gas permeable parts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M29/00Means for introduction, extraction or recirculation of materials, e.g. pumps
    • C12M29/04Filters; Permeable or porous membranes or plates, e.g. dialysis

Definitions

  • the oxygen generator is an electrolyzer device which electrolyzes water into oxygen and hydrogen.
  • the oxygen generator is in the form of a thin, multilayer electrolyzer sheet and is permeable to gas and water vapor but impermeable to liquids and dissolved material.
  • the oxygen generator comprises a multilayer electrolyzer sheet having a proton exchange membrane sandwiched by an anode layer and a cathode layer.
  • the device comprises a multilayer electolyzer sheet adapted for mating to a container containing cells.
  • the oxygen generator is provided as an in-line oxygenator through which blood, plasma, and other bodily fluids may flow.
  • the oxygen generator is mated to, contained within, or is an integral part of, a hollow tube through which blood, plasma, and other bodily fluids, or culture medium may flow.
  • the invention also relates to a system for delivering oxygen in situ to cells which are not contained within a cell encapsulating device.
  • the system for delivering oxygen in situ comprises an oxygen generator placed in proximity to cell-containing microcapsules which are free to migrate within an intraperitoneal space.
  • the system comprises an oxygen generator positioned in proximity to a cell-supporting, biocompatible, polymeric scaffold within the body of an organism. In this embodiment, the oxygen generator can be used to maintain, and support the growth of artificial tissues.
  • FIG. 1 is a schematic diagram of an oxygen generator according to one embodiment of the invention.
  • the oxygen generator 18 comprises two catalytic electrodes, i.e., an anode 21 and a cathode 23 , separated by a solid polymer electrolyte proton exchange membrane (PEM) 24 which allows passage of hydrated H + but has much lower permeability to gases such as O 2 and H 2 .
  • PEM solid polymer electrolyte proton exchange membrane
  • the system may be used to deliver cells 14 which can provide a function lacking in a body of a patient.
  • the function may be naturally provided by these cells 14 , or the cells 14 may be genetically modified to provide the function.
  • the system may be used in methods of treatment of conditions, especially chronic conditions, including, but not limited to, the treatment of diabetes, hemophilia, dwarfism, anemia, kidney failure, hepatic failure, immunodeficiency disorders, pituitary disorders, and central nervous system disorders.
  • Nervous system disorders which may be treated include, but are not limited to, chronic pain, Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis.
  • the space between the two semipermeable barrier layer 9 membranes contains the cells 14 to be implanted.
  • the semipermeable barrier layer 9 membranes are both distensible and conformable. Distensible semipermeable barrier layer 9 membranes allow for the expansion of the cell encapsulating chamber 10 , which is important when proliferating cell lines are used.
  • the conformable semipermeable barrier layer 9 membranes partially conform to the shape of the cells 14 , leading to a structure akin to peanut brittle.
  • the oxygen generator 18 can be provided proximal to, mated with, or an integral part of, a hollow tube through which blood flows.
  • cells 14 are implanted outside the tube.
  • the cells 14 are surrounded by a housing.
  • the present invention thus provides a solution to two practical problems: reduction of the size of the cell encapsulating chamber 10 so that it can be implanted in a human, and minimization of the cost of the implantable tissue (e.g., islet tissue) by maximizing the number of cells 14 that can survive implantation.
  • implantable tissue e.g., islet tissue
  • the present invention supplements oxygen provided to isolated cells 14 by the blood stream by providing an oxygen generator 18 in close proximity to the cell encapsulating chamber 10 .
  • close proximity refers to a distance through which sufficient oxygen can diffuse to enhance cell 14 viability and/or proliferation within the cell encapsulating chamber 10 in comparison with a device lacking an oxygen generator 18 .
  • the oxygen generator 18 is adjacent to the cell encapsulating chamber 10 .
  • the oxygen generator 18 provides a continuous supply of reactant water from the surrounding host cells/tissue/fluids, continuous diffusion of generated gases out of the oxygen generator/electrolyzer 18 , and exclusion of biological components that would otherwise contaminate the oxygen generator/electrolyzer 18 . It is clear that the oxygen generator 18 can supply oxygen to the transplanted cells 14 at a pO 2 far higher than is possible from any physiological source. Furthermore, it is possible to completely prevent exposure of the transplanted system to the sustained hypoxic environment following transplantation. However, the pO 2 value should be optimized to limit oxygen toxicity that may result from chronic exposure at high pO 2 .
  • F is Faraday's constant (96,500 A ⁇ s/equivalent).
  • N e M A e ( 2 )
  • an understanding of oxygen diffusion and consumption in the system provides a framework to use to calculate the necessary parameters of the oxygen generator 18 which may be optimized to maximize oxygen generation and delivery to cells 14 while minimizing the size of the oxygen generator 18 .
  • P Se and P Sm are the values of P at the tissue-semipermeable barrier layer 9 interfaces at the oxygen generator 18 and medium side, respectively
  • D is the effective diffusion coefficient of oxygen in tissue
  • is the Bunsen solubility of oxygen in tissue.
  • P Se and P Sm are not known a priori but can be calculated in terms of the known quantities N e (the imposed oxygen flux from the oxygen generator/electrolyzer 18 ) and P med (the oxygen partial pressure in the medium bulk):
  • the oxygen generator 18 is coupled to an energy source 199 , such as a battery or a current controller.
  • an energy source 199 such as a battery or a current controller.
  • a small implanted battery with modest current densities and power requirements which is periodically recharged transcutaneously can provide a viable transplanted tissue thickness in excess of 200 ⁇ m based on mathematical models which describe oxygen diffusion and consumption in implanted immunobarrier devices. Given a pO 2 at the host-device interface or an imposed oxygen flux, the maximum tissue thickness (L) that can be supported by one face and the maximum pO 2 to which the tissue is exposed can be readily estimated.
  • a source 199 of energy for the oxygen generator 18 is 2 AA-sized nickel-cadmium batteries.
  • the stored energy of about 450 mAh would require recharging after about 4 days.
  • Button cells with up to 3600 mAh capacity are also encompassed within the scope of the invention and would require recharging every 4 weeks or so.
  • the recharging of an energy source 199 in communication with the oxygen generator 18 is performed transcutaneously.
  • this is achieved with a transcutaneous energy transfer (TET) system of the type developed to power implantable devices such as artificial hearts, left ventricular assist devices, defibrillators, and electrical stimulators which are well known in the art. These devices have been under development since the 1960s.
  • TET transcutaneous energy transfer
  • Cells 14 were cultured in Dulbecco's Modification of Eagle's Medium (DMEM, Mediatech, Herndon, Va.) with 400 mg/dl glucose, supplemented by 10% (v/v) fetal bovine serum (FBS, HyClone, Logan, Utah), 100 IU penicillin, and 100 ⁇ g/ml streptomycin (Mediatech). Prior to loading the chamber, cells 14 were detached from T-flasks using trypsin-EDTA solution (Mediatech) and washed three times by centrifugation, removal of the supernatant, and resuspension in culture medium.
  • DMEM Dulbecco's Modification of Eagle's Medium
  • FBS fetal bovine serum
  • FBS HyClone, Logan, Utah
  • streptomycin 100 ⁇ g/ml streptomycin
  • the assembled cell encapsulating ring chamber 10 was immersed in 10 mM BaCl 2 solution for 30 seconds to crosslink the alginate. After three washes by immersion and shaking for 10 s in Ca 2+ and Mg 2+ -free PBS, the loaded ring chamber was ready to be cultured in vitro. Prior to loading, sterilization was achieved by autoclaving the titanium rings 32 and washers 33 , placing the oxygen generator/electrolyzer 18 in boiling water for at least 15 min, and immersing the semipermeable barrier layer 9 laminated membrane in 95% ethanol, 80% ethanol, and three times in PBS for at least 5 min in each. Sterilization was maintained by assembling the device in a laminar flow hood.
  • the cell encapsulating chamber 10 was oriented so that oxygen generation occurred below the bottom face of the ring immunoisolation chamber 10 .
  • the flask was then placed in an incubator and the oxygen generator 18 was connected to a power source 70 outside the incubator. Control experiments were carried out simultaneously with the oxygen generator 18 not connected to a power source 70 .
  • TET transcutaneous energy transfer

Abstract

An apparatus and method to enhance oxygen delivery to tissue encapsulated in an immunobarrier device. In one embodiment, in situ electrochemical oxygen generation by electrolysis of water into oxygen and hydrogen supplies oxygen to the tissue. In one embodiment a thin, multilayer sheet electrolyzer is enclosed in silicone rubber membranes permeable to gas and water vapor but not liquids. The anode side of the electrolyzer is in contact with one face of the device. Water vapor diffuses into the electrolyzer, decomposes at the anode, and oxygen diffuses back through the membrane to the implanted tissue, thus providing a continuous supply of water, continuous diffusion of O2 and H2 out of the electrolyzer, and exclusion of biological components that might contaminate the electrolyzer.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims priority under 35 U.S.C. 35 §119 (e) to U.S. Provisional Application No. 60/093,147, filed Jul. 17, 1998, the entire disclosure of which is incorporated herein by reference.[0001]
  • FIELD OF THE INVENTION
  • The invention relates generally to devices for delivering oxygen to cells and more specifically to devices which deliver oxygen in situ to cells in the body of an organism. [0002]
  • BACKGROUND
  • Techniques to transplant cells into people in need of the functions that these cells provide have application in the treatment of a variety of chronic conditions including diabetes, hemophilia, dwarfism, anemia, kidney failure, hepatic failure, familial hypercholesterolemia, immunodeficiency disorders, pituitary disorders, and central nervous system disorders. [0003]
  • Cell implantation techniques are typically limited by shortages of cells. For example, successful transplantation of insulin-secreting cells or tissue into people with diabetes has been a challenge because of the obvious shortage of human islet tissue. The approximately 3000 cadaver pancreases that could be available each year in the U.S.A. come nowhere near to meeting the needs of people with Insulin Dependent Diabetes Mellitus (IDDM). Use of cells/tissue from other species as xenogeneic cells may therefore provide the fastest path to clinical application. [0004]
  • Immunobarrier devices have been developed as a means of protecting xenogeneic cells from transplantation rejection by a host organism. The xenogeneic cells are encapsulated at a high, tissue-like density or are dispersed in the form of individual cells or cell aggregates (e.g., islets of Langerhans) in an extracellular gel matrix such as agar, alginate, or chitosan, within these devices. [0005]
  • High-density culture, if attainable, is advantageous because it minimizes the size of the implanted device used in a particular application. This is desirable because the complexity and difficulty of the application increases with the volume of implanted cells/tissue. Consequently, applications which have tended to require the least amount of transplanted cells/tissue, such as central nervous system applications, have been the first to advance to clinical testing. [0006]
  • Maintenance of the viability and function of implanted cells within an immunobarrier device is essential and limited by the supply of nutrients and oxygen which can be provided to the cells from the host. Apoptosis in transplanted tissues has been observed and may be a general response to severe hypoxia, as well as to methods of isolation and culture, glycemic state, and the nonspecific inflammatory reaction associated with the transplantation procedure. [0007]
  • The impact of hypoxia is also influenced by the type of cells/tissues being implanted. For example, pancreatic islet cells are especially prone to oxygen supply limitations because they have a relatively high oxygen consumption rate. They are normally highly vascularized and are supplied blood at arterial pO[0008] 2. When cultured in vitro under normoxic conditions, islets develop a necrotic core, the size of which increases with increasing islet size, as is to be expected as a result of oxygen diffusion and consumption within the islet. However, the death of implanted cells due to hypoxia is not the only concern. Oxygen levels high enough to keep cells alive can nonetheless have deleterious effects on cell functions that require higher cellular ATP concentrations, for example, ATP-dependent insulin secretion.
  • Only scant attention has been paid to the issue of islet viability within implanted immunobarrier devices. However, recently, critical parameters such as the number and volume of viable islet cells that can be supported by such devices, and the development of islet necrosis and fibrosis in such devices has begun to be examined. It is clear from these recent studies that all attempts to support larger volumes of islet tissue in high-density culture (i.e., where all or most of the internal device volume is occupied by viable islet tissue) have led to massive islet necrosis, invariably in regions furthest from the oxygen source. As with transplantation of naked cells, the hypoxic environment for several days following transplantation appears to be a critical problem. For example, most of the loss of viable β-cell mass undoubtedly occurs during the first few days after transplantation within these devices. [0009]
  • With few exceptions, only by suspending islets in an extracellular gel matrix at very low islet volume fractions (e.g., 1 to 5%), which greatly increases the size of the implanted device, have investigators been able to maintain the viability of the initially loaded islets. However, use of such low tissue density puts undesirable constraints on the maximum number of islets that can be supported in a device of a size suitable for surgical implantation. [0010]
  • Attempts to modify the design of immunobarrier devices have been made to try to overcome these limitations. A biohybrid artificial pancreas for insulin secretion known in the art consists of a semipermeable membrane tube through which arterial blood flows. The membrane tube is surrounded by the implanted tissue which is, in turn, contained in a housing. This approach provides the highest available pO[0011] 2 (100 mm Hg) but suffers from the need to open the cardiovascular system; thus, it may be limited to only a small fraction of patients.
  • One alternative is an extravascular device in the form of a planar or cylindrical diffusion chamber implanted, for example, in subcutaneous tissue or intraperitoneally. Such devices are exposed to the mean pO[0012] 2 of the microvasculature (about 40 mm Hg) limiting the steady state thickness of viable tissue that can be supported. Further limits are imposed when such devices are implanted into soft tissue. If a foreign body response occurs, an avascular fibrotic tissue layer adjacent to the chamber can be produced, typically on the order of 100 μm thick. This fibrotic tissue increases the distance between blood vessels and implant, and the fibroblasts in fibrotic tissue layer also consume oxygen. Oxygen deficits are especially likely during the first few days after implantation before neovascularization has a chance to occur. Anoxia may exist within regions of the device, leading to death of a substantial fraction of the initially implanted tissue.
  • Microporous membranes that induce neovascularization at the device-host tissue interface have also been used. This angiogenic process takes 2-3 weeks for completion, and the vascular structures induced remain indefinitely. By bringing some blood vessels close to the implant, oxygen delivery is improved. Oxygen delivery also may be improved by prevascularizing the device, e.g, by infusion of an angiogenic factor(s) through the membranes into the surrounding tissue. [0013]
  • Another means of implanting cells in an extravascular environment involves the use of spherical microcapsules. The microcapsules comprise small quantities of cells enclosed in a semipermeable membrane and can be implanted in an extravascular space, for example, in the peritoneal space. However, the large volume of microcapsules employed, and the tendency for most to permanently attach to peritoneal surfaces, may lead to clinical problems. Thus, despite encouraging results with various tissues and applications, the problem of oxygen transport limitations remain. [0014]
  • The present invention improves the viability and function of encapsulated tissue. [0015]
  • SUMMARY OF THE INVENTION
  • The invention provides an oxygen generator device for delivering oxygen to cells or to a cell compatible fluid. The oxygen generator device disclosed herein has application for in vitro or in vivo use. In one aspect the device is placed in proximity to a cell compatible fluid. In another aspect, the oxygen generator is placed in proximity to cells for which supplemental oxygen is desired. In a further aspect of the invention, the oxygen generator is placed in proximity to a cell encapsulating device. The oxygen generator disclosed herein provides a system to deliver oxygen to cells in situ in the body of an organism. [0016]
  • In one embodiment of the invention, the oxygen generator is an electrolyzer device which electrolyzes water into oxygen and hydrogen. In another embodiment of the invention, the oxygen generator is in the form of a thin, multilayer electrolyzer sheet and is permeable to gas and water vapor but impermeable to liquids and dissolved material. In a further embodiment of the invention, the oxygen generator comprises a multilayer electrolyzer sheet having a proton exchange membrane sandwiched by an anode layer and a cathode layer. In a further embodiment of the invention, the device comprises a multilayer electolyzer sheet adapted for mating to a container containing cells. [0017]
  • In one embodiment of the invention, the oxygen generator is in communication with an energy source, such as a battery. In another embodiment of the invention, the battery is rechargeable transcutaneously. In a further embodiment of the invention, the battery is recharged using a transcutaneous energy transfer system (TET) system. In a further embodiment, the invention may be operated directly and continuously from a battery-powered TET system. [0018]
  • In one embodiment of the invention, the oxygen generator is provided in proximity to cells in vitro. The oxygen generator is provided, either within a container containing the cells, or as an integral part of the container. In one embodiment of the invention, the oxygen generator is provided, either within, or as an integral part of, a cell-containing cartridge in an extracorporeal circuit device. [0019]
  • In a further embodiment of the invention, the oxygen generator is provided as an in-line oxygenator through which blood, plasma, and other bodily fluids may flow. In this embodiment, the oxygen generator is mated to, contained within, or is an integral part of, a hollow tube through which blood, plasma, and other bodily fluids, or culture medium may flow. [0020]
  • In one embodiment, the invention provides a system for delivering oxygen in situ to cells within the body of an organism. In this embodiment of the invention, the system comprises an oxygen generator positioned in proximity to a cell encapsulating chamber and is implanted within the body of an organism. The cell encapsulating chamber comprises a containment space for cells bounded by a semipermeable barrier layer which acts as a selective diffusion layer, allowing selected components to enter and leave the cell encapsulating chamber. [0021]
  • In a further embodiment of the invention, the system includes an oxygen generator which comprises a multilayer electrolyzer sheet mated to a cell encapsulating chamber comprising two semipermeable membranes sealed together by a ring seal. [0022]
  • In another embodiment of the invention, the system for delivering oxygen in situ comprises a cell encapsulating chamber which defines an immunoisolation chamber. In this embodiment, the semipermeable barrier layer of the immunoisolation chamber immunoisolates cells contained within the chamber when the device is exposed to components of the immune system. In a further embodiment, the invention relates to a system implantable in the body of an organism for growing tissue in immunoisolation while providing supplemental oxygen to the tissue. [0023]
  • In another embodiment of the invention, the system for delivering oxygen in situ comprises an oxygen generator provided in proximity to, or mated with, a semipermeable membrane tube through which blood, plasma, and other bodily fluids may flow. In another embodiment of the invention, the membrane tube is surrounded by implanted tissue. In a further embodiment of the invention, the membrane tube and tissue is contained within a housing. [0024]
  • The invention also relates to a system for delivering oxygen in situ to cells which are not contained within a cell encapsulating device. In a one embodiment of the invention, the system for delivering oxygen in situ comprises an oxygen generator placed in proximity to cell-containing microcapsules which are free to migrate within an intraperitoneal space. In another embodiment of the invention, the system comprises an oxygen generator positioned in proximity to a cell-supporting, biocompatible, polymeric scaffold within the body of an organism. In this embodiment, the oxygen generator can be used to maintain, and support the growth of artificial tissues. [0025]
  • In a further embodiment of the invention, two oxygen generators are placed back to back with cells on both sides of the oxygen generator, maximizing the amount of oxygen that can be delivered to cells. [0026]
  • In another embodiment, the system is designed to deliver oxygen in situ to cells located at a distance from the oxygen generator. A tube with low oxygen permeability is attached to the oxygen generator. Generated oxygen is transferred through the tube to a flexible oxygen distributor fabricated from oxygen-permeable membranes. In one aspect of the invention, the oxygen distributor is placed in proximity to cells, tissues, or organs, for which supplemental oxygen is desired. In another aspect of the invention, the oxygen distributor is placed in proximity to a cell encapsulating device. The flexible oxygen distributor provides a means to deliver oxygen from an oxygen generator located at a distance to cells, tissues, or organs located anywhere, and having any shape, within the body of an organism. [0027]
  • In a further embodiment of the invention, the oxygen generator is an oxygen transfer device which electrochemically transfers oxygen from the cathode side to the anode side of the oxygen generator substantially without the generation of hydrogen. [0028]
  • The invention also relates to methods of delivering oxygen to cells in vitro comprising positioning an oxygen generator in proximity to the cells. In one embodiment of the invention, the method comprises delivering oxygen to cells contained within a container, such as a cell culture dish or a flask. In another embodiment of the invention, the method comprises placing an oxygen generator in proximity to the cells within a cell-containing cartridge in an extracorporeal circuit or in culture medium in a perfusion circuit. In another embodiment of the device, the oxygen generator is provided as an in-line oxygenator through which culture medium flows. In this embodiment, oxygenated culture medium flows through the cell-containing cartridge and is then discarded or recycled through the oxygenator for reoxygenation. In a further embodiment of the invention, the oxygenated culture medium is made to flow in and around an organ for transplantation, such as heart, kidney, liver, or pancreas, during the period when it is stored or shipped. [0029]
  • The invention also relates to a method of delivering oxygen in situ to cells within a body of an organism. In this embodiment of the invention, an oxygen generator is placed in proximity to cells which are free to migrate within an intraperitoneal space. In another embodiment of the invention, the method includes implanting a system within the body of an organism, the system comprising an oxygen generator which is in proximity to a cell encapsulating chamber, such as an immunoisolation chamber. [0030]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • In the drawings, like reference characters generally refer to the same parts throughout the different views. Also, the drawings are not necessarily to scale, emphasis instead generally being placed upon illustrating the principles of the invention. [0031]
  • FIG. 1 is a schematic diagram of an oxygen generator according to one embodiment of the invention. [0032]
  • FIG. 2 is a schematic diagram of a system comprising an oxygen generator in proximity to a cell encapsulating chamber in an in vitro embodiment of the invention. [0033]
  • FIG. 3 is is a schematic diagram of a system comprising an oxygen generator in proximity to a cell encapsulating chamber in an in vivo embodiment of the invention. [0034]
  • FIG. 4 shows a graph of viable tissue thickness vs. oxygen flux when using an oxygen generator according to one embodiment of the invention. [0035]
  • FIG. 5 is a schematic TET system used with an oxygen generating device according to one embodiment of the invention. [0036]
  • FIG. 6 is a schematic diagram of a ring cell encapsulation chamber according to one embodiment of the invention. [0037]
  • FIGS. 7[0038] a-f show photomicrographs of hemotoxylin-stained sections of βTC3 cells cultured in vitro in barium alginate within a planar immunoisolation chamber according to one embodiment of the invention.
  • FIGS. 8[0039] a-e show photomicrographs of hemotoxylin-stained sections of βTC3 cells cultured in vitro in barium alginate within a planar immunoisolation chamber according to another embodiment of the invention.
  • FIGS. 9[0040] a-h show photomicrographs of hemotoxylin-stained sections of βTC3 cells cultured in vitro in barium alginate within a planar immunoisolation chamber according to another embodiment of the invention.
  • FIGS. 10[0041] a and 10 b are photomicrographs of hemotoxylin-stained sections of an in vitro cultured planar immunoisolation chamber according to one embodiment of the invention loaded with rat islets (500 islets/device) in barium alginate. FIG. 10a shows cells after four days in the absence of in situ oxygen generation (with the oxygen generator disconnected from its power source). FIG. 10b shows cells after four days with in situ oxygen generation (with the oxygen generator connected to its power source).
  • FIG. 11 shows a graph of oxygen partial pressure vs. distance from the oxygen generator/electrolyzer side/semipermeable barrier layer interface for different values of oxygen flux generated by the electrolyzer.[0042]
  • DETAILED DESCRIPTION
  • The invention relates to an oxygen generator which may be used to deliver oxygen to cells in vitro or in vitro. FIG. 1 is an exploded schematic diagram of the components of an oxygen generator (electrolyzer) [0043] 18, according to one embodiment of the invention, that carries out electrolysis of water. The water is obtained from a cell compatible media, i.e., cell culture media (synthetic or natural), or bodily fluids, such as blood or plasma.
  • The [0044] oxygen generator 18 comprises two catalytic electrodes, i.e., an anode 21 and a cathode 23, separated by a solid polymer electrolyte proton exchange membrane (PEM) 24 which allows passage of hydrated H+ but has much lower permeability to gases such as O2 and H2. As a consequence, the half-cell reactions at each electrode (21 and 23) are compartmentalized. For normal acid electrolysis, the reactions are
    Figure US20030087427A1-20030508-C00001
  • O[0045] 2 is formed at the anode 21. H+, which is also formed at the anode 21, is transported through the PEM 24 under the influence of a potential gradient imposed by a constant current controller 66 in an external circuit 40 shown in phantom in FIG. 1. The hydrogen ions arriving at the cathode 23 recombine with electrons passed through the external circuit 40 from the anode 21 so as to form molecular hydrogen.
  • Because of the permeability properties of the [0046] PEM 24, virtually all of the O2 diffuses into the cells in proximity to the oxygen generator 18, whereas the H2 diffuses away from the oxygen generator 18. The quantity of hydrogen generated is expected to be less than the stoichiometric 2:1 ratio relative to oxygen for normal electrolysis because oxygen diffusing into the cathode 23 side of the oxygen generator 18 from the cell media is preferentially reduced as compared to H+. With oxygen present at the cathode 23, the reaction
    Figure US20030087427A1-20030508-C00002
  • competes with the reaction [0047]
    Figure US20030087427A1-20030508-C00003
  • thereby decreasing the net hydrogen production. The extent of the decrease in net hydrogen production increases with an increase in the rate at which oxygen reaches the cathode surface. [0048]
  • In another embodiment of the invention, both electrodes, [0049] 21 and 23, are covered by silicone rubber membranes 20, so that water enters the oxygen generator 18 only in its vapor form and the interior of the oxygen generator 18 is kept free of all other components of biological fluids. In a further embodiment of the invention, metal contact rings 25 are provided which provide connections to the external circuit 40.
  • An [0050] oxygen generator 18 according to another embodiment of the invention is shown in FIG. 2. The active components of the oxygen generator 18 are the catalytic electrodes, 21 and 23, and the PEM 24. In one embodiment of the invention, the PEM 24 is Nafion 117 (1100 equivalent weight, about 1250 μm thick hydrated, DuPont Co., Wilmington, Del.) which has O2 and H2 permeabilities more than two orders of magnitude less than silicone rubber. The Nafion membrane is received in the Na+ form and is exchanged in acid to convert it to the proton form. The electrodes 21 and 23 are placed in intimate contact with the PEM 24.
  • In a further embodiment of the invention, a substrate is provided for the electrodes, [0051] anode 21 and cathode 23, in the form of a very fine platinum (Pt) screen (150 mesh with 43-μm diameter wire). The cathode 23 catalyst is Pt (25 m2/g), and the anode 21 catalyst is Pt-iridium oxide (100 m2/g). The platinum screens comprise high surface area particles mixed with polytetrafluoroethylene (PTFE) particles and pressed onto a fine-mesh substrate screen which in turn is pressed onto the PEM 24.
  • To make electrical contact with the [0052] electrodes 21 and 23, contact rings 25 are provided which comprise rings of tantalum foil, provided with a tab 67, resistance welded to the edge of the electrodes 21 and 23 before bonding to the PEM 24. The silicone rubber membranes 20 are then hot pressed on either side of the contact rings 25. In a further embodiment of the invention, a disk of fine mesh support screen 19 comprising a biocompatible, inert metal (e.g., titanium) is added over the outside of the silicon rubber membranes 20. In one embodiment of the invention, the entire thickness of the oxygen generator, including the silicon rubber membranes 20 and outer support screens 19, is about 0.75 mm.
  • In another embodiment of the invention, [0053] oxygen generator 18 is enclosed in a housing 68 (e.g., a polysulfone housing) incorporating wire leads 69 for connection to a current control circuit 40 and a power supply 70 (the current control circuit and power supply are shown in phantom in FIG. 2). In a further embodiment of the invention, the housing 68 is adapted for coupling to a container 100, which may be mated to the housing 68 at one face of the housing 68.
  • In operation, the [0054] oxygen generator 18 is provided in communication with a power supply 70 and a constant current controller 40, in order to maintain a constant rate of oxygen generation in the presence of resistance variations in the oxygen generator 18 and declining battery voltage (when a battery/energy source 199 is used). In one embodiment of the invention, a constant current control 40 is implemented with a Howland circuit, a voltage-controlled current source which utilizes positive and negative feedback to provide precise current control. In a further embodiment of the invention, the current control circuit 40 is powered by an AC-DC converter or by a single battery/energy source 199. In a further embodiment of the invention, a potentiometer is provided to adjust current from about 0 to about 200 μA.
  • In a further embodiment of the invention, the [0055] oxygen generator 18 is an oxygen transfer device, in which hydrogen generation at the cathode 23 is suppressed, when the cathode 23 has access to a continuous supply of oxygen, such as air or dissolved oxygen, at a rate in excess of the oxygen generation rate at the anode 21. Oxygen is preferentially reduced at the cathode 23, when available, in lieu of the reduction of protons to hydrogen in the pure electrolysis reaction; the same oxygen generation reaction occurs at the anode in both cases.
  • In a further embodiment of the invention, the [0056] oxygen generator 18 is an oxygen transfer device in which hydrogen generation at the cathode 23 is suppressed by an alternative means; the oxygen generator 18 is made to have a larger geometric area than the cell encapsulating chamber 10, thereby providing the cathode 23 with access to dissolved oxygen in excess of the oxygen generation rate at the anode 21.
  • The oxygen generation rate M (mol/s) of the [0057] oxygen generator 18 is related to the applied current I (A) by
  • I=nFM  (1)
  • where n=4 is the number of electrons required to make one molecule of oxygen, and F is Faraday's constant (96,500 A·s/equivalent). The imposed flux of oxygen N[0058] e from the oxygen generator/electrolyzer 18 into the cell encapsulating chamber/planar diffusion chamber 10 (mol/cm2·s) is N e = M A e ( 2 )
    Figure US20030087427A1-20030508-M00001
  • where A[0059] e=0.317 cm2 is the cross-sectional area of the gap between the oxygen generator/electrolyzer 18 and the cell encapsulating chamber/diffusion chamber 10. Equation (2) can thus be written as
  • i=nFN e  (3)
  • where i=I/A[0060] e is the effective current density at the electrodes 21 and 23.
  • The [0061] oxygen generator 18 disclosed herein may be used in a variety of in vitro and in vivo applications. In one embodiment of the invention, the oxygen generator 18 is provided in proximity to cells in vitro, and is either contained within, or is an integral part of the container 100. Types of containers 100 encompassed within the scope of the invention include, but are not limited to, cell culture plates, flat-bottom tissue culture flasks, roller cell culture flasks, microtiter plates, and the like. In this embodiment of the invention, the oxygen generator18 may be used to provide supplemental oxygen to cells in culture and may be used to enhance the proliferation of these cells. In another embodiment of the invention, the container 100 further comprise an oxygen sensor or other type of biosensor to monitor the level of metabolites in the cell media in which cells are cultured.
  • In another embodiment of the invention, the [0062] oxygen generator 18 is positioned in proximity to cells within the cell-cartridge of an extracorporeal circuit device or within a cell cartridge in a cell perfusion circuit. Such devices are well known in the art. In a further embodiment of the invention the oxygen generator provides oxygen which delivers oxygenated fluids to cells and does not have to be in proximity to the cells themselves. In this embodiment of the invention, the oxygen generator is either mated to, contained within, or is an integral part of, a hollow tube, flat plat channel or other conduit through which cell compatible media, such as cell culture media, plasma, blood, and the like, may flow.
  • The invention also relates to a system for delivering oxygen in situ to cells within the body of an organism. In this embodiment of the invention, the system (shown in FIG. 3) comprises an [0063] oxygen generator 18 positioned in proximity to a cell encapsulating chamber 10 and is implanted within the body of an organism. The cell encapsulating chamber 10 comprises a containment space for cells bounded by a semipermeable barrier layer 9 which acts as a selective diffusion layer, allowing selected components to enter and leave the cell encapsulating chamber 10. The oxygen generator 18 enhances the viability and/or proliferation of cells 14 within the containment space. In a second embodiment of the invention, the oxygen generator 18 includes an energy source 199 (e.g., a battery, current controller, fuel cell, and the like).
  • The system may be used to deliver [0064] cells 14 which can provide a function lacking in a body of a patient. The function may be naturally provided by these cells 14, or the cells 14 may be genetically modified to provide the function. The system may be used in methods of treatment of conditions, especially chronic conditions, including, but not limited to, the treatment of diabetes, hemophilia, dwarfism, anemia, kidney failure, hepatic failure, immunodeficiency disorders, pituitary disorders, and central nervous system disorders. Nervous system disorders which may be treated include, but are not limited to, chronic pain, Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis. The cells 14 may come from a variety of sources, including, but not limited to, pancreas cells, hepatocytes, kidney cells, lung cells, neural cells, pituitary cells, parathyroid cells, thyroid cells, and adrenal cells. Multiple types of cells 14 may be mixed in a single cell encapsulating chamber 10 (e.g., hepatocytes and pancreas cells may be provided within a single isolation chamber 10). As used herein, the term “cells” 14 encompasses individual or isolated cells, cell lines, tissues, such as islets, and cell aggregates. In an embodiment of the invention, a bioartificial organ is provided. As used herein, the term “bioartificial organ” encompasses any device that incorporates both synthetic and living material.
  • In one embodiment of the invention, the βTC3 cell line is used as a source of [0065] cells 14. βTC3 cells are derived from an insulinoma from a transgenic β6DZF1 mouse carrying a hybrid insulin-promoted SV40 T-antigen gene. The βTC3 line is among those genetically engineered β-cell lines which can be used for the treatment of diabetes.
  • The concentration of [0066] cells 14 required for transplantation is determined by the secretion rate of the desired agent by the cell 14 and the amount of active agent required by the body. For example, the average patient with IDDM requires approximately 30 Units of insulin per day to control blood glucose levels. The amount of insulin (Units/cells/day) produced by a population of islet cells is then determined in culture and the number of transplanted cells 14 needed to provide the patient's required insulin dose is loaded within the immunoisolation chamber 10. Typically, this is on the order of about 109 cells 14. In the case of cells 14 that proliferate, a determination of the insulin production rate in culture permits an estimation of the number of cells 14 required in a given volume within a cell encapsulating chamber 10. A small number of cells 14 can be used which will then proliferate to fill the volume of the chamber 10 and provide the necessary amount of insulin. It is readily apparent, that similar calculations may be performed for any application for which a required dosage of active agent is known, or is determinable, and for which the amount of bioactive agent produced by cells 14 may be measured.
  • In addition to [0067] cells 14, the system may be used to deliver a variety of bioactive molecules, including derivatives, analogs, and active fragment of these molecules. Bioactive molecules include, but are not limited to, hormones, growth factors, cytokines, vascularizing agents, receptors, ligands, antibodies, antibody fragments, fusion proteins, peptides (natural or synthetic), drugs, nucleic acids (including naked DNA, DNA encapsulated in liposomes, viral particles, or microspheres, antisense molecules, ribozymes, triple helix DNA, PNA molecules, and aptamers). Autologous molecules, as would be generated by tissues derived from transgenic animals, can be used to minimize immune responses by the host organism. While the system provides a means to immunoisolate transplanted living cells 14 and to prevent the immune system of the recipient organism from attacking the transplanted cells 14, the system may also be used as a bioreactor in in vitro applications.
  • In a further embodiment of the invention, the disclosed system provides a [0068] cell encapsulating chamber 10 which immunoisolates transplanted cells 14 by surrounding cells 14 with a semipermeable barrier layer 9. The semipermeable barrier layer 9 protects the transplanted cells 14 from components of both the cellular and humoral immune system but permits passage of the agent of interest, for example, secreted insulin. The transport properties of the implanted cells 14, semipermeable barrier layer 9, and surrounding host cells/tissue/fluids must permit a sufficient supply of nutrients and oxygen, as well as the removal of metabolic waste products, by diffusion from, or to, the nearest blood supply.
  • As used herein, the term “semipermeable barrier layer” [0069] 9 includes biocompatible encapsulants, such as membranes, which provide a selective diffusion layer which controls the entry and exit of molecules into and out of the cell encapsulating chamber 10. Minimally the barrier layer 9, must be have a pore size, pore density, percent porosity, molecular weight cut off to keep cells within the cell encapsulating chamber 10. Particularly in embodiments where allogeneic or autologous cells 14 are transplanted, the pore size may be quite large as about 0.1 μm but must completely prevent passage of cells 14. In the embodiment of the invention where the cell encapsulating chamber 10 is a xenogeneic immuonisolation chamber, the membranes may have any type of pore size, pore density, percent porosity, molecular weight cutoff, sufficient to keep selected components of the immune system out, yet allowing for the transport of nutrients, oxygen, secreted cellular products (e.g., secreted insulin), metabolic wastes, ions, and other bioactive molecules. When xenogeneic cells 14 are used, the molecular weight cutoff should be about 100,000 daltons or less, so as to prevent components of the humoral immune system from entering into the cell encapsulating chamber 10 and to prevent endogenous retroviruses transmittable by the cells 14, or other infectious macromolecules which might be secreted by the cells, from exiting the cell encapsulating chamber 10.
  • [0070] Semipermeable barrier layer 9 material is selected from the group including, but not limited to, acrylic, polyurethanes, cellulose acetate, agar, alginate, chitosan, or other types of hydrogels, polysulfone, polyether sulfones, polyvinyl chloride, polyvinyl alcohols, polyvinylidene fluoride, polytetrafluoroethylene, polyacryl nitrites, and including derivatives, and/or mixtures thereof. Any porous polymeric material is encompassed within the scope of the invention which has the desired membrane strength (i.e., ability to survive handling, transplantation, and the environment inside the body) and the desired permeability. The cell encapsulating chamber 10 is preferably easily removable from the body and replaceable in the body.
  • The [0071] cell encapsulating chamber 10 can be a variety of shapes including tubes or cylinders, cubes, spheres, discs, or sheets, so long as it is able to provide a sufficient containment surface for the number of cells 14 suitable for a given application. In one embodiment (shown in FIG. 6), the cell encapsulating chamber 10 is a ring chamber which includes two thin disc-shaped semipermeable barrier layer 9 membranes sealed together on their circular periphery by a ring seal 32. Suitable ring seal 32 materials include, but are not limited to, biocompatible, noncorrosive metals (e.g., titanium), Teflon®, plastic and the like. In this configuration, the space between the two semipermeable barrier layer 9 membranes contains the cells 14 to be implanted. The semipermeable barrier layer 9 membranes are both distensible and conformable. Distensible semipermeable barrier layer 9 membranes allow for the expansion of the cell encapsulating chamber 10, which is important when proliferating cell lines are used. When islets are the source of cells 14, the conformable semipermeable barrier layer 9 membranes partially conform to the shape of the cells 14, leading to a structure akin to peanut brittle. In a further embodiment of the invention the oxygen generator 18 can be provided proximal to, mated with, or an integral part of, a hollow tube through which blood flows. In this embodiment of the invention, cells 14 are implanted outside the tube. In a further embdiment of the invention, the cells 14 are surrounded by a housing.
  • The [0072] semipermeable barrier layer 9 of the cell encapsulating chamber 10 may be coated by other biocompatible molecules on the interior (i.e., the side proximal to the encapsulated cells 14), such as polymeric scaffolds which may also be coated by bioactive molecules such as ECM proteins, morphogenic proteins, growth factors, cytokines, and/or polysaccharides. As shown in FIG. 3, the interior of the semipermeable barrier layer 9 membrane in one embodiment further includes a gel matrix 15 (e.g., agar, alginate, chitosan, polyglycolic acid, polylactic acid, and the like) in which the cells 14 are embedded. In another embodiment, cells 14 in the cell encapsulating chamber 10 may themselves be encapsulated within microcapsules. The semipermeable barrier layer 9 membrane may also be coated with an anti-microbial and/or an anti-thrombotic agent on its outside surface.
  • The present invention provides a means of minimizing the size of a [0073] cell encapsulating device 10. For illustrative purposes, consider a planar diffusion immunoisolation chamber and the limiting case in which islet cells 14 occupy all available internal volume. The surface area A required by the device is given by A=V/h, where V is the required islet volume and h is the maximum total thickness of tissue that can be supported. If V=1 cm3 and h=50 μm, then A=200 cm2 (about 5.6 inches square). If enhanced oxygen delivery allows an increase to h=200 μm, then A=50 cm2 (about 2.8 inches square). This reduction in required area makes it feasible to implant a single or small number of planar immunobarrier device(s).
  • The present invention thus provides a solution to two practical problems: reduction of the size of the [0074] cell encapsulating chamber 10 so that it can be implanted in a human, and minimization of the cost of the implantable tissue (e.g., islet tissue) by maximizing the number of cells 14 that can survive implantation.
  • The present invention supplements oxygen provided to [0075] isolated cells 14 by the blood stream by providing an oxygen generator 18 in close proximity to the cell encapsulating chamber 10. As used herein, the term “close proximity” refers to a distance through which sufficient oxygen can diffuse to enhance cell 14 viability and/or proliferation within the cell encapsulating chamber 10 in comparison with a device lacking an oxygen generator 18. In one embodiment of the invention, the oxygen generator 18 is adjacent to the cell encapsulating chamber 10.
  • In one embodiment of the invention, the system for providing an oxygen to cells in situ comprises an [0076] oxygen generator 18 which is a multilayer electrolyzer, of the type discussed above, comprising an anode layer 21 and a cathode layer 23 sandwiching a proton exchange membrane 24. Oxygen is supplied to implanted tissue by diffusion from the host side and the anode side 21 of the oxygen generator 18. In contrast, nutrients are supplied from, and wastes and secreted insulin are removed from, the faces of the implantable device which are in contact with host cells/tissue/fluid (e.g., the faces of the cell encapsulation chamber 10 not in contact with the oxygen generator 18). The maximum total amount of cells 14 that can be viably maintained within the immunoisolation chamber 10 is the sum of that which can be substituted by each side of the device.
  • The [0077] oxygen generator 18 provides a continuous supply of reactant water from the surrounding host cells/tissue/fluids, continuous diffusion of generated gases out of the oxygen generator/electrolyzer 18, and exclusion of biological components that would otherwise contaminate the oxygen generator/electrolyzer 18. It is clear that the oxygen generator 18 can supply oxygen to the transplanted cells 14 at a pO2 far higher than is possible from any physiological source. Furthermore, it is possible to completely prevent exposure of the transplanted system to the sustained hypoxic environment following transplantation. However, the pO2 value should be optimized to limit oxygen toxicity that may result from chronic exposure at high pO2.
  • In the instant invention, the rate of oxygen generated per unit area of the oxygen generator/electrolyzer [0078] 18 (oxygen flux, mol O2/cm2·s) is determined quantitatively by the current density between the anode layer 21 and cathode layer 23, which can be set at will to provide the optimum oxygen flux required to maintain the maximum viability of the cells 14. Factors such as the site of implantation and the local oxygen partial pressure (pO2) in the blood, the spatial distribution of host blood vessels in the vicinity of the implant surface, the oxygen permeability of the semipermeable barrier layer 9, the oxygen consumption rate of the encapsulated cells 14, the geometrical characteristics of the implant system, and the tissue density and spatial arrangement of the encapsulated cells 14 will be considerations in optimizing oxygen flux.
  • The oxygen generation rate M (mol/s) of the electrolyzer is related to the applied current I (A) by[0079]
  • I=nFM  (1)
  • where n=4 is the number of electrons required to make one molecule of oxygen, and F is Faraday's constant (96,500 A·s/equivalent). The imposed flux of oxygen N[0080] e from the oxygen generator/electrolyzer 18 into the cell encapsulating chamber/planar diffusion chamber 10 (mol/cm2·s) is N e = M A e ( 2 )
    Figure US20030087427A1-20030508-M00002
  • where A[0081] e =0.317 cm2 is the cross-sectional area of the gap between the oxygen genreator/electrolyzer 18 the cell encapsulating chamber/diffusion chamber 10. Equation (1) can thus be written as
  • i=nFN e  (3)
  • where i=I/A[0082] e is the effective current density at the electrodes 21 and 23.
  • As discussed below, an understanding of oxygen diffusion and consumption in the system provides a framework to use to calculate the necessary parameters of the [0083] oxygen generator 18 which may be optimized to maximize oxygen generation and delivery to cells 14 while minimizing the size of the oxygen generator 18.
  • We consider one dimensional oxygen diffusion, perpendicular to the [0084] membranes 9 in Cartesian coordinates. Oxygen consumption is assumed to follow Michaelis-Menten kinetics with the local rate per unit volume V given by V=Vmax·(1−ε)·P/(Km+P) for P>PC, where P is the local oxygen partial pressure, Vmax is the maximum oxygen consumption rate per unit volume of tissue, 1−ε is the live tissue volume fraction in the tissue layer, Km is the Michaelis-Menten constant, and PC is the critical value of P below which loss of tissue viability occurs due to hypoxia, and oxygen consumption ceases. We present here the analytical solution to the oxygen diffusion-reaction equation using zero-order kinetics (Km set equal to 0, V=Vmax (1−ε)=constant) and then examine the small difference that results from the solution obtained with numerical methods using the nonlinear Michaelis-Menten kinetics.
  • Assuming that all the tissue layer within the [0085] cell encapsulating chamber 10 is exposed to P>PC, the solution for the local oxygen partial pressure is given by P ( x ) = P Se + P Sm 2 + ( P Se - P Sm ) x L - V 2 D α [ ( L 2 ) 2 - x 2 ] ( 4 )
    Figure US20030087427A1-20030508-M00003
  • where x is the distance from the center plane of a tissue slab having a thickness L, considered positive in the direction towards the [0086] oxygen generator 18, PSe and PSm are the values of P at the tissue-semipermeable barrier layer 9 interfaces at the oxygen generator 18 and medium side, respectively, D is the effective diffusion coefficient of oxygen in tissue, and α is the Bunsen solubility of oxygen in tissue. PSe and PSm are not known a priori but can be calculated in terms of the known quantities Ne (the imposed oxygen flux from the oxygen generator/electrolyzer 18) and Pmed (the oxygen partial pressure in the medium bulk):
  • P Se =P med+(N e −VL)R ext  (5)
  • and [0087] P Se = P med + ( N e - VL ) ( L D α + R ext ) + V L 2 2 D α ( 6 )
    Figure US20030087427A1-20030508-M00004
  • where R[0088] ext is the sum of the diffusion resistances in series external to the tissue, i.e. those imposed by the semipermeable barrier layer 9 membrane laminate and boundary layer, given by R ext = 1 k c α med + ( L D α ) M 2 + ( L D α ) M 1 ( 7 )
    Figure US20030087427A1-20030508-M00005
  • M[0089] 1, and M2 refer to the cell 14 retentive and vascularizing membranes, respectively, kc=Dmedc is the boundary layer mass transfer coefficient between the stirred medium and the vascularizing membrane M2, and δc is the concentration boundary layer thickness (including the outermost polyester mesh).
  • The maximum thickness of viable tissue L[0090] max that can be supported in the system is
  • L max =L e +L m  (8)
  • where L[0091] e and Lm are the maximum tissue thicknesses that can be supported by the imposed oxygen flux and by oxygen diffusion from the bulk medium, respectively. Le can be calculated by making a mass balance around the tissue layer
  • N m =Ne−VL  (9)
  • and setting the oxygen flux at the tissue-[0092] semipermeable barrier layer 9 membrane interface at the medium side Nm equal to 0: L e = N e V ( 10 )
    Figure US20030087427A1-20030508-M00006
  • L[0093] m, equal to the distance from the tissue-semipermeable barrier layer 9 membrane interface at the medium side (xC=−L/2) to the point where the oxygen flux is equals 0 and P=PC, is given by L m = - D α R ext + [ ( D α R ext ) 2 + 2 D α V ( P med - P c ) ] 1 / 2 ( 11 )
    Figure US20030087427A1-20030508-M00007
  • When the tissue thickness is not large enough for P to fall to P[0094] C, the minimum value of oxygen partial pressure Pmin can be calculated in a similar way, by replacing PC by Pmin and Lm by L−Le to yield P min = P med - V ( L - L e ) R ext - V 2 D α ( L - L e ) 2 ( 12 )
    Figure US20030087427A1-20030508-M00008
  • This minimum occurs at distance x[0095] min=L/2−Le from the tissue-semipermeable barrier layer 9 membrane interface at the oxygen generator/electrolyzer 18 side, provided that Le≦L.
  • For purpose of illustration, the oxygen partial pressure profiles were calculated in immunoisolated tissue supported on one face by an imposed oxygen flux from an [0096] oxygen generator 18 for the case that L≧Le so that P decreases to PC in the interior of the cell encapsulating chamber 10. Transport from the medium was not included. For zero-order kinetics, Equations (4) and (10) were used to calculate the maximum supportable thickness Le, the local partial pressure P(z) where z=L/2−x, and the surface partial pressure PSe. Calculation of the same dependent variables using Michaelis-Menten kinetics was carried out numerically. Parameter values selected for these calculations were 1−ε=0.75, Vmax=2.76×10−8 mol/cm3·s, V=2.07×10−8 mol/cm3·s, Dα=1.70×10−14 mol/cm3·mm Hg·s, 22 Pc=0.1 mm Hg, and Km=0.44 mm Hg. Oxygen partial pressures are plotted as a function of distance from the interface in FIG. 11 for values of imposed oxygen flux Ne ranging from 1 to 6×10−10 mol/cm2·s. These values of Ne correspond to current densities i calculated from Equation (3) of 39 to 232 μA/cm2 and to applied currents I of 12 to 73 μA (with Ae=0.317 cm2 in our system). With increasing oxygen flux, both the interface partial pressure and the maximum thickness increase. The underprediction of P(z) by zero order as compared to Michaelis-Menten kinetics is hardly noticeable on the linear plot but is apparent on the semi-logarithmic plot (lower panel). At low partial pressures, the relative, difference in P(z) can be large.
  • FIG. 4 is a plot of the interface oxygen partial pressure P[0097] se and the maximum viable tissue thickness supported by the oxygen generator/electrolyzer 18 Le as a function of the imposed oxygen flux Ne. At the highest oxygen flux investigated, Le is about 300 μm, and Pse is about 500 mm Hg. Oxygen toxicity at these Pse levels may be of concern with some cells and tissues, thereby setting a maximum limit on the imposed oxygen flux.
  • There is virtually no difference in FIG. 4 between the two predictions for P[0098] se, but the zero-order model underpredicts Le by nearly 20 μm, except at very low Ne, as a consequence of the difference in profiles that develops at very low P(z) (FIG. 11). Because the error in estimating oxygen partial pressure profiles is negligible and the error in Le relatively small, the zero-order kinetic model is appropriate for optimizing performance of the oxygen generator 18.
  • The above parameters provide a means to calculate the optimal amount of oxygen generation that may be produced by a given [0099] oxygen generator 18.
  • While the production of oxygen by the [0100] oxygen generator 18 should be monitored and optimized, hydrogen production in vivo is not a problem. Hydrogen produced at the cathode side 23 of the oxygen generator 18 diffuses through the silicone rubber membrane 20 into the surrounding tissue to the bloodstream from which it is expelled in the lungs. Hydrogen is biologically inert in mammals under normal atmosphere conditions and is not oxidized by mammalian tissues under hyperbaric conditions.
  • In operation, the [0101] oxygen generator 18 is coupled to an energy source 199, such as a battery or a current controller. A small implanted battery with modest current densities and power requirements which is periodically recharged transcutaneously can provide a viable transplanted tissue thickness in excess of 200 μm based on mathematical models which describe oxygen diffusion and consumption in implanted immunobarrier devices. Given a pO2 at the host-device interface or an imposed oxygen flux, the maximum tissue thickness (L) that can be supported by one face and the maximum pO2 to which the tissue is exposed can be readily estimated. Using host interface values of 10, 40, and 100 mm Hg, and oxygen generation with maximum pO2 of 100 and 300 mm Hg, the estimate for L is 22, 47, 78, 102, and 169 μm, respectively. Thus, the optimal total thickness supported is 78+169=247 μm, which would lead to a planar immunobarrier device having 1 cm3 high-density tissue volume with a surface area of about 40 cm2. The (maximum) oxygen flux would be about 5×10−10 mole/cm2·s, requiring a current density of 200 μA/cm2, a very modest level, a total current of 8 mA, and about 16 mwatts (with power supplied at 2V DC).
  • In one embodiment of the invention, a [0102] source 199 of energy for the oxygen generator 18 is 2 AA-sized nickel-cadmium batteries. In this embodiment, the stored energy of about 450 mAh would require recharging after about 4 days. Button cells with up to 3600 mAh capacity are also encompassed within the scope of the invention and would require recharging every 4 weeks or so.
  • In a further embodiment of the invention, the recharging of an [0103] energy source 199 in communication with the oxygen generator 18 is performed transcutaneously. In one embodiment of the invention, this is achieved with a transcutaneous energy transfer (TET) system of the type developed to power implantable devices such as artificial hearts, left ventricular assist devices, defibrillators, and electrical stimulators which are well known in the art. These devices have been under development since the 1960s.
  • Power from 5 to 70 watts has been transmitted through skin thicknesses of 3-15 mm at 60 to 80% efficiency using TET systems and implantable devices using TET systems can tolerate radial misalignments of up to 20 mm. In one embodiment of the invention, shown in FIG. 5, a TET system is provided which comprises a [0104] primary transmitter coil 34 and a secondary receiver coil 35. The primary coil 34 is outside of the body 36 and transmits a transmitted signal 38 to a secondary coil 35 inside the body 36. The secondary coil 35 is in communication with the energy source/battery 199, which in turn, is in communication with the oxygen generator 18 and can recharge energy within the energy source/battery 199. The TET system, thus ensures a continuous supply of energy to the oxygen generator 18, and therefore a continuous supply of oxygen, to the cell encapsulating chamber 10. In a further embodiment of the invention, a voltage control unit 37 is provided which controls the voltage and current in the primary coil 34 in response to a feedback signal 39 from the secondary coil 35. The TET secondary coil 35 can provide output voltages from 10 to 25 V for current loads of 0.5 to 4.0 amps. These capabilities greatly exceed the requirements for a biohybrid artificial pancreas. Thus, in one embodiment of the invention, a TET system is used for recharging implanted batteries on an overnight basis every few days (depending on implanted battery capacity). The TET system may also be worn continuously, using only small interchangeable external battery packs to drive the TET.
  • In another embodiment of the invention, the [0105] oxygen generator 18 may be used to provide supplemental oxygen to cells 14 in situ which are not encapsulated within a cell encapsulating device 10. In this embodiment of the invention, the oxygen generator 18 is implanted in the body in proximity to a desired site containing cells 14 for which supplemental oxygen is desired. In one embodiment of the invention, the oxygen generator 18 is provided in proximity to cells 14 encapsulated in microcapsules which are free to migrate within an intraperitoneal space. Methods of microencapsulating cells are well known in the art. In another embodiment, the oxygen generator 18 is provided in proximity to a cell-supporting, biocompatible, polymeric scaffold within the body of an organism, such as those used to fabricate artificial tissues. In all of these embodiments, the oxygen generator 18 may also be provided coupled to an energy source 199 such as a battery, which may be rechargeable by a recharging system, such as a TET system.
  • In another embodiment, the system is used to deliver oxygen in situ to [0106] cells 14 located at a distance from the oxygen generator 18. A tube with low oxygen permeability is attached to the oxygen generator 18. Generated oxygen is transferred through the tube to a flexible oxygen distributor fabricated from oxygen permeable membranes. In one aspect of the invention, the oxygen distributor is placed in proximity to cells, tissues, or organs, for which supplemental oxygen is desired. In another aspect of the invention, the oxygen distributor is placed in proximity to a cell encapsulating chamber 10. The flexible oxygen distributor provides a means to deliver oxygen from an oxygen generator 18 located at a distance to cells, tissues, or organs located anywhere, and having any shape within the body of an organism.
  • EXAMPLE 1
  • In vitro experiments in which [0107] βTC3 cells 14 are cultured in a cell encapsulating chamber 10 mated to an oxygen generator/electrolyzer 18 in a stirred flask have been performed. In this embodiment of the invention, the cell encapsulating chamber 10 was in the form of a ring immunoisolation chamber (shown schematically in FIG. 6) having a semipermeable barrier layer 9 membrane consisting of three layers: (a) a hydrophilic polytetrafluoroethylene (PTFE) inner membrane with nominal pore size of 0.45 μm and thickness of 25-35 μm (Biopore Millipore Corp, Bedford, Mass.), which retained cells and would function in vivo as an immunobarrier layer; (b) a hydrophobic PTFE outer membrane with nominal pore size of 5 μm and thickness of approximately 15 μm (W. L. Gore and Associates, Elkton, Md.), which would function in vivo as the vascularizing layer; and (c) a highly open outer meshwork about 125 μm thick of polyester fibers (about 50 μm in diameter) to provide support. The space between the two pieces of membrane was defined by a silicone rubber washer 33 (inner diameter 6.6 mm, cross-sectional thickness 100 μm after the device is compressed during assembly).
  • [0108] Cells 14 were cultured in Dulbecco's Modification of Eagle's Medium (DMEM, Mediatech, Herndon, Va.) with 400 mg/dl glucose, supplemented by 10% (v/v) fetal bovine serum (FBS, HyClone, Logan, Utah), 100 IU penicillin, and 100 μg/ml streptomycin (Mediatech). Prior to loading the chamber, cells 14 were detached from T-flasks using trypsin-EDTA solution (Mediatech) and washed three times by centrifugation, removal of the supernatant, and resuspension in culture medium. After the first wash, cells 14 were suspended in 20 ml culture medium, and four aliquots of 200 μl each were taken, and counted in a hemocytometer with Trypan Blue staining. The average of these four counts was used to calculate the total cell 14 numbers in the original cell 14 suspension. Only batches with viability greater than 95% were used for loading. After the final wash, a thin Pasteur pipette (made by pulling a regular Pasteur pipette over flame) was used to remove as much supernatant as possible without removing cells 14, and the cells 14 were resuspended in either phosphate buffered saline (PBS) or 1.7% (w/w) alginate solution to achieve the desired cell concentration. The suspension was carefully mixed without forming any air bubbles. A semipermeable barrier layer 9 consisitng of a laminated membrane was placed inside the titanium housing ring 32 with the hydrophilic side up and the silicone rubber washer 33 was placed on top of the membrane. A volume of 3.5 μl of cell 14 suspension was drawn into a 20 μl pipette tip and discharged at once on the center of the membrane. The other piece of membrane, with the hydrophilic side down, followed by the titanium sealing ring, were then placed on top of the cell 14 suspension, and the whole cell encapsulating ring chamber 10 was pressed together with a hand press. When alginate was used, the assembled cell encapsulating ring chamber 10 was immersed in 10 mM BaCl2 solution for 30 seconds to crosslink the alginate. After three washes by immersion and shaking for 10 s in Ca2+ and Mg2+-free PBS, the loaded ring chamber was ready to be cultured in vitro. Prior to loading, sterilization was achieved by autoclaving the titanium rings 32 and washers 33, placing the oxygen generator/electrolyzer 18 in boiling water for at least 15 min, and immersing the semipermeable barrier layer 9 laminated membrane in 95% ethanol, 80% ethanol, and three times in PBS for at least 5 min in each. Sterilization was maintained by assembling the device in a laminar flow hood.
  • The [0109] cell encapsulating chamber 10 was oriented so that oxygen generation occurred below the bottom face of the ring immunoisolation chamber 10. The flask was then placed in an incubator and the oxygen generator 18 was connected to a power source 70 outside the incubator. Control experiments were carried out simultaneously with the oxygen generator 18 not connected to a power source 70.
  • The current was initially set at 11 μA/cm[0110] 2 corresponding to a maximum calculated tissue pO2 of 77 mm Hg, and the current density was doubled every two days to account for cell growth. Culture medium was changed every two days. After 4 and 9 days, both control cells 14 and cells 14 provided with in situ oxygen generation were removed, embedded in paraffin, cut into 5 μm sections, and stained with hematoxylin.
  • Appropriate to this initial feasibility study, five runs were carried out comprising of 18 sets of in vitro experiments. In each run, the imposed oxygen flux (or a specified pattern of changes with time) and the number of cells intended to be added to the cell encapsulating chamber [0111] 10 (from a single cell 14 suspension) were held constant. Within a single run, each set of experiments included cell encapsulating chambers 10 cultured for a specific time period. A single set included experiments with, and without, oxygen generation with stirred medium. In some cases, controls were also carried out with neither oxygen generation nor stirring. Some sets of experiments were carried out with alginate in the cell encapsulating chambers 10, some without. Digital images taken from 5 μm paraffin sections through the device are presented here for three of the five runs. In runs 1 (FIG. 7) and 2 (FIG. 8), I=45 μA, Ne=3.7×10−10 mol/cm2·s, and 2.3 or 2.5×106 cells 14 were intended to be added to the cell encapsulating chamber 10, respectively. In run 3 (FIG. 9), fewer cells 14 were intended to be added (0.35×106), and the low initial value of I=4.3 μA (Ne=0.35×10−10 mol/cm2·s) was increased by a factor of every {square root}{square root over (2)} every 24 hr so as to parallel the doubling time of about 2 days observed with βTC3 cells 14 in culture.
  • In runs 1 and 3, three [0112] cell encapsulating chambers 10 were processed for histology immediately after being loaded in order to examine the initial cell 14 distribution. In all devices, cells 14 were usually distributed continuously within a region centered roughly around the center of the cell encapsulating chamber 10, with a diameter of about 2 to 3.5 mm. In run 1, the cell 14 layer ranged from one to four cells 14 (about 10 to 40 mm) thick and was usually thinnest at the periphery and thickest close to the center. FIG. 7a is a representative section of the layers observed.
  • Histological section from [0113] cell encapsulating chambers 10 that underwent culture for time t=1 to 9 days displayed cell 14 layer thickness patterns qualitatively similar to those at t=0. The sections selected for presentation here met the criteria of being in focus, having no sectioning artifacts, and being representative of the thickest cell 14 layer observed in the cell encapsulating chamber 10.
  • In run 1 (FIG. 7), initial (t=0) tissue thickness L was about 40 μm, and all [0114] cells 14 were alive. After 2 days culture, L≈45 μm in the control cell encapsulating chamber 10, with many condensed nuclei characteristic of apoptotic cells. In the stirred experiments without and with O2 generation, L≈50 and 70 μm, respectively. Cell 14 viability was better with O2 generation than without. After 6 days, L increased only to about 50 m in the control cell encapsulating chamber 10, and there was substantial evidence of cell 14 death. With stirring and O2 generation at 6 days, L increased to 100 μm and virtually all cells 14 were viable.
  • Results from [0115] run 2 are summarized in FIG. 8. After 1 day of culture, the control device had a tissue thickness of 20 μm, with substantial loss of cell 14 viability, whereas with stirring and O2 generation L≈45 μm with much fewer dead cells 14. After 2 days in the control cell encapsulating chamber 10, L≈45 μm, but both the volume fraction and viability of cells 14 were very low. With stirring and without O2 generation, L≈65 μm, but there were many apoptotic cells indicative of low cell viability. With O2 generation, L≈60 μm, and virtually all cells were viable.
  • FIG. 9 shows the results from stirred experiments in [0116] run 3, in which cell 14 loading and initial oxygen flux were much lower than in runs 1 and 2. At t=2 days, tissue thickness (L≈30 μm) and viability were similar with or without O2 generation. At 3 days, L was still 25 μm without O2 generation, while it increased to 50 μm with O2 generation. At 9 days, most of cells 14 in the experiment without O2 generation were dead. With O2 generation, L≈70 μm, but there were apoptotic cells 14 and cell debris 14 near the semipermeable barrier layer 9 membrane adjacent to the oxygen generator/electrolyzer 18 where the oxygen partial pressure was highest. By 9 days and four doublings, Ne had increased to the relatively high value of 5.6×10−10 mol/cm2·s, thereby suggesting the possibility of oxygen toxicity to the cells 14 nearest the oxygen generator/electrolyzer 18 in this experiment. Up to this point, all of the experiments presented were carried out with alginate. FIGS. 9g and 9 h show results from experiments without alginate. Without O2 generation, thickness was similar to that with alginate (L≈25 μm) after 3 days (FIG. 9c). With O2 generation, L≈75 μm, higher than with alginate (FIG. 9d). It is possible that cross-linked alginate poses a restriction on cell growth.
  • In all of these experiments, the thickness of the [0117] cell 14 layer (and thus the growth rate of the cells 14), and their viability was highest in the stirred experiments with O2 generation and lowest in the unstirred static controls (in which oxygen transport was most limited). These results verify the hypothesis that oxygen transport limitations can limit the growth and viability of cells 14 in immunobarrier devices. Furthermore, they demonstrate that in situ oxygen generation leading to an imposed oxygen flux into the tissue compartment can have a beneficial effect on cell 14 growth and viability.
  • The theoretical model developed earlier was used to analyze oxygen transport in nine of the experiments. Estimated values of parameters used in the calculations are summarized in Table 1. Each experiment is designated by its figure number. Vmax was based upon the value of 1.5 μmol/min·10[0118] 9 cells 14 for a monolayer of βTC3 cells 14 and converted to a volume basis for cells with dc=12 μm to yield 2.76×10−8 mol/cm3·s. Permeability of the cells 14 was assumed to be the same as for islet tissue, 1.24×10−14 mol/cm·mm Hg·s. The values of V and Dα varied between experiments because of variation in the tissue volume fraction 1−ε. Table 1 also contains estimates of the dependent variables Le, Lm, PSe, PSm, and Pmin which were calculated from Equations (10), (11), (6), (5), and (12), respectively.
  • The experiments analyzed represent three groups: (1) those with high imposed oxygen flux (experiments 7a, 7d, and 7f), (2) those with low oxygen flux (9b and 9h), and (3) those without oxygen flux (7a, 7c, 9a, and 9g). In the first group, L[0119] e>L, and there is an efflux of unconsumed oxygen from the device, described by Equation (9). In the second group, Le<L, and there is an influx of oxygen into the cell encapsulating chamber 10 from both sides, hence, a minimum in the P profile develops. In the third group, oxygen is supplied only from the medium, so the maximum value of P occurs at y=0 and Pmin occurs at z=0.
  • Although the bench-top [0120] current controller 40 used in our in vitro experiments is large because it provides displays and controls, we have also constructed a miniaturized controller 40 (6.5×3.5×5.4 cm) that can be worn by a rat. Furthermore, microelectronic technology can be used to make a miniaturized implantable device with a size suitable for implantation. The current can be preset or controlled by telemetry. We have calculated that an oxygen generation rate sufficient to support a biohybrid artificial pancreas in a human could be powered by a an implantable battery 199 with 3000 mAh capacity which would require recharging every four weeks. Recharging could be achieved with a transcutaneous energy transfer (TET) system of the type developed to power implantable devices such as artificial hearts, defibrillators, and electrical stimulators. The power transmission requirements for biohybrid artificial pancreas is many orders of magnitude smaller than the capacity of currently available TET systems.
  • EXAMPLE 2
  • Islets isolated from Sprague-Dawley rats were used in another set of experiments. The loading density was 500 islets/[0121] cell encapsulating chamber 10. The current density, 100 μA/cm2, which corresponds to a maximum predicted pO2 of 300 mm Hg, was fixed at its initial value because islets do not proliferate. The result is shown in FIGS. 10a
    TABLE 1
    Values of important parameters in analyzed experiments
    Parameters Experiments
    Figure Number 7a 7a 7c 7d 7f 9a 9b 9g 9h
    Culture Time, t (d)  0  0 2 2 6 2  2 3 3
    Oxygen Flux, N   0a    3.7a 0 3.7 3.7 0    0.50 0 0.70
    (mol/cm2 · s × 1010)
    Tissue Thickness, L (μm)  40  40 50 70 100 30  25 25 75
    Tissue Volume Fraction, 1 − ε    0.52    0.52 0.53 0.69 0.60 0.74    0.78 0.76 0.70
    Oxygen Consumption Rate, V    1.4    1.4 1.5 1.9 1.7 2.0    2.1 2.1 1.9
    (mol/cm3 · s × 108)
    Oxygen Permeability, Dα    2.2    2.2 2.1 1.8 2.0 1.7    1.6 1.7 1.8
    (mol/cm · mmHg · s × 1014)
    Tissue Thickness Supportable  0 250 0 190 220 0  23 0 37
    from Electrolyzer, Le (μm)
    Tissue Thickness Supportable 140 140 140 110 130 110 100 100 110
    from Medium, Lm (μm)
    Interface P at x = L/2, PSe 116 313 108 340 356 115 144 120 115
    (mmHg)
    Interface P at x = −L/2, PSm 122 250 116 224 212 121  141b 124 116
    (mmHg)
    Minimum P, Pmin (mmHg)  141b 108
    # 10−8 mol/cm3 · s, Pc = 0.1 mm Hg, and Km = 0.44 mm Hg.
  • and [0122] 10 b. After 4 days of culturing, the tissue thickness in the control experiment was only half of the tissue thickness in the experiment using in situ oxygen generation.
  • Although the examples above have been described in terms of transplantation of islets of Langerhans, it is generally applicable to implantation of any type of [0123] cells 14 or tissues for therapeutic application. The invention concerns a novel approach to improve growth and maintenance of cells 14 and to oxygenating cell 14-compatible fluids.
  • Having thus described certain embodiments of the present invention, various alterations, modifications, and improvements will be obvious to those skilled in the art. Such variations, modifications and improvements are intended to be within the spirit and scope of the invention. The materials employed, as well as their shapes and dimensions, generally can vary. Accordingly, the foregoing description is by way of example only and is not intended to be limiting.[0124]

Claims (27)

What is claimed is:
1. A cell encapsulating device, comprising:
a semipermeable barrier layer, said semipermeable barrier layer defining a containment space for cells; and
an oxygen generator in close proximity to said semipermeable barrier layer, said oxygen generator supplying a source of oxygen for said cells.
2. The cell encapsulating device of claim 1, wherein said semipermeable barrier layer defines an immunoisolation chamber which immunosiolates said cells when the device is exposed to components of the immune system.
3. The cell encapsulating device of claim 2, wherein said immunoisolation chamber is a ring immunoisolation chamber comprising two semipermeable membranes sealed together by a ring seal.
4. The cell encapsulating device of claim 1, wherein said semipermeable barrier layer has a molecular weight cutoff of from about 50,000 daltons through about 300,000 daltons.
5. The cell encapsulating device of claim 1, further comprising cells, said cells selected from the group consisting of pancreas cells, hepatocytes, kidney cells, lung cells, neural cells, pituitary cells, parathyroid cells, thyroid cells, adrenal cells, proliferating cell lines, genetically modified cells, and combinations thereof.
6. The cell encapsulating device of claim 1, further comprising cells, said cells provided in amounts from about 106 cells per ml through about 109 cells per ml.
7. The cell encapsulating device of claim 1, further comprising cells and at least one bioactive molecule.
8. The cell encapsulating device of claim 7, wherein said bioactive molecules are selected from the group consisting of hormones, growth factors, cytokines, vascularizing agents, receptors, ligands, antibody fragments, fusion proteins, natural peptides, synthetic peptides, drugs, naked DNA, encapsulated DNA, antisense molecules, ribozymes, triple helix DNA, PNA molecules, and aptamers.
9. The cell encapsulating device of claim 1, wherein said oxygen generator electrolyzes water into oxygen and hydrogen.
10. The cell encapsulating device of claim 2, wherein the oxygen generator comprises a multilayer electrolyzer sheet.
11. The cell encapsulating device of claim 10, wherein the oxygen generator comprises an anode side and a cathode side, wherein the anode side is proximal to said semipermeable barrier layer, and wherein, when said device is implanted within a host body, said cathode side is proximal to the host body's cells and/or fluids
12. The cell encapsulating device of claim 11, further comprising a proton exchange membrane.
13. The cell encapsulating device of claim 10, wherein said multilayer electrolyzer sheet is adapted for press-fitted coupling to a ring immunoisolation chamber, said ring immunosiolation chamber comprising two semipermeable membranes sealed together by a ring seal.
14. The cell encapsulating device of claim 1, wherein said oxygen generator is in communication with an energy source.
15. The cell encapsulating device of claim 14, wherein said energy source is a battery.
16. The cell encapsulating device of claim 15, wherein said battery is rechargeable transcutaneously.
17. A method of maintaining cells in a body, comprising:
providing an immunoisolation chamber containing said cells;
generating oxygen in situ in said body; and
providing said oxygen to said immunoisolation chamber.
18. The method of claim 17, wherein said body lacks a function and said cells provide said function.
19. The method of claim 18, wherein said cells are genetically modified to provide said function.
20. The method of claim 18, further comprising providing said cells to a body with a condition selected from the group consisting of diabetes, hemophilia B, dwarfism, anemia, kidney failure, hepatic failure, familial hypercholesterolemia, immunodeficiency disorders, pitutitary disorders, and central nervous system disorders.
21. The method of claim 17, further comprising providing at least one bioactive molecule in said immunoisolation chamber.
22. The method of claim 17, wherein said immunoisolation chamber and said cells comprise a bioartificial organ.
23. The method of claim 17, wherein said oxygen generator is in communication with an energy source.
24. The method of claim 23, wherein said energy source is a battery.
25. The method of 25, further comprising the step of recharging said battery transcutaneously.
26. The method of claim 23, wherein said energy source is a transcutaneous energy transfer device.
27. The method of claim 17, wherein said oxygen generator, further comprises a cathode side and an anode side, and wherein said oxygen generator electrochemically transfers oxygen from the cathode side to the anode side of the oxygen generator substantially without the without generation of hydrogen.
US10/040,278 1998-07-17 2001-10-24 Method and apparatus for delivering oxygen to cells Abandoned US20030087427A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/040,278 US20030087427A1 (en) 1998-07-17 2001-10-24 Method and apparatus for delivering oxygen to cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9314798P 1998-07-17 1998-07-17
US09/356,079 US6368592B1 (en) 1998-07-17 1999-07-16 Method of delivering oxygen to cells by electrolyzing water
US10/040,278 US20030087427A1 (en) 1998-07-17 2001-10-24 Method and apparatus for delivering oxygen to cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/356,079 Continuation US6368592B1 (en) 1998-07-17 1999-07-16 Method of delivering oxygen to cells by electrolyzing water

Publications (1)

Publication Number Publication Date
US20030087427A1 true US20030087427A1 (en) 2003-05-08

Family

ID=26787195

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/356,079 Expired - Fee Related US6368592B1 (en) 1998-07-17 1999-07-16 Method of delivering oxygen to cells by electrolyzing water
US10/040,278 Abandoned US20030087427A1 (en) 1998-07-17 2001-10-24 Method and apparatus for delivering oxygen to cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/356,079 Expired - Fee Related US6368592B1 (en) 1998-07-17 1999-07-16 Method of delivering oxygen to cells by electrolyzing water

Country Status (1)

Country Link
US (2) US6368592B1 (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030056805A1 (en) * 2001-09-14 2003-03-27 Osao Sumita Electrolytic cell for producing charged anode water suitable for surface cleaning or treatment, and method for producing the same and use of the same
US20040133188A1 (en) * 2000-01-12 2004-07-08 Pnina Vardi Implantable device
US20050121334A1 (en) * 2001-12-05 2005-06-09 Osao Sumita Method and apparatus for producting negative and positive oxidative reductive potential (orp) water
US20050136092A1 (en) * 2000-01-01 2005-06-23 Beta-O2 Technologies Ltd. Implantable device
US20050139808A1 (en) * 2003-12-30 2005-06-30 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and process for producing same
US20050196462A1 (en) * 2003-12-30 2005-09-08 Oculus Innovative Sciences, Inc. Topical formulation containing oxidative reductive potential water solution and method for using same
WO2006006166A2 (en) 2004-07-14 2006-01-19 Glusense, Ltd. Implantable power sources and sensors
US20060235350A1 (en) * 2005-03-23 2006-10-19 Oculus Innovative Sciences, Inc. Method of treating skin ulcers using oxidative reductive potential water solution
US20070173755A1 (en) * 2006-01-20 2007-07-26 Oculus Innovative Sciences, Inc. Methods of treating or preventing peritonitis with oxidative reductive potential water solution
US20090012502A1 (en) * 2000-01-12 2009-01-08 Beta-O2 Technologies Ltd. Oxygen Supply for Cell Transplant and Vascularization
US20100047311A1 (en) * 2006-11-22 2010-02-25 Beta O2 Technologies Ltd. Protecting algae from body fluids
US20100130916A1 (en) * 2008-11-26 2010-05-27 Yaki Stern Apparatus for transportation of oxygen to implanted cells
US20100160749A1 (en) * 2008-12-24 2010-06-24 Glusense Ltd. Implantable optical glucose sensing
WO2010091074A2 (en) * 2009-02-03 2010-08-12 Purdue Research Foundation Method and apparatus for improving local hypoxicity for enhanced therapy
US20100291610A1 (en) * 2006-03-08 2010-11-18 Yael Porat Regulating Stem Cells
US20100312165A1 (en) * 2007-09-07 2010-12-09 Beta 02 Technologies Ltd. Air gap for supporting cells
US20100330547A1 (en) * 2009-06-18 2010-12-30 Tempelman Linda A Perfusing an organ with an in situ generated gas
US20110165219A1 (en) * 2008-09-17 2011-07-07 Beta O2 Technologies Ltd. Optimization of alginate encapsulation of islets for transplantation
US20110300191A1 (en) * 2010-06-07 2011-12-08 Beta-O2 Technologies Ltd. Multiple-layer immune barrier for donor cells
US8685724B2 (en) 2004-06-01 2014-04-01 Kwalata Trading Limited In vitro techniques for use with stem cells
WO2015048184A1 (en) * 2013-09-24 2015-04-02 Giner, Inc. System for gas treatment of a cell implant
US9037205B2 (en) 2011-06-30 2015-05-19 Glusense, Ltd Implantable optical glucose sensing
US9168318B2 (en) 2003-12-30 2015-10-27 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and methods of using the same
US9357764B2 (en) 2009-06-18 2016-06-07 Giner, Inc. System for fluid perfusion of biological matter comprising tissue
US9498548B2 (en) 2005-05-02 2016-11-22 Oculus Innovative Sciences, Inc. Method of using oxidative reductive potential water solution in dental applications
WO2017137842A1 (en) * 2016-02-08 2017-08-17 Beta-O2 Technologies Ltd. Systems and methods for providing oxygen to transplanted cells
US20170304528A1 (en) * 2016-04-22 2017-10-26 DeepScience Ltd. Implantable medical device for delivering cells
WO2018085714A1 (en) 2016-11-03 2018-05-11 The Arizona Board Of Regents On Behalf Of The University Of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
WO2018093956A1 (en) 2016-11-15 2018-05-24 Giner, Inc. Percutaneous gas diffusion device suitable for use with a subcutaneous implant
WO2018093940A1 (en) 2016-11-15 2018-05-24 Giner, Inc. Self-regulating electrolytic gas generator and implant system comprising the same
WO2018102077A2 (en) 2016-11-03 2018-06-07 The Arizona Board Of Regents On Behalf Of The University Of Arizona Methods, systems, and implantable devices for enhancing blood glucose regulation
WO2018144098A1 (en) 2016-11-03 2018-08-09 The Arizona Board Of Regents On Behalf Of The University Of Arizona Stacked tissue encapsulation device systems with or without oxygen delivery
WO2018144099A1 (en) 2016-11-03 2018-08-09 The Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for real-time assessment of cells in encapsulation devices pre-and post-transplantation
WO2018204867A1 (en) * 2017-05-04 2018-11-08 Giner, Inc. Robust, implantable gas delivery device and methods, systems and devices including same
WO2019021528A1 (en) * 2017-07-28 2019-01-31 株式会社日立製作所 Oxygen supply mechanism
WO2019106996A1 (en) 2017-11-30 2019-06-06 株式会社日立製作所 Immunoisolation device
US10342825B2 (en) 2009-06-15 2019-07-09 Sonoma Pharmaceuticals, Inc. Solution containing hypochlorous acid and methods of using same
WO2019222704A1 (en) * 2018-05-17 2019-11-21 Giner Life Sciences, Inc. Electrolytic gas generator with combined lead and gas port terminals
WO2019222273A1 (en) * 2018-05-14 2019-11-21 Giner Life Sciences, Inc. System and method for controlling oxygen delivery to implanted cells
US20200030598A1 (en) * 2018-07-26 2020-01-30 University Of North Texas Hydrogel-based biomedical devices for therapeutic hydrogen treatment of skin and tissues and methods of using them
US10575765B2 (en) 2014-10-13 2020-03-03 Glusense Ltd. Analyte-sensing device
CN111819308A (en) * 2017-12-22 2020-10-23 格勒诺布尔-阿尔卑斯大学 Implantable device for generating hydrogen
US10871487B2 (en) 2016-04-20 2020-12-22 Glusense Ltd. FRET-based glucose-detection molecules
US20220134074A1 (en) * 2020-10-30 2022-05-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for encapsulation devices for housing cells and agents
WO2023205302A3 (en) * 2022-04-20 2023-12-07 Northwestern University Oxygen generation system and applications thereof

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7165551B2 (en) * 1998-02-19 2007-01-23 Curon Medical, Inc. Apparatus to detect and treat aberrant myoelectric activity
US9119705B2 (en) 1998-06-08 2015-09-01 Thermotek, Inc. Method and system for thermal and compression therapy relative to the prevention of deep vein thrombosis
US6500319B2 (en) * 2001-04-05 2002-12-31 Giner Electrochemical Systems, Llc Proton exchange membrane (PEM) electrochemical cell having an integral, electrically-conductive, compression pad
CA2476576A1 (en) * 2002-02-22 2003-09-04 Aqua Innovations, Inc. Microbubbles of oxygen
US20040106951A1 (en) * 2002-11-22 2004-06-03 Edman Carl Frederick Use of electric fields to minimize rejection of implanted devices and materials
US8100956B2 (en) * 2006-05-09 2012-01-24 Thermotek, Inc. Method of and system for thermally augmented wound care oxygenation
US8778005B2 (en) 2003-07-18 2014-07-15 Thermotek, Inc. Method and system for thermal and compression therapy relative to the prevention of deep vein thrombosis
US8128672B2 (en) 2006-05-09 2012-03-06 Thermotek, Inc. Wound care method and system with one or both of vacuum-light therapy and thermally augmented oxygenation
US8574278B2 (en) 2006-05-09 2013-11-05 Thermotek, Inc. Wound care method and system with one or both of vacuum-light therapy and thermally augmented oxygenation
DE602004032027D1 (en) 2003-07-18 2011-05-12 Thermotek Inc THERMAL SYSTEM FOR A BLANKET
USD679023S1 (en) 2004-07-19 2013-03-26 Thermotek, Inc. Foot wrap
US10016583B2 (en) 2013-03-11 2018-07-10 Thermotek, Inc. Wound care and infusion method and system utilizing a thermally-treated therapeutic agent
US10765785B2 (en) 2004-07-19 2020-09-08 Thermotek, Inc. Wound care and infusion method and system utilizing a therapeutic agent
US20060034053A1 (en) * 2004-08-12 2006-02-16 Thermotek, Inc. Thermal control system for rack mounting
GB0504445D0 (en) 2005-03-03 2005-04-06 Univ Cambridge Tech Oxygen generation apparatus and method
US20070141405A1 (en) * 2005-11-16 2007-06-21 General Motors Corporation Method of making a membrane electrode assembly comprising a vapor barrier layer, a gas diffusion layer, or both
US9017717B2 (en) * 2006-01-16 2015-04-28 Peach Technologies Llc Bandage for facilitating transdermal respiration and healing
US20100210982A1 (en) * 2006-04-11 2010-08-19 Niran Balachandran Method And System For Providing Segmental Gradient Compression
USD664260S1 (en) 2007-04-10 2012-07-24 Thermotek, Inc. Calf wrap
US8083821B2 (en) * 2007-04-13 2011-12-27 Giner, Inc. System for modifying the atmosphere within an enclosed space and incubator system including the same
US8758419B1 (en) 2008-01-31 2014-06-24 Thermotek, Inc. Contact cooler for skin cooling applications
US8986725B2 (en) 2009-04-15 2015-03-24 University Of Miami Silicone-peroxide compositions for long-term, controlled oxygen release
US8594806B2 (en) 2010-04-30 2013-11-26 Cyberonics, Inc. Recharging and communication lead for an implantable device
US10512587B2 (en) 2011-07-27 2019-12-24 Thermotek, Inc. Method and apparatus for scalp thermal treatment
US10149927B2 (en) 2012-04-24 2018-12-11 Thermotek, Inc. Method and system for therapeutic use of ultra-violet light
US9343923B2 (en) 2012-08-23 2016-05-17 Cyberonics, Inc. Implantable medical device with backscatter signal based communication
US9935498B2 (en) 2012-09-25 2018-04-03 Cyberonics, Inc. Communication efficiency with an implantable medical device using a circulator and a backscatter signal
JP6335904B2 (en) 2012-10-05 2018-05-30 ユニバーシティ オブ サザン カリフォルニア Oxygen supply
US10300180B1 (en) 2013-03-11 2019-05-28 Thermotek, Inc. Wound care and infusion method and system utilizing a therapeutic agent
EP3068481B1 (en) 2013-11-11 2020-01-01 Thermotek, Inc. System for wound care
EP4197491A1 (en) * 2021-12-20 2023-06-21 Technische Universität Dresden Modular implantable device for the macroencapsulation of cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6315721B2 (en) * 1997-02-26 2001-11-13 Alfred E. Mann Foundation For Scientific Research System of implantable devices for monitoring and/or affecting body parameters

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
DE3915251A1 (en) 1989-05-10 1990-11-22 Annemarie Schloegl Ges M B H IMPLANTABLE DEVICE FOR DISPENSING DISPOSAL OF MEDICINES IN HUMAN BODIES
TW279133B (en) 1990-12-13 1996-06-21 Elan Med Tech
US5391164A (en) 1991-05-03 1995-02-21 Giampapa; Vincent C. Subcutaneous implantable multiple-agent delivery system
US5201728A (en) 1991-05-03 1993-04-13 Giampapa Vincent C Subcutaneous implantable multiple-agent delivery system
US5635039A (en) 1993-07-13 1997-06-03 Lynntech, Inc. Membrane with internal passages to permit fluid flow and an electrochemical cell containing the same
JPH0858897A (en) 1994-08-12 1996-03-05 Japan Storage Battery Co Ltd Fluid supply device
US5575770A (en) 1995-04-05 1996-11-19 Therex Corporation Implantable drug infusion system with safe bolus capability
US5578022A (en) 1995-04-12 1996-11-26 Scherson; Daniel A. Oxygen producing bandage and method
US5788682A (en) 1995-04-28 1998-08-04 Maget; Henri J.R. Apparatus and method for controlling oxygen concentration in the vicinity of a wound
IE77523B1 (en) 1995-09-11 1997-12-17 Elan Med Tech Medicament delivery device
US5945766A (en) 1996-01-18 1999-08-31 Amotron Co., Ltd. Coreless-type BLDC motor and method of producing stator assembly having axial vibration attenuation arrangement
US5899381A (en) 1997-02-21 1999-05-04 Ceramatec, Inc. Electrochemical device for delivery of volatile substances
US5928194A (en) 1997-04-07 1999-07-27 Maget; Henri J. R. Self-contained liquid microdispenser
US5961796A (en) 1997-06-03 1999-10-05 Lynntech, Inc. Bipolar membranes with fluid distribution passages
US5938640A (en) 1997-06-04 1999-08-17 M&R Consulting Services Two-part fluid dispenser
US5914199A (en) 1997-08-04 1999-06-22 Lockheed Martin Tactical Defense Systems, Inc. Pressure equalized fuel cell stack
US5951538A (en) 1997-08-07 1999-09-14 Ceramatec, Inc. Gas generating device for delivering beneficial agents to a body cavity
US5921251A (en) 1997-08-07 1999-07-13 Ceramatec, Inc. Brush that delivers beneficial agents
US5971722A (en) 1997-09-05 1999-10-26 Baxter International Inc Electrochemical syringe pump having a sealed storage reservoir for a charge transfer medium
US5957895A (en) 1998-02-20 1999-09-28 Becton Dickinson And Company Low-profile automatic injection device with self-emptying reservoir

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6315721B2 (en) * 1997-02-26 2001-11-13 Alfred E. Mann Foundation For Scientific Research System of implantable devices for monitoring and/or affecting body parameters

Cited By (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050136092A1 (en) * 2000-01-01 2005-06-23 Beta-O2 Technologies Ltd. Implantable device
US7892222B2 (en) 2000-01-12 2011-02-22 Beta O 2 Technologies Ltd. Implantable device
US20040133188A1 (en) * 2000-01-12 2004-07-08 Pnina Vardi Implantable device
US8444630B2 (en) 2000-01-12 2013-05-21 Beta-O2 Technologies Ltd. Oxygen supply for cell transplant and vascularization
US20090012502A1 (en) * 2000-01-12 2009-01-08 Beta-O2 Technologies Ltd. Oxygen Supply for Cell Transplant and Vascularization
US20060272954A1 (en) * 2001-09-14 2006-12-07 Oculus Innovative Sciences, Inc. Electrolytic cell for producing charger anode water suitable for surface cleaning or treatment, and method for producing the same and use of the same
US20030056805A1 (en) * 2001-09-14 2003-03-27 Osao Sumita Electrolytic cell for producing charged anode water suitable for surface cleaning or treatment, and method for producing the same and use of the same
US8062500B2 (en) 2001-12-05 2011-11-22 Oculus Innovative Sciences, Inc. Method and apparatus for producing negative and positive oxidative reductive potential (ORP) water
US20050121334A1 (en) * 2001-12-05 2005-06-09 Osao Sumita Method and apparatus for producting negative and positive oxidative reductive potential (orp) water
US10016455B2 (en) 2003-12-30 2018-07-10 Sonoma Pharmaceuticals, Inc. Method of preventing or treating influenza with oxidative reductive potential water solution
US9168318B2 (en) 2003-12-30 2015-10-27 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and methods of using the same
US9642876B2 (en) 2003-12-30 2017-05-09 Sonoma Pharmaceuticals, Inc. Method of preventing or treating sinusitis with oxidative reductive potential water solution
US20050196462A1 (en) * 2003-12-30 2005-09-08 Oculus Innovative Sciences, Inc. Topical formulation containing oxidative reductive potential water solution and method for using same
US20050139808A1 (en) * 2003-12-30 2005-06-30 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and process for producing same
US8685724B2 (en) 2004-06-01 2014-04-01 Kwalata Trading Limited In vitro techniques for use with stem cells
US20080319287A1 (en) * 2004-07-14 2008-12-25 Yossi Gross Implantable Power Sources and Sensors
EP2532356A1 (en) 2004-07-14 2012-12-12 Glusense Ltd. Implantable power sources and sensors
US7951357B2 (en) 2004-07-14 2011-05-31 Glusense Ltd. Implantable power sources and sensors
WO2006006166A2 (en) 2004-07-14 2006-01-19 Glusense, Ltd. Implantable power sources and sensors
US8840873B2 (en) 2005-03-23 2014-09-23 Oculus Innovative Sciences, Inc. Method of treating second and third degree burns using oxidative reductive potential water solution
US8323252B2 (en) 2005-03-23 2012-12-04 Oculus Innovative Sciences, Inc. Method of treating skin ulcers using oxidative reductive potential water solution
US20060241546A1 (en) * 2005-03-23 2006-10-26 Oculus Innovative Sciences, Inc. Method of treating second and third degree burns using oxidative reductive potential water solution
US20060235350A1 (en) * 2005-03-23 2006-10-19 Oculus Innovative Sciences, Inc. Method of treating skin ulcers using oxidative reductive potential water solution
US9498548B2 (en) 2005-05-02 2016-11-22 Oculus Innovative Sciences, Inc. Method of using oxidative reductive potential water solution in dental applications
US20070173755A1 (en) * 2006-01-20 2007-07-26 Oculus Innovative Sciences, Inc. Methods of treating or preventing peritonitis with oxidative reductive potential water solution
US8147444B2 (en) 2006-01-20 2012-04-03 Oculus Innovative Sciences, Inc. Methods of treating or preventing peritonitis with oxidative reductive potential water solution
US9072726B2 (en) 2006-01-20 2015-07-07 Oculus Innovative Sciences, Inc. Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
US9782434B2 (en) 2006-01-20 2017-10-10 Sonoma Pharmaceuticals, Inc. Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
US8834445B2 (en) 2006-01-20 2014-09-16 Oculus Innovative Sciences, Inc. Methods of treating or preventing peritonitis with oxidative reductive potential water solution
US20070196434A1 (en) * 2006-01-20 2007-08-23 Oculus Innovative Sciences, Inc. Methods of preventing or treating sinusitis with oxidative reductive potential water solution
US20100092399A1 (en) * 2006-01-20 2010-04-15 Oculus Innovative Sciences, Inc. Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
US9234173B2 (en) 2006-03-08 2016-01-12 Kwalata Trading Ltd. Regulating stem cells
US20100291610A1 (en) * 2006-03-08 2010-11-18 Yael Porat Regulating Stem Cells
US8541232B2 (en) 2006-03-08 2013-09-24 Kwalata Trading Limited Composition comprising a progenitor/precursor cell population
US10358629B2 (en) 2006-03-08 2019-07-23 Kwalata Trading Limited Regulating stem cells
US20100047311A1 (en) * 2006-11-22 2010-02-25 Beta O2 Technologies Ltd. Protecting algae from body fluids
US20100312165A1 (en) * 2007-09-07 2010-12-09 Beta 02 Technologies Ltd. Air gap for supporting cells
US8821431B2 (en) 2007-09-07 2014-09-02 Beta O2 Technologies Ltd. Air gap for supporting cells
US9463083B2 (en) 2007-09-07 2016-10-11 Beta-O2 Technologies Ltd. Air gap for supporting cells
US9540630B2 (en) 2008-09-17 2017-01-10 Beta O2 Technologies Ltd. Optimization of alginate encapsulation of islets for transplantation
US20110165219A1 (en) * 2008-09-17 2011-07-07 Beta O2 Technologies Ltd. Optimization of alginate encapsulation of islets for transplantation
US8043271B2 (en) 2008-11-26 2011-10-25 Beta 02 Technologies Ltd. Apparatus for transportation of oxygen to implanted cells
AU2009321193B2 (en) * 2008-11-26 2015-07-09 Beta O2 Technologies Ltd. Apparatus for transportation of oxygen to implanted cells
CN102292049A (en) * 2008-11-26 2011-12-21 贝塔02技术有限公司 Apparatus for transportation of oxygen to implanted cells
US20100130916A1 (en) * 2008-11-26 2010-05-27 Yaki Stern Apparatus for transportation of oxygen to implanted cells
WO2010061387A3 (en) * 2008-11-26 2010-08-12 Beta O2 Technologies Ltd. Apparatus for transportation of oxygen to implanted cells
US20100160749A1 (en) * 2008-12-24 2010-06-24 Glusense Ltd. Implantable optical glucose sensing
WO2010091074A3 (en) * 2009-02-03 2010-11-25 Purdue Research Foundation Method and apparatus for improving local hypoxicity for enhanced therapy
US9005187B2 (en) 2009-02-03 2015-04-14 Purdue Research Foundation Method and apparatus for improving local hypoxicity for enhanced therapy
WO2010091074A2 (en) * 2009-02-03 2010-08-12 Purdue Research Foundation Method and apparatus for improving local hypoxicity for enhanced therapy
US10342825B2 (en) 2009-06-15 2019-07-09 Sonoma Pharmaceuticals, Inc. Solution containing hypochlorous acid and methods of using same
US9357764B2 (en) 2009-06-18 2016-06-07 Giner, Inc. System for fluid perfusion of biological matter comprising tissue
US20100330547A1 (en) * 2009-06-18 2010-12-30 Tempelman Linda A Perfusing an organ with an in situ generated gas
US10091985B2 (en) * 2009-06-18 2018-10-09 Giner, Inc. Perfusing an organ with an in situ generated gas
US9446168B2 (en) * 2010-06-07 2016-09-20 Beta-O2 Technologies Ltd. Multiple-layer immune barrier for donor cells
US20110300191A1 (en) * 2010-06-07 2011-12-08 Beta-O2 Technologies Ltd. Multiple-layer immune barrier for donor cells
US9037205B2 (en) 2011-06-30 2015-05-19 Glusense, Ltd Implantable optical glucose sensing
EP3049020A4 (en) * 2013-09-24 2017-07-26 Giner, Inc. System for gas treatment of a cell implant
JP2016530980A (en) * 2013-09-24 2016-10-06 ガイナー,インク. System for gas treatment of cellular implants
US10231817B2 (en) 2013-09-24 2019-03-19 Giner Life Sciences, Inc. System for gas treatment of a cell implant
US11701215B2 (en) 2013-09-24 2023-07-18 Giner, Inc. System for gas treatment of a cell implant
KR102323291B1 (en) 2013-09-24 2021-11-05 기너 라이프 사이언시즈, 인크. System for gas treatment of a cell implant
AU2014326794B2 (en) * 2013-09-24 2019-03-21 Giner, Inc. System for gas treatment of a cell implant
KR20160060115A (en) * 2013-09-24 2016-05-27 기너 아이엔씨. System for gas treatment of a cell implant
CN105792775A (en) * 2013-09-24 2016-07-20 杜松子酒有限公司 System for gas treatment of a cell implant
WO2015048184A1 (en) * 2013-09-24 2015-04-02 Giner, Inc. System for gas treatment of a cell implant
US10575765B2 (en) 2014-10-13 2020-03-03 Glusense Ltd. Analyte-sensing device
JP2019503828A (en) * 2016-02-08 2019-02-14 ベータ−オーツー・テクノロジーズ・リミテッドBeta−O2 Technologies Ltd. System and method for providing oxygen to transplanted cells
WO2017137842A1 (en) * 2016-02-08 2017-08-17 Beta-O2 Technologies Ltd. Systems and methods for providing oxygen to transplanted cells
CN108882697A (en) * 2016-02-08 2018-11-23 贝塔O2技术有限公司 The system and method for oxygen are provided to transplanted cells
US10871487B2 (en) 2016-04-20 2020-12-22 Glusense Ltd. FRET-based glucose-detection molecules
US20170304528A1 (en) * 2016-04-22 2017-10-26 DeepScience Ltd. Implantable medical device for delivering cells
AU2017396754B2 (en) * 2016-11-03 2021-08-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for real-time assessment of cells in encapsulation devices pre-and post-transplantation
EP3534793A4 (en) * 2016-11-03 2020-05-27 Arizona Board of Regents on behalf of the University of Arizona Methods, systems, and implantable devices for enhancing blood glucose regulation
AU2021204321B2 (en) * 2016-11-03 2023-02-02 Arizona Board Of Regents On Behalf Of The University Of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
US11446133B2 (en) 2016-11-03 2022-09-20 Arizona Board Of Regents On Behalf Of The University Of Arizona Stacked tissue encapsulation device systems with or without oxygen delivery
KR102436392B1 (en) * 2016-11-03 2022-08-24 아리조나 보드 오브 리전츠 온 비해프 오브 더 유니버시티 오브 아리조나 Methods, systems and implantable devices for enhancing glycemic control
WO2018144099A1 (en) 2016-11-03 2018-08-09 The Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for real-time assessment of cells in encapsulation devices pre-and post-transplantation
CN110121312A (en) * 2016-11-03 2019-08-13 代表亚利桑那大学的亚利桑那董事会 With or without the system of the containment device with oxygen sensor of external source oxygen conveying
CN110139605A (en) * 2016-11-03 2019-08-16 代表亚利桑那大学的亚利桑那董事会 For improving the method, system and implantable device of blood glucose-control
CN110267526B (en) * 2016-11-03 2022-06-24 代表亚利桑那大学的亚利桑那董事会 Method and system for real-time assessment of cells in a packaged device before and after transplantation
KR20190104140A (en) * 2016-11-03 2019-09-06 아리조나 보드 오브 리전츠 온 비해프 오브 더 유니버시티 오브 아리조나 Methods, Systems, and Implant Devices for Enhancing Glucose Control
CN110267526A (en) * 2016-11-03 2019-09-20 代表亚利桑那大学的亚利桑那董事会 Method and system for cell in the rear containment device of assessment in real time before transplantation
US11723558B2 (en) 2016-11-03 2023-08-15 Arizona Board Of Regents On Behalf Of The University Of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
AU2017355528B2 (en) * 2016-11-03 2021-04-01 Arizona Board Of Regents On Behalf Of The University Of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
WO2018102077A2 (en) 2016-11-03 2018-06-07 The Arizona Board Of Regents On Behalf Of The University Of Arizona Methods, systems, and implantable devices for enhancing blood glucose regulation
US11746318B2 (en) 2016-11-03 2023-09-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for real-time assessment of cells in encapsulation devices pre-and post-transplantation
EP3534692A4 (en) * 2016-11-03 2020-06-17 Arizona Board of Regents on behalf of the University of Arizona Methods and systems for real-time assessment of cells in encapsulation devices pre-and post-transplantation
WO2018144098A1 (en) 2016-11-03 2018-08-09 The Arizona Board Of Regents On Behalf Of The University Of Arizona Stacked tissue encapsulation device systems with or without oxygen delivery
WO2018085714A1 (en) 2016-11-03 2018-05-11 The Arizona Board Of Regents On Behalf Of The University Of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
EP3534834A4 (en) * 2016-11-03 2020-05-13 Arizona Board of Regents on behalf of the University of Arizona Encapsulation device systems with oxygen sensors with or without exogenous oxygen delivery
WO2018093940A1 (en) 2016-11-15 2018-05-24 Giner, Inc. Self-regulating electrolytic gas generator and implant system comprising the same
EP3541450A4 (en) * 2016-11-15 2020-05-27 Giner Life Sciences, Inc. Self-regulating electrolytic gas generator and implant system comprising the same
US10557691B2 (en) 2016-11-15 2020-02-11 Giner Life Sciences, Inc. Self-regulating electrolytic gas generator and implant system comprising the same
CN110139683A (en) * 2016-11-15 2019-08-16 吉纳生命科学公司 Be self-regulated electrolytic gas generator and the implant system including it
EP3541449A4 (en) * 2016-11-15 2020-04-29 Giner Life Sciences, Inc. Percutaneous gas diffusion device suitable for use with a subcutaneous implant
JP2019534112A (en) * 2016-11-15 2019-11-28 ガイナー ライフ サイエンシズ,インク. Self-regulating electrolytic gas generator and implant system including the same
JP7044268B2 (en) 2016-11-15 2022-03-30 ガイナー ライフ サイエンシズ,インク. Self-regulating electrolytic gas generator and implant system with it
US11033666B2 (en) 2016-11-15 2021-06-15 Giner Life Sciences, Inc. Percutaneous gas diffusion device suitable for use with a subcutaneous implant
WO2018093956A1 (en) 2016-11-15 2018-05-24 Giner, Inc. Percutaneous gas diffusion device suitable for use with a subcutaneous implant
US20180318566A1 (en) * 2017-05-04 2018-11-08 Giner, Inc. Robust, implantable gas delivery device and methods, systems and devices including same
JP7199632B2 (en) 2017-05-04 2023-01-06 ガイナー,インク. Robust Implantable Gas Delivery Device and Methods, Systems and Devices Containing The Same
US11642501B2 (en) * 2017-05-04 2023-05-09 Giner, Inc. Robust, implantable gas delivery device and methods, systems and devices including same
JP2020518397A (en) * 2017-05-04 2020-06-25 ガイナー ライフ サイエンシズ,インク. Robust implantable gas delivery device and methods, systems and devices including same
WO2018204867A1 (en) * 2017-05-04 2018-11-08 Giner, Inc. Robust, implantable gas delivery device and methods, systems and devices including same
WO2019021528A1 (en) * 2017-07-28 2019-01-31 株式会社日立製作所 Oxygen supply mechanism
WO2019106996A1 (en) 2017-11-30 2019-06-06 株式会社日立製作所 Immunoisolation device
CN111819308A (en) * 2017-12-22 2020-10-23 格勒诺布尔-阿尔卑斯大学 Implantable device for generating hydrogen
CN113164721A (en) * 2018-05-14 2021-07-23 吉纳生命科学公司 System and method for controlling oxygen delivery to implanted cells
WO2019222273A1 (en) * 2018-05-14 2019-11-21 Giner Life Sciences, Inc. System and method for controlling oxygen delivery to implanted cells
WO2019222704A1 (en) * 2018-05-17 2019-11-21 Giner Life Sciences, Inc. Electrolytic gas generator with combined lead and gas port terminals
US11773496B2 (en) 2018-05-17 2023-10-03 Giner, Inc. Combined electrical lead and gas port terminals and electrolytic gas generator comprising same
US20200030598A1 (en) * 2018-07-26 2020-01-30 University Of North Texas Hydrogel-based biomedical devices for therapeutic hydrogen treatment of skin and tissues and methods of using them
US20220134074A1 (en) * 2020-10-30 2022-05-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and systems for encapsulation devices for housing cells and agents
WO2023205302A3 (en) * 2022-04-20 2023-12-07 Northwestern University Oxygen generation system and applications thereof

Also Published As

Publication number Publication date
US6368592B1 (en) 2002-04-09

Similar Documents

Publication Publication Date Title
US6368592B1 (en) Method of delivering oxygen to cells by electrolyzing water
Wu et al. In situ electrochemical oxygen generation with an immunoisolation device
JP6749239B2 (en) Cell implant gas treatment system
CN110139683B (en) Self-regulating electrolytic gas generator and implant system including the same
Drake et al. A tissue implantable fuel cell power supply
US11033666B2 (en) Percutaneous gas diffusion device suitable for use with a subcutaneous implant
US8012500B2 (en) Implantable device
Amiel et al. Renal therapy using tissue-engineered constructs and gene delivery
CA3100199C (en) System and method for controlling oxygen delivery to implanted cells
EP0577719B1 (en) Artificial pancreatic perfusion device with reseedable matrix
JP2000178180A (en) Material for forming neovascular bed for use in making capillary-rich tissue in vivo
Galletti Bioartificial organs
CN112587730B (en) Composite cell scaffold and preparation method thereof
Kerzenmacher Biofuel cells as sustainable power sources for implantable systems
WO2023205302A2 (en) Oxygen generation system and applications thereof
EP4197491A1 (en) Modular implantable device for the macroencapsulation of cells
CN214388359U (en) Composite cell scaffold
US20220411734A1 (en) Bioreactor system for tissue engineering
Lopes et al. Development of an all-human vascularized model of pancreatic islets on-chip: towards personalized medicine for type 1 diabetes
Kang et al. MEMS oxygen transport device for islet transplantation in the subcutaneous site
Kerzenmacher Abiotic (nonenzymatic) implantable biofuel cells
Do Glucose energy harvester for self-powering of remote distributed bioanalytical microsystems
CN117580545A (en) Matrix for containing pancreatic cells and improved artificial pancreatic device
CN112587730A (en) Composite cell scaffold and preparation method thereof
Javor Biofuel cell operating on activated THP-1 cells

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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