The present invention relates to implantable cell encapsulation systems, and scaffold sub-components thereof, as well as methods of manufacturing the scaffolds and implantable cell encapsulation systems, and methods of using the same.
Cell-based therapies are attractive treatments for a variety of diseases, such as diabetes (Shapiro et al., “Clinical Pancreatic Islet Transplantation,” Nat. Rev. Endocrinol. 13:268-277 (2017); Farina et al., “Cell Encapsulation: Overcoming Barriers in Cell Transplantation in Diabetes and Beyond,” Adv. Drug Deliv. Rev. 139:92-115 (2019)), liver diseases (Yu et al., “Cell Therapies for Liver Diseases,” Liver Transplantation 18:9-21 (2012)), and hemophilia (Ohmori et al., “New Approaches to Gene and Cell Therapy for Hemophilia,” J. Thromb. Haemost. 13 Suppl. 1:S133-142 (2015); Roth et al., “Nonviral Transfer of the Gene Encoding Coagulation Factor VIII in Patients with Severe Hemophilia A,” New Engl. J. Med. 344:1735-1742 (2001)). In particular, the delivery of islets (or stem cell-derived β-cells) represents a promising therapy for type 1 diabetes (T1D). Islet transplantation via the injection of isolated islets into the liver portal vein or onto the omentum has shown the potential to normalize glycemic control without exogenous insulin in clinical trials, but life-long recipient immunosuppression is required with this procedure (Shapiro et al., “International Trial of the Edmonton Protocol for Islet Transplantation,” New England Journal of Medicine 355:1318-1330 (2006); Baidal et al., “Bioengineering of an Intraabdominal Endocrine Pancreas,” New England Journal of Medicine 376:1887-1889 (2017)).
Cell encapsulation technology offers to protect cells from immune rejection by isolating them from the host using an artificial, semipermeable material, thereby overcoming the need for immunosuppressive agents (Farina et al., “Cell Encapsulation: Overcoming Barriers in Cell Transplantation in Diabetes and Beyond,” Adv. Drug Deliv. Rev. 139:92-115 (2019); Desai et al., “Advances in Islet Encapsulation Technologies,” Nature Reviews Drug Discovery 16:338-350 (2017)). Unlike traditional organ transplantations (e.g., pancreas transplantation), wherein the host circulatory system is connected to the transplanted organ via surgical vascular anastomosis (White et al., “Pancreas Transplantation,” Lancet 373:1808-1817 (2009)), most islet encapsulation devices (i.e., bioartificial pancreas) remain isolated from the host's bloodstream after transplantation. Therefore, encapsulated cells are entirely dependent on O2 and other nutrients by passive diffusion from the surrounding blood vessels at the exterior of the device (Popel, “Theory of Oxygen-Transport to Tissue,” Critical Reviews in Biomedical Engineering 17:257-321 (1989)). It is well documented that O2 is more severely limited than other nutrients because of the relative scarcity of extravascular O2 in vivo (Colton C K, “Oxygen Supply to Encapsulated Therapeutic Cells,” Advanced Drug Delivery Reviews 67-68:93-110 (2014)). In the intraperitoneal site, the O2 tension (pO2) is approximately 40 mmHg, and in the subcutaneous site, likely lower (Barkai et al., “Survival of Encapsulated Islets: More than a Membrane Story,” World J. Transplant. 6:69-90 (2016)).
A thoroughly investigated approach to improve graft oxygenation is to supply exogeneous O2 in situ. The βAir device (Beta-02), for example, supports injections of high concentration O2 into a gas-permeable chamber adjacent to hydrogel-encapsulated cells (Ludwig et al., “Favorable Outcome of Experimental Islet Xenotransplantation Without Immunosuppression in a Nonhuman Primate Model of Diabetes,” Proc. Natl. Acad. Sci. USA 114:11745-11750 (2017); Evron et al., “Long-Term Viability and Function of Transplanted Islets Macroencapsulated at High Density are Achieved by Enhanced Oxygen Supply,” Sci. Rep. 8:6508 (2018)). Another strategy is the local production of O2 using chemical reactions (Pedraza et al., “Preventing Hypoxia-Induced Cell Death in Beta Cells and Islets via Hydrolytically Activated, Oxygen-Generating Biomaterials,” Proc. Natl. Acad. Sci. USA 109:4245-4250 (2012); Wang et al., “An Inverse-Breathing Encapsulation System for Cell Delivery,” Science Advances 7:eabd5835 (2021)) or electrolysis (CA Patent Application No. 2924681 to Tempelman et al.; Wu et al., “In Situ Electrochemical Oxygen Generation with an Immunoisolation Device,” Annals of the New York Academy of Sciences 875:105-125 (1999)). Though these strategies have all demonstrated the benefit of adequate O2 supply to encapsulated cells, remaining limitations include increased device complexity and the requirement of patient compliance to maintain O2 provision. O2 transport in hydrogels is invariably dependent on its permeability, the product of the solubility and diffusivity coefficients, both of which are low in aqueous media such as hydrogels and tissue. An alternative, possibly simpler or complementary approach is thus to improve the O2 permeability of the encapsulating material.
In the absence of supplemental O2 provision, theoretical analyses suggest that islets should be within a few hundred micrometers from the blood stream in surrounding tissue to avoid hypoxia (Dulong et al., “A Theoretical Study of Oxygen Transfer Including Cell Necrosis for the Design of a Bioartificial Pancreas,” Biotechnol. Bioeng. 96:990-998 (2007); Iwata et al., “Design of Bioartificial Pancreases From the Standpoint of Oxygen Supply,” Artif Organs 42:E168-E185 (2018)). Based on this design principle, the cell module of an encapsulation system should be exceedingly thin to support favorable oxygenation (Barkai et al., “Survival of Encapsulated Islets: More than a Membrane Story,” World J. Transplant. 6:69-90 (2016); Papas et al., “Oxygenation Strategies for Encapsulated Islet and Beta Cell Transplants,” Adv. Drug Deliv. Rev. 139:139-156 (2019)). For spherical microcapsules which are endowed with a high surface area to volume ratio, a diameter of ˜1000 μm is widely used (Colton C K, “Oxygen Supply to Encapsulated Therapeutic Cells,” Advanced Drug Delivery Reviews 67-68:93-110 (2014); Liu et al., “Zwitterionically Modified Alginates Mitigate Cellular Overgrowth for Cell Encapsulation,” Nat. Commun. 10:5262 (2019); Bochenek et al., “Alginate Encapsulation as Long-Term Immune Protection of Allogeneic Pancreatic Islet Cells Transplanted into the Omental Bursa of Macaques,” Nat. Biomed. Eng. 2:810-821 (2018)). Similarly, cylindrical cell-laden hydrogel fibers are commonly designed from 350-1000 μm in diameter (Onoe et al., “Metre-Long Cell-Laden Microfibres Exhibit Tissue Morphologies and Functions,” Nature Materials 12:584-590 (2013); Watanabe et al., “Millimeter-Thick Xenoislet-Laden Fibers as Retrievable Transplants Mitigate Foreign Body Reactions for Long-Term Glycemic Control in Diabetic Mice,” Biomaterials 255:120162 (2020); An et al., “Developing Robust, Hydrogel-Based, Nanofiber-Enabled Encapsulation Devices (NEEDs) for Cell Therapies,” Biomaterials 37:40-48 (2015)), and planar slabs (the geometry endowed with the lowest surface area to volume ratio), typically from 250-600 μm in thickness (Desai et al., “Advances in Islet Encapsulation Technologies,” Nature Reviews Drug Discovery 16:338-350 (2017); Ludwig et al., “Favorable Outcome of Experimental Islet Xenotransplantation Without Immunosuppression in a Nonhuman Primate Model of Diabetes,” Proc. Natl. Acad. Sci. USA 114:11745-11750 (2017); Evron et al., “Long-Term Viability and Function of Transplanted Islets Macroencapsulated at High Density are Achieved by Enhanced Oxygen Supply,” Sci. Rep. 8:6508 (2018)).
Clinical islet transplantations require approximately 500 k islet equivalent (IEQ) of human islets (5-10 k IEQ per kg body weight) to reverse diabetes (Shapiro et al., “Clinical Pancreatic Islet Transplantation,” Nat. Rev. Endocrinol. 13:268-277 (2017)), and cellular treatments for liver diseases and hemophilia require similar cell volumes (Roth et al., “Nonviral Transfer of the Gene Encoding Coagulation Factor VIII in Patients with Severe Hemophilia A,” New England Journal of Medicine 344:1735-1742 (2001)). Because O2 diffusion limitations restrict hydrogel thickness, devices can only be scaled along one or two spatial dimensions to accommodate this requisite cell payload, and thus an unreasonably large estimated device length, surface area, or number is required. It is estimated that meters of a cylindrical fiber, hundreds of square centimeters of a planar slab, or ˜100,000 microcapsules are needed to deliver a curative islet dose. Increasing the system's O2 permeability would allow devices to be scaled in three dimensions, facilitating the design of reasonable and surgically convenient device geometries (i.e., shorter in length, smaller in surface area, or lower in number).
The present invention is directed to overcoming these and other deficiencies in the art.
A first aspect of the disclosure relates to an implantable cell containing device. This implantable cell containing device includes a scaffold, and a cell-containing hydrogel encapsulating the scaffold. The scaffold has a tracheal-like internal system of continuous air-filled, hydrophobic micro-channels that traverse the scaffold's dimensions and a hydrophilic external surface layer.
A second aspect of the disclosure relates to a scaffold for use in a cell encapsulation system. The scaffold has a three-dimensional structure including a network of hydrophobic microchannels throughout the three-dimensional structure; and a hydrophilic external surface of the three-dimensional structure.
A third aspect of the disclosure relates to a method of preparing a scaffold for a cell encapsulation system. The method includes the steps of: providing a mold; introducing a polymer solution into the mold and allowing the polymer to solidify and form a three-dimensional structure comprising a network of hydrophobic microchannels throughout the three-dimensional structure; and removing the mold to release the three-dimensional structure, thereby forming the scaffold.
A fourth aspect of the disclosure relates to a method of forming a cell encapsulation system suitable for implant. The method includes the steps of: providing a hydrogel precursor solution comprising one or more cells suspended in the hydrogel precursor solution; and combining the hydrogel precursor solution with a scaffold according to the second aspect in a contained state, and allowing the hydrogel precursors to cross-link to form the hydrogel with the scaffold and the one or more cells embedded within the hydrogel.
A fifth aspect of the disclosure relates to a method of delivering a therapeutic agent to a subject in need thereof. This method involves implanting the implantable cell containing device as described herein.
A sixth aspect of the disclosure relates to a method of treating diabetes in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having diabetes. One particular embodiment of this aspect involves the encapsulation of islet cells in the implantable cell containing device, where the islet cells produce insulin, glucagon, or a combination thereof.
A seventh aspect of the disclosure relates to a method of treating a bleeding disorder in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having a bleeding disorder. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more blood clotting factors.
An eighth aspect of the disclosure relates to a method of treating a lysosomal storage disease in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having the lysosomal storage disease. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more enzymes that are deficient in the subject and thereby treat the lysosomal storage disease.
A ninth aspect of the disclosure relates to a method of treating a neurological disorder in a subject. This method involves implanting the implantable cell containing device of as described herein into the subject having the neurological disorder. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more molecules that are effective to treat the neurological disorder.
A tenth aspect of the disclosure relates to a method of treating a cancer in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having cancer. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more molecules that are effective to treat the cancer.
An eleventh aspect of the disclosure relates to a method of treating a chronic eye disease in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having a chronic eye disease. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more molecules that are effective to treat the chronic eye disease.
A twelfth aspect of the disclosure relates to a method of treating kidney failure in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having a kidney failure. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more molecules that are effective to treat the kidney failure.
A thirteenth aspect of the disclosure relates to a method of treating a chronic pain in a subject. This method involves implanting the implantable cell containing device as described herein into the subject having a chronic pain. One particular embodiment of this aspect involves the encapsulation of cells in the implantable cell containing device, where the cells produce and release one or more molecules that are effective to treat the chronic pain.
Inadequate oxygenation is a major challenge in cell encapsulation, a therapy which holds potential to treat many diseases including type I diabetes. In such systems, cellular oxygen (O2) delivery is limited to slow passive diffusion from transplantation sites through the poorly O2-soluble encapsulating matrix, usually a hydrogel. This constrains the maximum permitted distance between the encapsulated cells and host site to within a few hundred micrometers to ensure cellular function.
The physiology of insects presents a creative solution to rapid O2 distribution across multi-millimeter scales. Instead of using circulatory blood for tissue oxygenation as in vertebrates (Pittman, “Regulation of Tissue Oxygenation,” Colloquium Series on Integrated Systems Physiology: from Molecule to Function, Morgan & Claypool Life Sciences (2011), which is hereby incorporated by reference in its entirety), insects transport gaseous oxygen via a gas-filled channel network, known as the tracheal system, which is distributed throughout their bodies. This system is also present in some aquatic insects without spiracles (Klowden, Physiological Systems in Insects, Third Edition, Chapter 7: “Circulatory Systems,” pp. 357-401, and Chapter 9: “Respiratory Systems,” pp. 433-461, Elsevier (2013), which is hereby incorporated by reference in its entirety). Inspired by this clever mechanism of rapid gas-phase O2 distribution, the present invention includes the design of an air-filled scaffold for islet encapsulation, which was pursued to overcome the thickness limitation for O2 diffusion and thereby termed Speedy Oxygenation Network for Islet Constructs (or SONIC). See
The bio-inspiration was highlighted by analyzing the tracheal anatomy of a larva of the mealworm beetle (Tenebrio molitor) (
The incorporation of the SONIC scaffold in a hydrogel-based cell encapsulation system described in the present application intentionally recapitulated the efficiency of rapid O2 transport of the insect tracheal network, oxygenating deeply encapsulated cells within the thick devices (
The present disclosure relates to implantable cell containing devices, methods of producing these devices, and methods of using the same.
One aspect of the disclosure relates to an implantable cell containing device. This implantable cell containing device includes a scaffold and a cell-containing hydrogel encapsulating the scaffold. The scaffold includes a tracheal-like internal system of continuous air-filled, hydrophobic micro-channels that traverse the scaffold's dimensions, and has a hydrophilic external surface.
The scaffold may have any of a variety of constructions such that the bulk of the hydrogel material that surrounds the scaffold, and the cells that are contained in the hydrogel, is not more than about 1000 μm from the external surface of the scaffold, such as about 900 μm from the external surface of the scaffold, about 800 μm from the external surface of the scaffold, about 700 μm from the external surface of the scaffold, or 600 μm from the external surface of the scaffold. In certain embodiments, the bulk of the hydrogel material that surrounds the scaffold, and the cells that are contained in the hydrogel, is not more than about 500 μm from the external surface of the scaffold, preferably not more than about 450 μm, or about 400 μm, or about 350 μm, or about 300 μm, or about 250 μm from the external surface of the scaffold. This helps to ensure that, via diffusion of oxygen from the scaffold into the hydrogel matrix, the cells contained in the hydrogel matrix remain viable.
The variety of scaffold constructions are not limited to any particular shape or configuration. Exemplary shapes or configurations are presented in the accompanying examples, and include a ladder-like geometry with varying heights and lengths (see
The scaffold has an internal microstructure that is bi-continuous and non-wettable (hydrophobic) to provide unobstructed microchannels for O2 flow similar to the insect tracheae. The scaffold also has an external surface that is rough and wettable (hydrophilic) to allow cell-laden hydrogel precursor to infiltrate. Although the architecture of the channels in the scaffold are largely in the micron range, it is to be understood that sub-micron channels may or may not be present. Thus, as used herein, the term ‘microchannels’ also encompasses channels that would be considered nanochannels due to their dimensions.
According to one embodiment, a hydrophobic polymer material is used to form the scaffold.
In some embodiments, the scaffold comprises a fluorinated polymer material. Suitable fluorinated polymer materials that can be used include, without limitation, poly(vinylidene fluoride-co-hexafluoropropylene) (PVDF-HFP), poly(vinylidene fluoride) (PVDF), polyvinylidene difluoride, polytetrafluoroethylene (PTFE), poly(vinylidene fluoride-co-trifluoroethylene) (P(VDF-TrFE)), poly(vinylidene fluoride-co-tetrafluoroethylene) (P(VDF-TFE)), poly(vinylidene fluoride-co-chlorotrifluoroethylene) (P(VDF-CTFE)), Teflon AF® family: copolymers made from 2,2-bistrifluoromethyl-4,5-difluoro-1,3-dioxole and tetrafluoroethylene, and polychlorotrifluoroethylene (PCTFE).
Other suitable polymer materials that can be used include, without limitation, silicone, PDMS, rubber, nylon, polyurethane, polysulfone, polyacrylonitrile, polyester such as polyethylene terephthalate and polybutester, polyacrylamide, poly(ethyl methacrylate), poly(methyl methacrylate), polyvinyl chloride, polyoxymethylene, polycarbonate, polypropylene, polyethylene, polybenzimidazole, polyaniline, polystyrene, polyvinylcarbazole, polyamide, poly vinyl phenol, cellulose acetate, polyacrylamide, poly(2-hydroxyethyl methacrylate), polyether imide, poly(ferrocenyldimethylsilane), poly(ethylene-co-vinylacetate), polyethylene-co-vinyl acetate, polyacrylic acid-polypyrene methanol, poly(ethylene-co-vinyl alcohol), polymetha-phenylene isophthalamide, poly(lactic acid), poly(ε-caprolactone), poly(lactic-co-glycolic acid), poly(1-lactide-co-ε-caprolactone), and combinations thereof.
In an alternative embodiment, when a non-hydrophobic polymer material is used to form the scaffold, the microchannels of the scaffold are subsequently treated to render the microchannels hydrophobic (non-wettable). In one approach, the microporous scaffold can be chemically modified with fluoroalkysilanes to render the microchannels hydrophobic. Alternatively, the microchannels in the scaffold can be modified to comprise SiO2 nanoparticles, attapulgite, ZnO nanoparticles or nanorods, and combinations thereof.
In yet another embodiment, the scaffold is formed of a carbon material. Suitable carbon materials that can be used include, without limitation, activated carbon, carbon microbelts, graphite, carbon nanoparticles, carbon soot, carbon nanofibers, graphene, and carbon nanotubes.
In alternative embodiments, a mixture of carbonaceous materials and polymer materials are used to form the scaffold.
For scaffold materials that are hydrophobic in nature, the external surface of the scaffold should be treated to render the external surface hydrophilic. In certain embodiments, a hydrophilic polymer coating is applied to the external surface. Suitable hydrophilic coating materials include, without limitation, polydopamine coatings and silane coatings, such as PEGylated silane coatings.
In an alternative embodiment, the external surface of the scaffold can be treated by exposing the scaffold to plasma (e.g., atmospheric), which will render the otherwise hydrophobic polymer surface hydrophilic.
The inventive scaffolds can be prepared by first preparing or providing a mold, and then introducing a polymer solution into the mold and allowing the polymer to solidify and form a three-dimensional structure comprising a network of microchannels throughout the three-dimensional structure, and then removing the mold to release the three-dimensional structure, thereby forming the scaffold. This is generally illustrated in
In certain embodiments, the mold is prepared using a three-dimensional printing technique that forms a patterned mold, which has a pattern that allows for development of the desired three-dimensional structure of the scaffold. The mold is shown to the left side of
The step of introducing a polymer solution into the mold and allowing the polymer to solidify and form a three-dimensional structure is carried out by phase separation technique. As demonstrated in the accompanying examples, phase separation is a method for forming the scaffold by precipitation of polymers from a polymer-poor phase and a polymer-rich phase. The advantage of the phase separation process is that it is a relatively simple procedure and requires minimal apparatus (i.e., the mold) and suitable solvent/nonsolvent for a given polymer. Most polymer solution systems have a suitable, corresponding poor solvent (nonsolvent) to create a porous structure using the phase separation technique. In this process, the polymer is dissolved in solution and the phase separation is induced, either thermally or through the addition of a non-solvent to the polymer solution to create a gel. The polymer solution under this condition becomes thermodynamically unstable and tends to separate into two phases. Water is then used to extract the solvent from the gel; the polymer-rich phase then solidifies on reducing the temperature to a 3-D porous composite scaffold. This is illustrated in the middle steps of
As alternatives to nonsolvent-induced phase separation, additional methods can also be used to create porous scaffold structures via phase separation. These include, without limitation, thermally induced phase separation (TIPS) (see Nam et al., “Porous Biodegradable Polymeric Scaffolds Prepared by Thermally Induced Phase Separation,” J. Biomed. Mat. Res. 47(1):8-17 (1999); Lee et al., “Bicontinuous Phase Separation of Lithium-ion Battery Electrodes for Ultrahigh Areal Loading,” Proc. Natl. Acad. Sci. USA 117(35): 21155-21161 (2020), each of which is hereby incorporated by reference in its entirety); supercritical gel drying (see Cardea et al., “Supercritical Gel Drying: A Powerful Tool for Tailoring Symmetric Porous PVDF-HFP Membranes,” ACS Appl. Mater. Interfaces 1(1):171-180 (2009), which is hereby incorporated by reference in its entirety); and salt leaching (see Corriera et al., “Strategies for the Development of Three Dimensional Scaffolds from Piezoelectric Poly(vinylidene fluoride),” Materials & Design 92:674-681 (2016), which is hereby incorporated by reference in its entirety). Any other suitable phase separation techniques can also be utilized.
Subsequent to forming the scaffold, the mold is removed from the scaffold. Removal of the mold is preferably carried out by dissolving the mold from the exterior of the scaffold. Using PLA as the mold material, the PLA can be dissolved using chloroform to liberate the three-dimensional structure of the scaffold. This is illustrated in the final step of
Once the scaffold is liberated from the mold, the scaffold is then modified—as described above—to render the microchannels hydrophobic and/or the external surface of the scaffold hydrophilic. In addition, to facilitate hydrogel cross-linking to the hydrophilic external surface of the scaffold, the scaffold can optionally be treated with one or more agents to promote cross-linking of the scaffold surface to the hydrogel matrix. With the scaffold having these properties, the scaffold is ready to be integrated with the hydrogel matrix (containing one or more types of cells) to form the implantable cell containing devices. The scaffold can optionally be sterilized by any suitable means prior to use.
Having prepared the scaffold, the scaffold can be encapsulated by a hydrogel matrix (containing one or more cells) to form an implantable cell containing device of the invention. Briefly, a hydrogel precursor solution containing one or more cells suspended in the hydrogel precursor solution is formed, and then the hydrogel precursor solution is combined with the scaffold of the invention in a contained state, while allowing the hydrogel precursors to cross-link to form the hydrogel with the scaffold and the one or more cells embedded within the hydrogel.
The hydrogel layer is preferably formed using a cell growth matrix material. Suitable cell growth matrix materials include, without limitation, a natural polymeric material, a synthetic polymeric material, or a combination thereof.
Exemplary synthetic polymer materials include, without limitation, polyethylene glycol (PEG), poly(acrylic acid), poly(ethylene oxide), poly(vinyl alcohol), polyphosphazene, poly(hydroxyethyl methacrylate), triazole-zwitterion hydrogels (TR-qCB, TR-CB, TR-SB), poly(sulfobetaine methacrylate), carboxybetaine methacrylate, poly-2-methacryloyloxyethyl phosphorylcholine, N-hydroxyethyl acrylamide, copolymers thereof, derivatives thereof, and combinations thereof.
Exemplary natural polymeric materials include, without limitation, hyaluronate, collagen, elastin, fibrin, gelatin, gelatin-methacryloyl, silk fibroin, glycosaminoglycans, dextran, alginate, agarose, chitosan, bacterial cellulose, keratin, matrigel, decellularized hydrogels, and derivatives or combinations thereof.
Preferred hydrogel materials include alginate, collagen, hyaluronate, fibrin, fibroin, agarose, chitosan, bacterial cellulose, elastin, keratin, polyethylene glycol, a polyethylene glycol derivative, poly(2-hydroxyethyl methacrylate), a poly(2-hydroxyethyl methacrylate) derivative, and combinations thereof. The alginate can be a pure alginate, a modified alginate, or a mixture of pure and modified alginate. Suitable modified alginates that can be used include zwitterionically modified alginate.
In any embodiment, the hydrogel layer has a thickness of about 100 μm to about 1000 μm. For example, and without limitation, the hydrogel layer of the implantable cell containing device may comprise a thickness of about 100 μm to about 200 μm, about 200 μm to about 300 μm, about 300 μm to about 400 μm, about 400 μm to about 500 μm, about 500 μm to about 750 μm, about 750 μm to about 1000 μm, about 100 μm to about 400 μm, about 100 μm to about 350 μm, about 100 μm to about 300 μm, about 150 μm to about 300 μm, or about 150 μm to about 300 μm.
The hydrogel layer comprises a preparation of cells. In any embodiment, the preparation of cells positioned or encapsulated in the hydrogel layer of the cell encapsulation device is a preparation of single cells such as a preparation of a single type of cells, and a preparation of multiple types of cells. In any embodiment, the preparation of a cells is a preparation of cell aggregates. In any embodiment, the preparation of cells is a preparation of single cells and cell aggregates.
The cells introduced into the hydrogel can be primary cells (autologous or allogenic) or immortalized cells.
In any embodiment, the preparation of cells can be mammalian cells, such as primate cells, rodent cells, canine cells, feline cells, equine cells, bovine cells, porcine cells, or human cells.
In certain embodiments, the preparation of cells positioned or encapsulated in the hydrogel layer includes stem cells or stem cell derived cells. The stem cells can be pluripotent, multipotent, oligopotent, or unipotent stem cells. For example, the preparation of stem cells may contain embryonic stem cells, epiblast cells, primitive ectoderm cells, primordial germ cells, and/or induced pluripotent stem cells.
In certain embodiments, the preparation of cells positioned or encapsulated in the hydrogel layer includes one or more of smooth muscle cells, cardiac myocytes, platelets, epithelial cells, endothelial cells, urothelial cells, fibroblasts, embryonic fibroblasts, myoblasts, chondrocytes, chondroblasts, osteoblasts, osteoclasts, keratinocytes, hepatocytes, bile duct cells, islet cells, thyroid, parathyroid, adrenal, hypothalamic, pituitary, ovarian, testicular, salivary gland cells, adipocytes, embryonic stem cells, mesenchymal stem cells, neural cells, endothelial progenitor cells, hematopoietic cells, precursor cells, mesenchymal stromal cells, Baby Hamster Kidney (BHK) cells, Chinese Hamster Ovary cells, Human Amniotic Epithelial (HAE) cells, choroid plexus cells, chromaffin cells, adrenal chromaffin cells, pheochomocytoma cell line PC12, human retinal pigment epithelium cells, recombinant human retinal pigment epithelium cells, NGF-secreting Baby Hamster Kidney (BHK) cells, human bone marrow-derived stem cells transfected with GLP-1, BDNF-producing fibroblasts, NGF-producing cells, CNTF-producing cells, BDNF-secreting Schwann cells, IL-2-secreting myoblasts, endostatin-secreting cells, and cytochrome P450 enzyme over-expressed feline kidney epithelial cells, myogenic cells, embryonic stem cell-derived neural progenitor cells, irradiated tumor cells, proximal tubule cells, neural precursor cells, astrocytes, genetically engineered cells.
In certain embodiments, the preparation of cells positioned or encapsulated in the hydrogel layer includes islet cells, such as a preparation of human islet cells, porcine islet cells, or rodent islet cells. The islet cells may produce insulin, glucagon, or both.
In certain embodiments, the preparation of cells positioned or encapsulated in the hydrogel layer includes cells that produce one or more of insulin, coagulation factors, albumin, urea, human cytochrome P450 enzymes, and combinations thereof.
In any embodiment, the cells may be present in the hydrogel layer at a cell density of between about 1×103 to about 1×1010 cells/mL. For example, the cell density may range from about 1×103 cells/mL up to about 1×104 cells/mL, about 1×104 cells/mL up to about 1×105 cells/mL, about 1×105 cells/mL up to about 1×106 cells/mL, about 1×106 cells/mL up to about 1×107 cells/mL, about 1×107 cells/mL up to about 1×108 cells/mL, about 1×108 cells/mL up to about 1×109 cells/mL, or about 1×109 cells/mL up to about 1×1010 cells/mL.
In any embodiment, the cells may be present in the hydrogel layer at a concentration of about 1%-40% v/v cells/hydrogel. For example, the cells are present in the hydrogel layer of the device at a concentration of about 5%-40% v/v, 10%-35% v/v, 15%-30% v/v, 20%-30% v/v, 10%-25% v/v, 15%-25% v/v, 5%-10% v/v, 5%-20% v/v, 5%-30% v/v, 35%-40% v/v, 30%-40% v/v, 25%-40% v/v, or 20%-40% v/v cells/hydrogel.
The hydrogel may optionally be supplemented with one or more cell factors or biologically active agents to enhance cell growth, differentiation, and/or survival of the cells positioned within the hydrogel material. Suitable biologically active agents include, without limitation, a protein, peptide, antibody or antibody fragment thereof, antibody mimetic, a nucleic acid, a small molecule, a hormone, a growth factor, an angiogenic factor, a cytokine, an anti-inflammatory agent, and any combination thereof.
Exemplary growth factors include, without limitation, an epidermal growth factor, fibroblast growth factor, transforming growth factor/bone morphogenetic protein, platelet derived growth factor, insulin growth factor, FGF, bFGF, acid FGF (aFGF), FGF-2, FGF-4, EGF, PDGF, TGF-beta, angiopoietin-1, angiopoietin-2, placental growth factor (PlGF), or VEGF.
Exemplary anti-inflammatory agents include, without limitation, diclofenac, diflunisal, etodolac, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, mefenamic acid, meloxicam, nabumetone, naproxen, oxaprozin, piroxicam, salsalate, sulindac, tolmetin, and combinations thereof.
The hydrogel may optionally be supplemented with one or more contrast agents to facilitate in vivo monitoring of the implantable cell containing device when implanted to determine device placement, location of the implanted device at some time point after implantation, health of the implanted device, deleterious effects on non-target cell types, inflammation, and/or fibrosis. Suitable contrast agents include, without limitation, nanoparticles, nanocrystals, gadolinium, iron oxide, iron platinum, manganese, iodine, barium, microbubbles, fluorescent dyes, and others known to those of skill in the art.
In certain embodiments, the preparation of cells positioned or encapsulated in the hydrogel layer includes islet cells that release insulin, glucagon, or both insulin and glucagon. In certain embodiments, the insulin producing cells may be a preparation of human SC-β cells. In certain embodiments, the preparation of islet cells and/or SC-β cells is a preparation of human islets and/or human SC-β cells, porcine islets and/or porcine SC-β cells, or rodent islets and/or rodent SC-β cells. In these embodiments, the preparation of cells comprises an islet density between about 1×103 to about 6×105 islet equivalents (IEQs)/mL. In these embodiments, the preparation of cells comprises an islet density between about 1×103 to about 6×104 islet equivalents (IEQs)/mL. For example, the islet equivalents may range from about 1×103 up to about 2×103, about 2×103 up to about 3×103, about 3×103 up to about 4×103, about 4×103 up to about 5×103, about 5×103 up to about 6×103, about 6×103 up to about 7×103, about 7×103 up to about 8×103, about 8×103 up to about 9×103, about 9×103 up to about 1×104, about 1×104 up to about 2×104, about 2×104 up to about 3×104, about 3×104 up to about 4×104, about 4×104 up to about 5×104, about 5×104 up to about 6×104, about 6×104 up to about 7×104, about 7×104 up to about 8×104, about 8×104 up to about 9×104, about 9×104 up to about 1×105, about 1×105 up to about 2×105, about 2×105 up to about 3×105, about 3×105 up to about 4×105, about 4×105 up to about 5×105, or about 5×105 up to about 6×105 islet equivalents (IEQs)/mL.
The implantable cell containing devices of the present invention are intended to be implanted in a subject to afford therapeutic benefit to that subject. In particular, the devices can be implanted using laproscopic surgical procedures or open surgical sites, and the devices can be placed subcutaneously, transcutaneously, preperitoneally, transperitoneally, or intraperitoneally. In some embodiments, implanting involves suturing the device or system to a body wall of the subject; anchoring the device to a body wall of the subject via a transabdominal portal; wrapping the delivery device or system in omentum of the subject; positioning the device in a cavity between the liver and the diaphragm; or anchoring the device to the diaphragm.
Both veterinary and medical uses are contemplated. Thus, exemplary subjects include, without limitation, a human, a mouse, a rat, a dog, a pig, a sheep, a cow, a horse, and a nonhuman primate.
By implanting the implantable cell containing devices, the devices can be used to deliver a therapeutic agent to a subject in need thereof. The treatment by implantation can be carried out for a limited duration over a period of days, weeks, or months. Thus, it is also contemplated that the method of treatment further involves retrieving the implantable cell containing device from the subject when no longer needed or when the device needs replacement, and optionally implanting a replacement implantable cell containing device after the initial device is retrieved.
As indicated above, the introduction of one or more contrast agents allows for monitoring of the device. Methods of in vivo monitoring include but are not limited to confocal microscopy, 2-photon microscopy, high frequency ultrasound, optical coherence tomography (OCT), photoacoustic tomography (PAT), computed tomography (CT), magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). These alone or combined can provide useful means to monitoring the implantable device. Monitoring of the device may be used to determine when to remove and replace a device, as necessary.
According to one embodiment, the subject has diabetes, is in need of diabetes treatment, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein, which comprises a preparation of cells that release insulin, glucagon, or a combination thereof for the treatment of diabetes in the subject. Exemplary cells that release the therapeutic agent include one or more of islet cells, islets derived from a preparation of stem cells such as pluripotent, multipotent, oligopotent, or unipotent stem cells, including embryonic stem cells, epiblast cells, primitive ectoderm cells, primordial germ cells, and induced pluripotent stem cells.
According to one embodiment, the subject has a bleeding disorder, is in need of treatment for the bleeding disorder, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein, which comprises a preparation of cells that release a therapeutic agent that treats the bleeding disorder. In accordance with this embodiment, the bleeding disorder can be any bleeding disorder, such as hemophilia A, hemophilia B, von Willebrand disease, Factor I deficiency, Factor II deficiency, Factor V deficiency, Factor VII deficiency, Factor X deficiency, Factor XI deficiency, Factor XII deficiency, and Factor XIII deficiency, and the therapeutic agent is a blood clotting factor selected from the group of Factor I, Factor II, Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of recombinant myoblasts, mesenchymal stromal cells, endothelial cells, induced pluripotent stem cell derived endothelial cells, induced pluripotent stem cell derived mesenchymal stromal cells, and a combination thereof.
In another embodiment, the subject has a lysosomal storage disorder, is in need of treatment for the lysosomal storage disorder, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein into the subject having the lysosomal storage disorder. In accordance with this embodiment, the therapeutic agent is an enzyme selected from the group of α-L-iduronidase, Iduronate-2-sulfatase, α-glucuronidase, Arylsulfatase A, alpha-Galactosidase A, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of hematopoietic stem cells, fibroblasts, myoblasts, Baby Hamster Kidney (BHK) cells, Chinese Hamster Ovary cells, Human Amniotic Epithelial (HAE) cells, mesenchymal stromal cells, induced pluripotent stem cell derived mesenchymal stromal cells, and combinations thereof.
According to one embodiment, the subject has a neurological disorder, is in need of treatment for the neurological disorder, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein, which comprises a preparation of cells that release a therapeutic agent that treats the neurological disorder. In accordance with this embodiment, the neurological disorder is Parkinson's disease, Alzheimer's disease, epilepsy, Huntington's disease, Amyotrophic lateral sclerosis, chronic pain, a sensory disorder such as visual loss, hearing loss, peripheral nerve injury, and spinal cord injury, and the therapeutic agent is selected from the group of cerebrospinal fluid, extracellular fluid, levodopa, nerve growth factor (NGF), ciliary neurotrophic factor (CNTF), BLP-1, brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), enkephalin, adrenaline, catecholamine, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of choroid plexus cells, chromaffin cells, pheochomocytoma cell line PC12, human retinal pigment epithelial cells, NGF-secreting Baby Hamster Kidney (BHK) cells, myoblasts, human bone marrow-derived stem cells transfected with GLP-1, BDNF-producing fibroblasts, NGF-producing cells, CNTF-producing cells, adrenal chromaffin cells, BDNF-secreting Schwann cells, myogenic cells, embryonic stem cell-derived neural progenitor cells, and combinations thereof.
In another embodiment, the subject has a cancerous condition, is in need of treatment for the cancerous condition, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein into the subject having the cancerous condition. In accordance with this embodiment, the therapeutic agent is one or more of IL-2, endostatin, cytochrome P450 enzyme, a tumor antigen, a cytokine, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of IL-2-secreting myoblasts, endostatin-secreting cells, Chinese Hamster Ovary cells, cytochrome P450 enzyme overexpressed feline kidney epithelial cells, irradiated tumor cells, and combinations thereof.
In another embodiment, the subject has a chronic eye disease, is in need of treatment for the chronic eye disease, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein into the subject having the chronic eye disease. The chronic eye disease may be any one of age-related macular degeneration, diabetic retinopathy, retinitis pigmentosa, glaucoma, macular telangiectasia, and combinations thereof. In accordance with this embodiment, the therapeutic agent is one or more trophic factors that protect compromised retinal neurons and restore neural circuits, such as any one or more of ciliary neurotrophic factor, antagonists against vascular endothelial growth factor and platelet-derived growth factor, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of human retinal pigment epithelium cells, recombinant human retinal pigment epithelium cells, and combinations thereof.
In one embodiment, the subject has kidney disease (kidney failure), is in need of treatment for the kidney disease (kidney failure), and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein into the subject having the kidney disease (kidney failure). In accordance with this embodiment, the therapeutic agent is dopamine, atrial natriuretic peptide, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of renal proximal tubule cells, mesenchymal stem cells, and combinations thereof.
In one embodiment, the subject has chronic pain, is in need of treatment for the chronic pain, and the method of delivering a therapeutic agent to the subject involves implanting an implantable cell containing device or system as described herein into the subject having the chronic pain. The chronic pain can be any chronic pain condition including, without limitation, those caused by degenerative back and knee, neuropathic back and knee, or cancer. In accordance with this embodiment, the therapeutic agent is catecholamine, opioid peptides, enkephalins, and combinations thereof. Exemplary cells that release the therapeutic agent include one or more of chromaffin cells, neural precursor cells, mesenchymal stem cells, astrocytes, and genetically engineered cells, and combinations thereof.
Wherever the word “about” is employed herein in the context of dimensions (e.g. distances, sizes), time, amounts (relative amounts, concentration, etc.), cell densities, etc., it will be appreciated that such variables are approximate and as such may vary by ±10%, for example ±5% and preferably ±2% (e.g. ±1%) from the numbers specified herein.
The examples below are intended to exemplify the practice of embodiments of the disclosure but are by no means intended to limit the scope thereof.
Materials: Poly(vinylidene fluoride-co-hexafluoropropylene) (PVDF-HFP, Mw=455 kDa), Tris hydrochloride (Tris-HCl), sodium hydroxide, dopamine hydrochloride, sodium chloride (NaCl), calcium chloride dihydrate (CaCl2·2H2O), barium chloride dihydrate (BaCl2·2H2O), calcium sulfate dihydrate (CaSO4·2H2O), Nile Red, gelatin, and D-glucose were purchased from Sigma-Aldrich. Poly(lactic acid) (PLA) filament was purchased from PRUSA. Ultrapure sodium alginate (Pronova SLG100) was purchased from NovaMatrix. Water was deionized to 18.2 MΩ·cm with a Synergy UV purification system (Millipore Sigma).
Animals: Male C57BL/6J mice (2 months old) were purchased from The Jackson Laboratory. The mice were maintained at a temperature of 70-72° F. with 30-70% humidity under a 14-hour light/10-hour dark cycle. Male Sprague-Dawley rats (weight of ˜300 g) were purchased from Charles River Laboratories.
Characterizations: High-resolution X-ray computer tomography (Nano-CT) scanning was conducted on a 3D X-ray microscope (ZEISS Xradia 520 Versa). Scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDS) element mapping were performed using a field emission scanning electron micro-analyzer (LEO 1550). Contact angle images were taken using a contact angle goniometer (Rame-Hart 500). Optical and fluorescent microscope images were taken using a digital microscope (EVOS FL). H&E staining images were taken using an Aperio Scanscope (CS2). Stereo microscope images were taken by a stereomicroscope (Olympus SZ61). Immunofluorescence images were taken using a confocal microscope (ZEISS LSM 710). OriginPro 8.5.1 software and GraphPad Prism 8 software were used for data plotting.
Fabrication of the SONIC Scaffold: PVDF-HFP was dissolved in acetone at concentration of 15 wt % under heat in a sealed glass vial. After cooling to room temperature, the PVDF-HFP solution was filled into a 3D printed PLA mold (Original Prusa i3 MK2S) and, after ˜1 minute, the polymer solution and mold were immersed in a water/ethanol (V/V=1/1) bath for ˜10 minutes to facilitate the phase separation process, and then transferred to a water bath for ˜30 minutes for a solidification process. Next, the solidified PVDF-HFP was immersed in ethanol and hexane for two dehydration steps of about ˜10 minutes each, followed by air drying at ambient temperature. Finally, the SONIC scaffold was obtained after the selective extraction of the PLA mold with chloroform. The SONIC scaffolds used for in vivo studies were sterilized by autoclave prior to use.
Nano-CT Imaging of the Mealworm and SONIC Scaffold: To prepare a mealworm specimen for Nano-CT scanning, a 2 cm-long mealworm was loaded in a 1 mL pipet tip and sacrificed by freezing at −20° C. 6 individual scans were performed on different sections of the mealworm using an “oversize scan” option to get a full image of the specimen. During the scans, the X-ray source was set to a voltage of 100 kV, and the scanning resolution was set as 5.19 μm per pixel under a binning mode of 2×2. Subsequently, 3D reconstruction of the obtained images was performed using Avizo software (version 8.1.1). A segmentation process was conducted to visualize the tracheal system of the mealworm based on the different absorption contrasts between the respiratory gases and mealworm tissues.
To prepare a SONIC scaffold specimen for the Nano-CT scanning, a small piece of scaffold (˜1 mm3) was cut and attached on a tip. During the scan, the X-ray source was set to a voltage of 100 kV, and the scanning resolution was set as 0.268 μm per pixel under a binning mode of 2×2. Subsequently, 3D reconstruction of the obtained images was performed using Avizo software. Network connectivity on the polymeric and the porous regions of the scaffold was performed using ImageJ.
Electron Paramagnetic Resonance (EPR) for O2 Mapping: O2 mapping was performed on a 25 mT EPR imager (JIVA-25, O2M Technologies, LLC). The JIVA-25 operates at 720 MHz using electron paramagnetic resonance oxygen imaging (EPROI) principles and utilizes oxygen sensitive electron spin-lattice relaxation rates (T1) of trityl radical probe OX063-D24 (methyl-tris[8-carboxy-2,2,6,6-tetrakis[(2-hydroxyethyl]benzo[1,2-d:4,5-d′]bis[1,3]dithiol-4-yl]-trisodium salt) for reporting pO2.
A SONIC scaffold or control scaffold was fixed at the bottom of a glass tube (VWR, 10×75 mm) using a dental vinyl polysiloxane impression material. Gelatin solution (1 mL) (1 wt %) containing spin probe (1 mM OX063-d24) was filled into the container with the top end of scaffold exposed above the gelatin. First, the system was deoxygenated using N2 to reduce pO2 close to 0 mmHg. After deoxygenation, the system was exposed to a gas mixture containing 5% O2 and 95% N2, and the pO2 change in gelatin was continuously monitored until a steady state (40 mmHg) was approached.
Average pO2 measurements were performed using inversion recovery electron spin echo (IRESE) sequence (Epel et al., “Absolute Oxygen R1e Imaging In Vivo with Pulse Electron Paramagnetic Resonance,” Magnetic Resonance in Medicine 72:362-368 (2014), which is hereby incorporated by reference in its entirety) with the following parameters: pulse lengths 60 ns, 16 phase cycles scheme with FID suppression, spin echo delay 500 ns, 80 logarithmically spaced delays from 400 ns-65 μs, 100 us repetition time. The curves were fitted using single exponential recovery to extract R1 (1/T1) values that were converted to pO2 (
Fabrication of the SONIC Device: The SONIC scaffold was immersed into a dopamine solution (2 mg/mL in 10 mM tris buffer, pH 8.5) overnight to create a hydrophilic polydopamine coating on the scaffold surface. Subsequently, CaSO4 was deposited onto the scaffold surface by dipping it into a CaSO4 saturated solution (0.24 wt % in water) and then drying it at 60° C., leaving CaSO4 crystals on the scaffold surface. Next, the scaffold was inserted into a glass tubing mold with sodium alginate (2%) solution. Alginate crosslinking then occurred by the Ca2+ ions diffused from the CaSO4. Finally, the SONIC device was pushed out from the tubing mold into a cross-linking buffer (95 mM CaCl2+5 mM BaCl2), leaving the device in buffer around 4 min for a further cross-linking. Constructs which contained INS-1 cells or islets were fabricated by premixing the alginate solution with the cells before application onto the scaffolds.
To fabricate control devices without the SONIC scaffold, a tubing mold was prepared by rolling a dialysis membrane (Spectra/Por®, MWCO 3500) into a tube with an inner diameter of ˜4 mm and sealing one end with a PDMS cap. Then, INS-1 cells or islets alginate solution were loaded into the mold and immersed in the buffer (95 mM CaCl2+5 mM BaCl2) for cross-linking by the Ca2+ and Ba2+ ions which diffused through the dialysis membrane. Next, the tubing mold was unrolled to leave the alginate in the buffer around 4 min for further cross-linking.
For the devices in mice studies, 500 IEQ of rat islets distributed in approximately 170 μL alginate were incorporated in each cylindrical (4.2 mm in diameter, 20.4 mm in length) SONIC device (
In Vitro Cell Viability Study: INS-1 cells were purchased from Sigma-Aldrich and cultured with RPMI 1640 medium (Gibco) supplemented with 10% FBS (Gibco), 10 mM HEPES (Gibco), 2 mM glutamine (Gibco), 1 mM sodium pyruvate (Gibco), 50 μM β-mercaptoethanol (Gibco), and 1% penicillin/streptomycin (Gibco). Trypsin-dissociated INS-1 cells were suspended in alginate solution at a density of 2.5 million cells/mL and incorporated into SONIC devices or control devices, and then were incubated in the above-mentioned medium in a hypoxic incubator with 5% O2, 5% CO2 at 37° C. After 48 hours, the cells in devices were stained with a LIVE/DEAD™ viability/cytotoxicity kit (Invitrogen).
Rat Islet Isolation and Purification: Sprague-Dawley rats were used for harvesting islets. The rats were anesthetized using 3% isoflurane in O2 throughout the whole surgery. Briefly, the pancreas was distended with 10 mL 0.15% Liberase (Roche) in M199 media (Gibco) through the bile duct. The pancreas was digested at 37° C. circulating water bath for ˜28 mins (digestion time varied slightly for different batches of Liberase). The digestion was stopped by adding cold M199 media with 10% FBS (Gibco). After vigorously shaking, the digested pancreases were washed twice with media (M199+10% FBS), filtered through a 450 μm sieve, and then suspended in a Histopaque 1077 (Sigma)/M199 media gradient and centrifuged at 1700 RCF with 0 break and 0 acceleration for 17 min at 4° C. This gradient centrifugation step was repeated for higher purity. Finally, the islets were collected from the gradient and further isolated by a series of gravity sedimentations, in which each top supernatant was discarded after 4 min of settling. Islet equivalent (IEQ) number of purified islets was counted by reported IEQ conversion factors (Buchwald et al., “Quantitative Assessment of Islet Cell Products: Estimating the Accuracy of the Existing Protocol and Accounting for Islet Size Distribution,” Cell Transplant 18:1223-1235 (2009), which is hereby incorporated by reference in its entirety). Islets were then washed once with islet culture media (RPMI 1640 supplemented with 10% FBS, 10 mM HEPES, and 1% penicillin/streptomycin) and cultured in this medium overnight before further use.
Implantation and Retrieval in Mice: C57BL/6J mice were administered an intraperitoneal injection of freshly prepared STZ solution (22.5 mg/mL in 100 mM sodium citrate buffer, pH 4.5) at a dosage of 150 mg STZ/kg mouse to induce diabetes one week before device implantation. Only mice with non-fasted blood glucose levels above 350 mg/dL were considered as diabetic. The diabetic mice were anesthetized with 3% isoflurane in O2 and the abdomen area was shaved and sterilized using betadine and 70% ethanol. A small skin incision (˜8 mm) was made along the midline of the abdomen, and then a following incision was made along the linea alba. The device was introduced into the peritoneal cavity through the incision. The peritoneal wall was closed using 5-0 absorbable polydioxanone (PDS II) sutures and the skin incision was closed using 5-0 nylon sutures.
For retrieval, the mice were treated with the same procedures as above. Then, the device was located and pulled out from the peritoneal cavity using a tweezer. The incisions were sutured and keep the mice alive for following BG monitoring after device retrieval.
Morphology and Immunohistochemistry of Islets in Retrieved Devices: The retrieved devices were fixed with 10% formalin, embedded in paraffin, and sectioned into 5 μm sections. Hematoxylin and eosin (H&E) staining was performed. For immunofluorescent insulin and glucagon staining, paraffin-embedded sections were deparaffinized in xylene and sequentially rehydrated in 100% ethanol, 95% ethanol, 75% ethanol, and PBS. Slides were then boiled in citric acid buffer (10 mM citric acid, 0.05% Tween 20, pH 6.0) for 30 min for antigen retrieval. After blocking with 5% donkey serum, primary rabbit anti-rat insulin (Abcam, ab63820, 1:200), and mouse anti-rat glucagon (Abcam, ab10988, 1:200) antibodies were applied and incubated overnight at 4° C. After washing with PBS, Alexa Fluor 594-conjugated goat anti-rabbit IgG (Thermofisher, A11037, 1:400) and Alexa Fluor 488-conjugated donkey anti-mouse IgG (Thermofisher, A21202, 1:400) were applied and incubated for 60 min. Finally, slides were washed with PBS, applied with antifade/DAPI, and covered with glass coverslips.
BG Monitoring and Intraperitoneal Glucose Tolerance Test (IPGTT): Mouse BG levels were measured by a commercial glucometer (Contour Next EZ, Bayer) with a drop of blood collected from the tail vein. For the IPGTT, mice were fasted for 16 hours and an intraperitoneal injection of 20% glucose solution was administered at a dosage of 2 g glucose/kg mouse. BG levels were measured at 0, 15, 30, 60, 90, and 120 min following the injection.
Ex Vivo Static Glucose-Stimulated Insulin Secretion (GSIS) Assay: Krebs Ringer Bicarbonate (KRB) buffer was prepared as follows: 98.5 mM NaCl, 4.9 mM KCl, 2.6 mM CaCl2·2H20, 1.2 mM MgSO4·7H2O, 1.2 mM KH2PO4, 25.9 mM NaHCO3, 0.1% BSA (all from Sigma-Aldrich), and 20 mM HEPES (Gibco). The retrieved devices were incubated in the KRB buffer for 2 hours at 37° C., 5% CO2. Devices were transferred and incubated in KRB buffer supplemented with 3.3 mM glucose, then 16.7 mM glucose for 75 min each. The buffer was collected after each incubation step, and insulin concentration was measured using an ultrasensitive rat insulin ELISA kit (ALPCO).
Computational Modeling: Five general finite element models were created to calculate theoretical O2 profiles in SONIC-enabled constructs and corresponding controls. In all models, O2 tension (pO2) was related to the concentration of O2 (co
In other words, O2 partitioning was governed by Henry's law where, in Equation 1, αo
Model 1 (
Here, Do
Model 2 simulated steady state O2 transport in cylindrical constructs (4.2 mm diameter, 20.4 mm length) containing alginate-encapsulated INS-1 cells with cell densities from 1.0 to 8.0 million cells per mL alginate (
Above, R02 represents O2 consumption by the encapsulated INS-1 cells, modeled using Michaelis-Menten kinetics and a step-down function (An et al., “An Atmosphere-Breathing Refillable Biphasic Device for Cell Replacement Therapy,” Advanced Materials 31:1905135 (2019), which is hereby incorporated by reference in its entirety):
In Equation 4, VINS-1=5.0×10−17 mol/(m3 s cell) represents the literature-retrieved INS-1 cellular O2 consumption rate (Cline et al., “Rates of Insulin Secretion in INS-1 Cells are Enhanced by Coupling to Anaplerosis and Kreb's Cycle Flux Independent of ATP Synthesis,” Biochemical and Biophysical Research Communications 415:30-35 (2011), which is hereby incorporated by reference in its entirety), Km=0.81 mmHg represents the half-maximum constant derived from studies on mitochondrial respiration (Wilson et al., “The Oxygen Dependence of Mitochondrial Oxidative Phosphorylation Measured by a New Optical Method for Measuring Oxygen Concentration,” J Biol. Chem. 263:2712-2718 (1988), which is hereby incorporated by reference in its entirety), and ρ represents the cell density which was implemented at 2.5 million cells/mL to match experimental conditions (
Model 3 simulated steady state O2 transport in all cylindrical constructs (4.2 mm in diameter, 20.4 mm or 6.4 mm in length) containing alginate-encapsulated rat islets (
where α=0.40 and β=112.6. These values were obtained empirically and were found to be similar to distributions observed in other animal islet sources (Jo et al., “Size Distribution of Mouse Langerhans Islets,” Biophysical Journal 93:2655-2666 (2007), which is hereby incorporated by reference in its entirety). Human islet diameters were selected from Weibull distribution (Buchwald et al., “Quantitative Assessment of Islet Cell Products: Estimating the Accuracy of the Existing Protocol and Accounting for Islet Size Distribution,” Cell Transplant 18:1223-1235 (2009), which is hereby incorporated by reference in its entirety), with the probability density function given by:
where α=1.5 and β=105. In specified cases (
Steady state pO2 profiles were obtained by solving the diffusion-reaction mass balance equation (Equation 3). Solubility and diffusivity in the islets were given by αo
where Vislets=0.0340 mol/(m3 s) represents the O2 consumption rate in rat islets (Avgoustiniatos et al., “Measurements of the Effective Diffusion Coefficient of Oxygen in Pancreatic Islets,” Industrial & Engineering Chemistry Research 46:6157-6163 (2007), which is hereby incorporated by reference in its entirety) and Vislets=0.0134 mol/(m3 s) in human islets (Papas et al., “Human Islet Oxygen Consumption Rate and DNA Measurements Predict Diabetes Reversal in Nude Mice,” Am. J. Transplantation 7:707-713 (2007), which is hereby incorporated by reference in its entirety), respectively. Model 3 was used to predict islet oxygenation and necrosis in cylindrical constructs implanted intraperitoneally in mice (
Model 4 simulated steady state O2 transport in two thick cubic (6.6×6.6×6.6 mm) devices, each containing 500 IEQ rat islets (
Model 5 simulated steady state O2 transport in the SONIC spiral device and empty control containing variable loading densities of human islets. A constant pO2 of 40 mmHg was imposed on the top and bottom boundaries whereas a no-flux condition was imposed on the lateral face, as the modeled geometry is intended to represent only the central region of a device which would be extruded radially. All other physics implementations were identical to those of Model 3, except for the dimensions, which are defined in
All models were solved in COMSOL Multiphysics or COMSOL Livelink for MATLAB. In Models 3-5, all calculations were repeated for at least 3 iterations, whereby the islets were reselected and repositioned at random each time. For all calculations, a mesh was implemented using COMSOL's “Free Tetrahedral” program with the following settings: maximum element size of 100 μm, minimum element size of 1 μm, curvature factor of 0.3, resolution of narrow domains of 3.3, and maximum growth rate of 1.25. It was ensured that all results were independent of the mesh.
Statistics: All results are expressed as raw data or as mean±SD. Data from random BG measurements (
To develop a SONIC scaffold suitable for cell encapsulation and supporting rapid O2 transport, two structural features, difficult to achieve simultaneously, were developed. The first feature was an internal microstructure that is bi-continuous and non-wettable to provide unobstructed microchannels for O2 flow similar to the insect tracheae. The second feature was an external surface that is wettable to allow cell-laden hydrogel precursor to infiltrate. It was expected that a hydrophobic material for the scaffold skeleton would resist water infiltration. A two-step process was designed, forming first a superhydrophobic (i.e., low surface energy and high roughness) scaffold skeleton with continuous internal pores and then an adherent hydrophilic coating only on the external surface of the scaffold.
Accordingly, poly(vinylidene fluoride-co-hexafluoropropylene) (PVDF-HFP) (
Briefly, the SONIC scaffold was fabricated using the process illustrated in
Structural analysis confirmed that the desired macro- and micro-architecture (
To integrate the SONIC scaffold into a hydrogel-based cell encapsulation device (
Following surface hydrophilicity modification, a cell-laden hydrogel was applied via a simple in situ procedure. CaSO4 was deposited onto the SONIC scaffold surface as a crosslinker source (
Scanning electron microscopy (SEM)/energy dispersive X-ray spectroscopy (EDS) mapping was used to analyze the component distribution on the SONIC scaffold. Fluorine (F), nitrogen (N), and sulfur (S) were chosen as the specific elements to identify PVDF-HFP, polydopamine (PDA) and CaSO4, respectively (
A perfusion study was performed to test ability of the SONIC scaffold to prevent water from wicking into the internal pores. A cylindrical scaffold was inserted into a vial containing a water phase and a chloroform phase (
O2 distribution mapping was performed on an electron paramagnetic resonance (EPR) O2 imager to characterize O2 transport through the SONIC scaffold (
A computational model, developed to simulate spatial O2 transfer over time in this system, was in general agreement with experimental results. In silico, the SONIC scaffold distributed O2 more efficiently through the gelatin in comparison to the PLA control. Likewise, a clear radial O2 gradient was observed emanating from the SONIC sample, whereas only a top-down gradient was observed in the control PLA simulation (
The EPR measurement procedure was repeated with two additional control scaffolds chosen to verify the vital role of the air channels in facilitating high oxygen permeability. One control was a commercial porous sponge comprised of hydrophilic melamine, which was selected as a simple example to confirm that a liquid-filled porous material would not provide benefit to O2 transport. The second control was a PVDF-HFP scaffold, modified by the following two-step process to render the internal microporous channels hydrophilic: (1) ethanol was added to the tris buffer during incubation with dopamine to allow buffer solution penetration into the micropores of the scaffold, therefore enabling the application of the hydrophilic polydopamine coating within the microchannels; (2) the scaffold was treated with radio frequency plasma to further ensure its hydrophilicity. O2 transport tests showed that these two scaffolds were significantly slower than SONIC at equilibrating the system to exposed O2 levels. Spatial O2 distributions of the sample containing the hydrophilic porous PVDF-HFP showed a top-to-bottom pO2 gradient throughout the system until a steady state was achieved, rather than the radial gradient emanating from the SONIC insert (
In vitro tests and a complementary theoretical analysis were performed to evaluate the advantages of the SONIC scaffold for improving the viability of hydrogel encapsulated cells (
Results from the in vitro test were consistent with model predictions. Cell-containing devices were incubated in a hypoxic incubator with 5% CO2 and 5% O2 (i.e., 40 mmHg). After culturing for 48 hours, cell viability was evaluated using live/dead staining kit. Cells in the central region of the control device were dead likely due to hypoxia, with surviving cells limited to a thin region near the surface (
The computational model was adapted to simulate O2 profiles in SONIC devices, empty control devices, and control scaffold devices containing islets. In this model, 500 IEQ of rat islets were considered as discrete spheres, with diameters (30-300 μm) selected from a distribution-simulating the natural size heterogeneity of islets—and dispersed randomly within the alginate domain. Cross sectional surface plots show deficient pO2 levels in the center of the control device (
†The coefficient for the hydrophilic porous PVDF-HFP/alginate was calculated by the composition volume fraction-weighted average of the coefficients for PVDF-HFP and alginate with the assumption that the PVDF-HFP's interior microporous channels were filled with alginate.
Quantification of the oxygenation of the islet population in devices showed that the average islet pO2 in the SONIC device was 1.8-fold higher than that in the control device (
The simulation was repeated, but with the adaptation that each islet was implemented as 150 μm in diameter (the standard size for definition of an islet equivalent) to isolate the effect of islet distance from the surface. In the control device, it was observed that islets near the device surface were well oxygenated, while those in the center were anoxic (
The in vitro study and theoretical analysis performed here confirm the benefit of SONIC scaffold to improve O2 delivery to encapsulated cells and elucidates its mechanism of action. As previously mentioned, it is often suggested that islets must be within a few hundred micrometers to the device-host boundary to avoid debilitating O2 diffusion resistances (Dulong et al., “A Theoretical Study of Oxygen Transfer Including Cell Necrosis for the Design of a Bioartificial Pancreas,” Biotechnol. Bioeng. 96:990-998 (2007); Iwata et al., “Design of Bioartificial Pancreases From the Standpoint of Oxygen Supply,” Artif Organs 42:E168-E185 (2018), which are hereby incorporated by reference in their entirety). It was expected that the pO2 levels in the SONIC scaffold are near those available in the transplantation site (i.e., intraperitoneal cavity). The SONIC device was designed such that any islet is within 300 μm from the SONIC scaffold or the device-host boundary. It follows that every encapsulated islet is thus within 300 μm to a high pO2 source, regardless of its distance to the host site tissue. This not only permits the construction of thick devices, but also should allow the incorporation of significantly higher cell densities without significant impact on cell oxygenation, especially with human islets which feature lower O2 consumption rates (Papas et al., “Human Islet Oxygen Consumption Rate and DNA Measurements Predict Diabetes Reversal in Nude Mice,” Am. J. Transplantation 7:707-713 (2007), which is hereby incorporated by reference in its entirety), and mitigate negative outcomes should the external pO2 environment be lower. The computational analysis described herein, and corresponding in vitro tests, suggest that the SONIC device significantly improves cell survival in hydrogel-based devices.
Following in vitro testing and theoretical analysis, in vivo studies were pursued to test the therapeutic capability of the SONIC device (
Intraperitoneal glucose tolerance tests (IPGTT) were conducted on day 60 (
A transverse section of a retrieved device at 6 months confirmed that both the islets close to surface and deep within the structure were viable (
The foreign-body reaction (FBR) is a ubiquitous phenomenon for almost all foreign implants, which induces fibrosis deposition on the implant surface, occasionally leading to a complete coverage by the formation of a collagenous fibrotic capsule (Grainger D W., “All Charged up About Implanted Biomaterials,” Nat. Biotechnol. 31:507-509 (2013); Welch et al., “Antifibrotic Strategies for Medical Devices,” Adv. Drug Deliv. Rev. 167:109-120 (2020), which are hereby incorporated by reference in their entirety). Deposited fibrosis restricts the mass transfer at the implant/host interface and affects the viability of the encapsulated cells. Alginate is one of the most widely used materials for cell encapsulation due to its good biocompatibility. In addition, it has been reported that alginate capsules or fibers larger than 1.5 mm in diameter have a significantly mitigated fibrotic response (Watanabe et al., “Millimeter-Thick Xenoislet-Laden Fibers as Retrievable Transplants Mitigate Foreign Body Reactions for Long-Term Glycemic Control in Diabetic Mice,” Biomaterials 255:120162 (2020); Vlahos et al., “Muted Fibrosis from Protected Islets,” Nature Biomedical Engineering 2:791-792 (2018); Veiseh et al., “Size- and Shape-Dependent Foreign Body Immune Response to Materials Implanted in Rodents and Non-Human Primates,” Nat. Mater. 14:643-651 (2015), which are hereby incorporated by reference in their entirety). The diameter of this SONIC device is around 4 mm which is suggested to mitigate the fibrotic response. In acellular studies, minimal cellular overgrowth was observed on the retrieved device surface. For the islet-containing devices, most regions of the retrieved devices remained free of fibrosis (
Successful in vivo results of the ˜4 mm diameter device motivated the exploration of yet a thicker device design (
A 4-layer scaffold was then fabricated using a new printed mold to test the model predictions (
The preceding Examples describe the design and testing of an insect-inspired scaffold (SONIC) which features internal continuous air channels for rapid O2 delivery to hydrogel encapsulated cells. Incorporation of this scaffold into bulk hydrogels containing islets (the SONIC device) supported high islet viability and robust function even in devices with a thickness of 6.6 mm. The SONIC scaffold was comprised of a hydrophobic polymer, the fluoropolymer PVDF-HFP, and the internal continuous air channels were created by a phase separation process. A hydrophilic polydopamine coating was applied to the external scaffold surface to provide a compatible interface between the hydrophobic SONIC scaffold and hydrophilic hydrogel, while maintaining internal hydrophobicity to avoid water penetration into the air channels. The latter feature was verified as critical for enabling the high O2 permeability of the scaffold. Finally, the alginate hydrogel phase was structurally interlocked with the skeleton of the SONIC scaffold to prevent hydrogel detachment during retrieval of the transplanted device.
O2 supply is a primary limiting factor for cell encapsulation, especially for cells with high O2 consumption rates such as islets. To overcome this limitation, much attention has been deservedly focused on exogeneous O2 supply, such as O2-generating (Pedraza et al., “Preventing Hypoxia-Induced Cell Death in Beta Cells and Islets via Hydrolytically Activated, Oxygen-Generating Biomaterials,” Proc. Natl. Acad. Sci. USA 109:4245-4250 (2012); Wang et al., “An Inverse-Breathing Encapsulation System for Cell Delivery,” Science Advances 7:eabd5835 (2021); C A Patent Application No. 2924681 to Tempelman et al.; Wu et al., “In Situ Electrochemical Oxygen Generation with an Immunoisolation Device,” Annals of the New York Academy of Sciences 875:105-125 (1999), which are hereby incorporated by reference in their entirety) and O2-filling devices (Ludwig et al., “Favorable Outcome of Experimental Islet Xenotransplantation Without Immunosuppression in a Nonhuman Primate Model of Diabetes,” Proc. Natl. Acad. Sci. USA 114:11745-11750 (2017); Evron et al., “Long-Term Viability and Function of Transplanted Islets Macroencapsulated at High Density are Achieved by Enhanced Oxygen Supply,” Sci. Rep. 8:6508 (2018), which are hereby incorporated by reference in their entirety). However, O2 transport within the hydrogel-encapsulated cell component is driven by gradients of O2 tension and remains slow due to the poor O2 permeability in aqueous media. Few efforts have focused on improving O2 transport within the cell encapsulation domain itself. Some reports have explored the effect of perfluorocarbon (PFC) emulsion incorporation in hydrogels due to the high O2 solubility (Goh et al., “Limited Beneficial Effects of Perfluorocarbon Emulsions on Encapsulated Cells in Culture: Experimental and Modeling Studies,” J Biotechnol. 150:232-239 (2010); White et al., “Perfluorocarbons Enhance Oxygen Transport in Alginate-Based Hydrogels,” Polymers for Advanced Technologies 25:1242-1246 (2014), which are hereby incorporated by reference in their entirety), and slightly higher diffusivity (O'Brien et al., “Diffusion-Coefficients of Respiratory Gases in a Perfluorocarbon Liquid,” Science 217:153-155 (1982), which is hereby incorporated by reference in its entirety) in PFC compared to hydrogels. However, these systems generally only yield modest benefits because of the limited improvement in O2 permeability in composite systems where the phase with the higher permeability (in this case, PFC) is dispersed (Lewis A S., “Eliminating Oxygen Supply Limitations for Transplanted Microencapsulated Islets in the Treatment of Type 1 Diabetes,” Thesis (2008), which is hereby incorporated by reference in its entirety). The bicontinuous gas phase, endowed with extremely high permeability, incorporated into the hydrogel by the SONIC scaffold facilitates the rapid permeation of O2 throughout the device, thereby enormously improving the effective O2 permeability of the system. Regardless of design, the current SONIC system is reliant on the O2 available in the transplantation site; however, it is believed that the SONIC scaffold could be used to further enhance O2 supply to hydrogel encapsulated cells even in devices which provide exogeneous supply.
The mechanism of improved O2 distribution via SONIC can be visualized accordingly: the continuous hydrogel phase of the device can be considered as comprising small cubic regions of ˜600×600×600 μm (
A successful islet delivery implant must not only maintain cell survival but also ensure timely release of insulin to prevent postprandial hyperglycemia and overcorrection into hypoglycemia. The delay in IPGTT observed in mice treated with the thick SONIC device (
3D printing of the mold used to form the SONIC scaffold enables the scaffold to be fabricated in scaled-up dimensions (e.g., in multiple layers or extended in length to tens of centimeters) in a wide range of designs (e.g., toroidal, spiral). See, e.g.,
In summary, this invention provides a solution to the poor transport of O2 in traditionally employed bulk hydrogels of cell encapsulation systems. SONIC's mimicry of the insect tracheal system yields a cell encapsulation device that is amenable to increased cell density, fibrotic blockage, and—most notably—substantially increased device thickness without sacrificing cell oxygenation, in effect, decoupling cell survival from its distance to the external supply. Ultimately, these advantages imparted by the SONIC scaffold represent a promising platform for translatable encapsulation devices requiring high cell payloads.
Although preferred embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be made without departing from the spirit of the invention and these are therefore considered to be within the scope of the invention as defined in the claims which follow.
This application claims the priority benefit of U.S. Provisional Patent Application Ser. No. 63/174,739, filed Apr. 14, 2021, which is hereby incorporated by reference in its entirety.
This invention was made with government support under grant numbers 1R01DK105967-01A1 awarded by the National Institutes of Health and DGE-1650441 awarded by the National Science Foundation. The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2022/024800 | 4/14/2022 | WO |
Number | Date | Country | |
---|---|---|---|
63174739 | Apr 2021 | US |