US20070141164A1 - Methods and gel compositions for encapsulating living cells and organic molecules - Google Patents

Methods and gel compositions for encapsulating living cells and organic molecules Download PDF

Info

Publication number
US20070141164A1
US20070141164A1 US11/670,061 US67006107A US2007141164A1 US 20070141164 A1 US20070141164 A1 US 20070141164A1 US 67006107 A US67006107 A US 67006107A US 2007141164 A1 US2007141164 A1 US 2007141164A1
Authority
US
United States
Prior art keywords
hydrogel
isocyanate
thiol
prepolymer
biologic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/670,061
Inventor
Soonkap Hahn
Roberto Fagnani
Xiaofan Dong
Carl Edman
Pavel Tsinberg
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.)
Biocept Inc
Original Assignee
Biocept Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biocept Inc filed Critical Biocept Inc
Priority to US11/670,061 priority Critical patent/US20070141164A1/en
Publication of US20070141164A1 publication Critical patent/US20070141164A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/5436Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals with ligand physically entrapped within the solid phase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0046Sequential or parallel reactions, e.g. for the synthesis of polypeptides or polynucleotides; Apparatus and devices for combinatorial chemistry or for making molecular arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G18/00Polymeric products of isocyanates or isothiocyanates
    • C08G18/06Polymeric products of isocyanates or isothiocyanates with compounds having active hydrogen
    • C08G18/08Processes
    • C08G18/10Prepolymer processes involving reaction of isocyanates or isothiocyanates with compounds having active hydrogen in a first reaction step
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/33348Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing isocyanate group
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/334Polymers modified by chemical after-treatment with organic compounds containing sulfur
    • C08G65/3342Polymers modified by chemical after-treatment with organic compounds containing sulfur having sulfur bound to carbon and hydrogen
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N11/00Carrier-bound or immobilised enzymes; Carrier-bound or immobilised microbial cells; Preparation thereof
    • C12N11/02Enzymes or microbial cells immobilised on or in an organic carrier
    • C12N11/04Enzymes or microbial cells immobilised on or in an organic carrier entrapped within the carrier, e.g. gel or hollow fibres
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00351Means for dispensing and evacuation of reagents
    • B01J2219/00364Pipettes
    • B01J2219/00367Pipettes capillary
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • B01J2219/00527Sheets
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00596Solid-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00639Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium
    • B01J2219/00644Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium the porous medium being present in discrete locations, e.g. gel pads
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00677Ex-situ synthesis followed by deposition on the substrate
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00722Nucleotides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00725Peptides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/0074Biological products
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/0074Biological products
    • B01J2219/00743Cells
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G2210/00Compositions for preparing hydrogels
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L2203/00Applications
    • C08L2203/02Applications for biomedical use
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/10Libraries containing peptides or polypeptides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/14Apparatus specially adapted for use in combinatorial chemistry or with libraries for creating libraries

Definitions

  • the present invention relates to systems and methods for forming polyurethane hydrogels useful for encapsulating biologics, such as living cells, proteins, enzymes, antibodies and small organic molecules, and to the compositions which result therefrom. More specifically, the present invention relates to the formulation and use of a polymerization process employing biocompatible polymers and biocompatible polymerization conditions, such as neutral pH, where there is maintenance of an aqueous environment and preservation of physiologically relevant osmolarity throughout the polymerization process, as well as to bioassays utilizing such improved resultant products.
  • This invention represents a significant development in the art of encapsulation of certain materials and a significant advancement, from certain standpoints, of the process described in U.S. Pat. No. 6,174,683, which is assigned to the assignee of this application.
  • hydrogels are defined as water-containing polymeric matrices.
  • hydrogels provide a support for biomaterials that more closely resembles the native, aqueous, cellular environment, as opposed to a more denaturing environment that results when proteins or other materials are directly attached to a solid support surface using other molecular scale linkages, such as coatings.
  • hydrogels have previously been described as matrix supports for biomolecules and/or living cells, and these include alginates, alginates modified to permit cross-linking, acrylamide-based hydrogels, and polyethylene oxide-based hydrogels.
  • alginates alginates modified to permit cross-linking
  • acrylamide-based hydrogels alginates modified to permit cross-linking
  • polyethylene oxide-based hydrogels polyethylene oxide-based hydrogels.
  • many of the materials suitable for these gentle conditions e.g. alginate-based polymers, lack the structural requirements and/or biostability necessary for broad applications.
  • Alginate gels have been widely utilized for immobilization of eukaryotic cells and proteins. This form of hydrogel is generally benign and biocompatible during the encapsulation process; however, it can suffer from a lack of structural stability. Alginates are thus sometimes combined with multivalent cations to form more stable, ionically cross-linked gels. However, upon exposure to physiologically relevant buffers and environments, divalent cations tend to exchange with monovalent species, and the polymer often loses structural integrity. As a result, alginates are somewhat undesirable hydrogels for encapsulating biomolecules and living cells. Moreover, the overall manufacturability of alginate gels is difficult, further lessening the desirability and applicability of such a gel system.
  • Polyacrylamide hydrogel systems have also received considerable attention as matrices for attaching biomolecules and encapsulation vehicles.
  • Arenkov, et al. Anal. Biochem. 278, 123-131 (2000)
  • Proteins are then covalently linked to each gel pad following application to the micromatrix either by crosslinking with glutaraldehyde or by chemical modification of carbohydrate moieties present on select proteins to allow subsequent chemical linkage to the gel support.
  • such a method of linkage can be potentially very damaging to the integrity and/or activity of the protein, and it may also require the presence of sugar residues not ubiquitously found on all proteins.
  • polyacrylamide-based hydrogels are the use of those composed primarily of polyethylene oxide (PEO) polymerization units.
  • PEO polyethylene oxide
  • These polymers can offer a number of distinct advantages in the areas of biocompatibility, diffusion of small molecules and manufacturing process control.
  • the grafting of PEO onto serum albumin significantly reduces immunogencity of the native albumin (Abuchowski, et al. 1977).
  • Hubbell, et al. U.S. Pat. No. 5,573,934 and related patents teach the use of polyethylene glycol polymers for encapsulating cells using a dye-based photoinitiated free radical-based polymerization process.
  • initiation of polymerization requires the addition of a separate, photoactivatable catalyst and/or the addition of free radical-generating polymerization accelerators, separate and distinct from the polymer components or subunits.
  • Chudzik and Anderson teach the use of polymer initiator groups which are pendant from the polymerizable groups and thus avoid the separate addition of initiator components.
  • Wood, et al. teach the use of various cross-linking polymer systems, including a polyurethane-based hydrogel formed from isocyanate-functional prepolymers, to form a cross-linked polymer to encapsulate microbial cells (U.S. Pat. Nos. 4,436,813 and 4,732,851). Also described are methods using polyazetidine prepolymers and carboxymethylcellulose which can be crosslinked with polyvalent ions. Direct contact of isocyanates with the microbial cells which occurs in the encapsulation within such a polyurethane-based hydrogel and exposure to other potentially toxic conditions may not be suitable for the encapsulation of certain sensitive biological materials.
  • a polyurethane-based hydrogel prepolymer is used to simultaneously derivatize biomolecules, such as nucleic acid probes, within its structure during polymerization.
  • biomolecules such as nucleic acid probes
  • Such a polymerization process can use PEG-based prepolymers and is advantageous from its avoidance of free radicals or other agents as a result of its employ of water to initiate polymerization.
  • organic solvents are often employed in the prepolymer formation, derivatization and/or solubilization, the process may still be toxic to certain sensitive biological materials, such as living mammalian cells.
  • the polymerization process is truly biocompatible as it employs no organic solvents.
  • This novel process utilizes thiol-based crosslinkers which reduces the crosslinking of biomaterials within the hydrogel, thereby rendering the process capable of encapsulating and attaching such biological material in forms particularly suitable for diagnostic and therapeutic use, for example, microarrays of proteins or cells or other organic compounds for high-throughput testing.
  • the method of polymerization employs thiol-containing crosslinkers and selective reaction conditions, specifically neutral pH and aqueous buffers, to preferentially favor the reaction of sulfhydryl groups, as opposed to amines, as the preferred conjugation nucleophile where water is present during polymerization; this provides mild, non-radical reaction conditions that allow gentle encapsulation which is of particular importance to biomolecules and living cells.
  • the porosity of the encapsulating polymer can be advantageously varied, and the encapsulation process permits deposition or dispensing, onto glass slides or other surfaces, of discrete hydrogel droplets in a desired physical form, e.g.
  • the overall encapsulation/polymerization process of preparation comprises fewer steps than comparable methodologies, thereby simplifying and easing process development. Because the resultant hydrogel polymers can provide antibody or enzymatic arrays and viable cell encapsulation, the potential employment of such materials in bioreactors, biosensors, biochips and artificial organs is facilitated.
  • compositions containing such hydrogel encapsulated cells are expected to serve as a logical extension of bioassay development for complex biopathway screening, and encapsulated cells will be useful tools in bioreactors for economically generating complex therapeutic agents and materials.
  • encapsulated living cells may potentially serve as artificial organs or biosensors, responding as needed to altered or toxic environments.
  • Microarrays of encapsulated cells or other such biologics are also expected to be useful in high throughput biological testing.
  • FIG. 1 is a diagrammatic view showing a mechanism of cross-linking prepolymers.
  • FIG. 2 is a diagrammatic view, similar to FIG. 1 , of an alternative cross-linking reaction embodying various features of the present invention.
  • Water is often added to cure or initiate the crosslinking of isocyanate-functional prepolymers, particularly polyurethane prepolymers. This is in contrast to processes employing free radical-based methodology, e.g. UV-induced photopolymerization, that is used to generate reactive species suitable for forming covalent linkages between prepolymer units.
  • Molecules having isocyanate-functional groups are covalently linked to a polymer of choice to provide a prepolymer, and addition of water produces an active primary amine at a certain frequency by conversion of some isocyanate moieties, based upon temperature and pH.
  • Such primary amines subsequently react with other isocyanates attached to other prepolymer units, thereby covalently linking the prepolymer units together, as illustrated in FIG. 1 ; this is generally representative of certain reactions utilized in the '683 patent.
  • This process leads to the generation of a suitable optically transparent, urea-linkage-based hydrogel, so long as reactivity of the polyurethane prepolymer and reaction conditions are controlled to prevent gas bubble formation and/or precipitation of the polymer.
  • various biological entities or small molecules i.e. biologics, can be present or can be later added to create biologically active hydrogels.
  • biologics for purposes of this patent application, should be understood to include living cells, proteins, such as antibodies, other bioactive materials, both natural or synthetic, and small organic molecules which function bioactively. It can thus be seen that the size of a biologic may vary substantially, and as explained hereinafter, molecules of small size may desirably be provided with anchoring moieties.
  • the '683 patent describes the addition of primary amine-derivatized oligonucleotides to isocyanate-functional prepolymers in order to produce oligonucleotide arrays that are attached to a solid support surface.
  • An advantageous feature of such a process is that, during the completion of the polymerization reactions between isocyanate prepolymer units, the oligonucleotides will become covalently linked to the polymer matrix.
  • such a method based upon amine conjugation, may not be suitable for certain sensitive biologics, e.g. certain proteins and living cells. Because primary amines are components of all proteins including those present on the surface of living cells, e.g.
  • Thiol-based crosslinking agents serve as mediators of the cross-linking reaction between isocyanate groups on different prepolymers, as opposed to employing amine functionalities.
  • a neutral pH i.e. about 6.5 to 7.6
  • a neutral pH of about 6.5 to 7.5 is preferred; about 6.6 to 7.1 is more preferred; about 6.9 to 7.1 is still more preferred; and approximately pH 7.0 is most preferred.
  • the presence of such thiol-containing species will cross-link unreacted isocyanate groups so as to effectively carry out the polymerization process.
  • thiol crosslinkers are of course composed of a variety of amino acids, some of which contain side chain primary amines that are potentially reactive during the overall polymerization process. Linking to such amine functionalities may well hinder the natural movement and conformation of the protein, and in the case of living cells, it may likely alter the pattern and responsiveness of extracellular and plasma membrane proteins.
  • the present method avoids or substantially limits occurrence of such links and the negative aspects thereof.
  • the pKa value for the side chain primary amine of the amino acid lysine is quite basic, approximately 10.5, and that for arginine is even more basic, i.e. over 12. If the pH of the polymerization mixture is maintained approximately neutral, then the proportion of free amine suitable for participating in a nucleophilic addition, such as that shown in FIG. 1 , is less than 1/1000th of the total primary amine population represented by lysine side chains. In contrast, thiol groups remain nucleophilic and very reactive at neutral pH values.
  • cysteine residues in proteins contain a thiol side chain
  • the frequency of cysteines within proteins is generally more than 3-fold lower than that of lysine, and when present, cysteines are frequently oxidized so as to form intramolecular cysteine linkages in native proteins, thereby further lowering the number of available sulfhydryl groups.
  • the overall result is a very substantial reduction in the number of multiple, potentially denaturing links between embedded proteins or cells and the polymer matrix; thus, such thiol-mediated crosslinking of hydrogel prepolymers provides improved formulations for encapsulating sensitive biological molecules and living cells.
  • this encapsulation method which depends upon thiol reactions, also provides a very effective way of anchoring small organic molecules, for example organic molecules having a molecular weight between 100 and 2000 and particularly those having a molecular weight not greater than about 500, in a manner so that they fully retain their effectiveness in the hydrogel.
  • small organic molecules for example organic molecules having a molecular weight between 100 and 2000 and particularly those having a molecular weight not greater than about 500, in a manner so that they fully retain their effectiveness in the hydrogel.
  • These small molecules are derivatized to place a thiol group at a location in the molecule where it will not interfere with the secondary or tertiary spatial configuration of the small molecule, for example, at one end of a generally linear molecule.
  • the small molecule might be of such a size that it would not necessarily be retained in an encapsulating matrix of this type, the presence of the thiol group will result in a linking to an isocyanate group on the polymer and thus anchor the small organic molecule within or upon the gel in a manner such that it can assume its normal active configuration.
  • the encapsulation method can be used to create what might be termed chemical chips, as well as protein chips, cellular chips and the like.
  • Isocyanate-functional prepolymers are often prepared from relatively high molecular weight polyoxyalkylene diols or polyols that are reacted with difunctional or polyfunctional isocyanate compounds.
  • Preferred prepolymers are ones made from polyoxyalkylene diols or polyols that comprise homopolymers of ethylene oxide units or block or random copolymers containing mixtures of ethylene oxide units and propylene oxide or butylene oxide units. In the case of such block or random copolymers, at least 75% of the units are preferably ethylene oxide units.
  • Such polyoxyalkylene diol or polyol molecular weight is preferably from 2,000 to 30,000 and more preferably from 5,000 to 30,000.
  • Suitable prepolymers may be prepared by reacting selected polyoxyalkylene diols or polyols with polyisocyanate, at an isocyanate-to-hydroxyl ratio of about 1.2 to about 2.2, so that essentially all of the hydroxyl groups are capped with polyisocyanate.
  • Aliphatic, rather than aromatic isocyanates, are preferred as they provide more easily controlled polymerization.
  • polyethylene glycol (PEG), polypropylene glycol (PPG) or copolymers thereof are preferred.
  • the isocyanate-functional prepolymers being used preferably contain active isocyanates in an amount of about 0.1 meq/g to about 1 meq/g, and more preferably about 0.2 meq/g to about 0.8 meq/g.
  • relatively low molecular weight prepolymers e.g. less than 2,000
  • they preferably contain a relatively high isocyanate content (about 1 meq/g or even higher).
  • the polymerization rate of such smaller prepolymers may require more precise control to avoid too rapid polymerization, and thus would be less preferred for fabricating microarrays and the like.
  • prepolymers with a fairly high isocyanate content may have a relatively high content of free amines after polymerization, and the positive charges on such amine functionalities, at neutral pH, may increase non-specific binding of negatively charged biomolecules with the potential of resulting in higher levels of undesirable background signals.
  • higher molecular weight prepolymers which contain a relatively low isocyanate content are preferred.
  • prepolymer In order to enhance the diffusability of large biological molecules, it may be desirable to use low ratios of prepolymer (about 3-5 weight %) relative to the total volume of the ultimate formulation following the mixture of the components. Such relatively low percentages aid in producing hydrogel compositions having the desired porosity for use in assays, bioreactors and the like, and about 3.5 weight % of prepolymer in the ultimate formulation may be preferred.
  • Longer dithiol crosslinkers provide formulations for hydrogel polymerization that are more easily controlled.
  • Crosslinkers having a backbone of PEG and/or PPG units are one class of dithiols that provide biocompatibility and structural advantages, and such water-soluble crosslinkers of molecular weight between about 500 and 10,000 are preferred, with those between about 2,000 and 6,000 being more preferred and those between about 3,000 and 4,000 being most preferred.
  • the molar ratio of dithiol crosslinker to isocyanate (from the prepolymer) is preferably not higher than about 0.3 dithiol per isocyanate, and more preferably not higher than about 0.2 dithiol per isocyanate to provide a suitable reaction product.
  • formulations having a ratio slightly less than about 0.1 dithiol per isocyanate are preferably supplied with an auxiliary water-soluble bidentate crosslinker having two different isocyanate-reactive functional groups, one of which is preferably a thiol group, e.g. cysteine which has a side chain thiol group and a less reactive primary ⁇ -amine group, which is of course more nucleophilic than the ⁇ -carboxyl under these conditions.
  • bidentate crosslinkers that might be used include 2-mercaptoethanol, 2-aminoethanethiol, homocysteine, 2-mercaptopropanol and other short chain compounds having a thiol group and another nucleophilic group.
  • an auxiliary bidentate crosslinker can be advantageous in controlling the polymerization reaction in obtaining completion within desirable time limits and in obtaining hydrogel compositions that are stable, have a high water content and excellent structural strength. Accordingly, the employment of the combination of a dithiol crosslinker of relatively high molecular weight, e.g.
  • auxiliary crosslinker When such an auxiliary crosslinker is used, it is generally used in a molar amount from about 1 to 3 times the molar amount of the dithiol, and preferably from about 1.5 to about 2.5 times the moles of the dithiol crosslinker, in which amount it has been found to moderate the polymerization reaction and result in satisfactory curing.
  • prepolymers of this general type allows the use of chemically functional surfaces to also achieve covalent attachment of the polymer to a substrate during polymerization.
  • Such surfaces may be provided upon substrates which will facilitate the handling and instrumented examination of the polymerized hydrogel and encapsulated biological matter; for example, fabrication of a microarray containing different bioactive material or biologics encapsulated into individual spots or regions of polymerized hydrogel placed in a known pattern on such a substrate.
  • Neutral pH is preferably maintained throughout the polymerization process by the use of 50 mM phosphate buffer supplemented with NaCl, typically 10 mM to 80 mM; osmotic pressure is preferably maintained at physiological levels, approximately 300 milliosmoles. It is found that such formulations can be made without using organic solvents by mixing isocyanate-derivatized prepolymers rapidly in phosphate buffer/NaCl and then rapidly adding a premixed solution of cells or proteins and water-soluble dithiol crosslinker in phosphate buffer/NaCl.
  • the polymerization process will then generally occur within 20 to 60 minutes, typically less than 30 minutes, during which time the cells or proteins remain in a hydrated and an osmotically balanced state at physiological pH in the resultant reaction product.
  • pH and osmolality are maintained between 6.9 to 7.6 and between 250 to 400 mOsm/kg, respectively.
  • thiol-crosslinked polyurethane hydrogels are suitable for encapsulating a wide range of biologics.
  • One criteria for the success of encapsulation of living cells is an assessment of continued cell viability, and typically, trypan blue exclusion is a technique frequently favored by biologists to easily determine cell viability.
  • trypan blue exclusion is a technique frequently favored by biologists to easily determine cell viability.
  • the hydrogel absorbs a significant amount of the trypan blue dye, determination of cell color and intensity of the intracellular dye using this method was unreliable.
  • AlamarBlueTM (Trek Diagnostic Systems, Inc.) was used. AlamarBlue is a dye that becomes fluorescent upon reduction by metabolic processes.
  • Goat lymphocytes having been determined by trypan blue exclusion to have both viable and dead cells present within the cell mixture, were chosen, and the prepolymer and cell suspension/dithiol crosslinker/bidentate crosslinker were mixed.
  • the lymphocytes became encapsulated within the thiol-crosslinked hydrogel when droplets of the mixture were deposited as spots (approximately 300-1,000 microns in diameter, with a height equal to or greater than 20 microns) upon glass slides and then cured in a high humidity chamber at room temperature; mixing and curing took just less than 20 minutes.
  • the relative humidity (RH) is at least about 90% and more preferably is about 95% or higher.
  • Protein encapsulation was also examined, formulating the gel essentially as just described. Protein encapsulation was demonstrated by the sequestration/encapsulation of anti-transferrin antibody within the gel matrix during polymerization. Verification of the antibody's functionality was demonstrated by the specific binding of fluorescent dye-labeled transferrin to those sites containing the anti-transferrin antibody and not at other sites containing different antibodies or no antibodies.
  • encapsulated living cells and/or proteins within such a thiol-crosslinked hydrogel might be deposited in pre-identified areas as spots or regions upon support surfaces, such as glass slides, or within microwells or microchambers, such as would be present in standard 96 well, 384 well or 1536 well microtiter plates. Distinct advantages are present with both approaches. In repeatedly depositing a number of discrete spots upon a single surface, each spot might contain a different entity, allowing a single incubation followed by the supply of wash solutions to contact all spots simultaneously. The use of individual microchambers would allow robotic handling of the plates and permit the use of low volumes of individual test solutions at each well. Combinations of these two approaches may also be used whereby individual chambers, arranged in a standard 96 well array or similar format, are each supplied with one or more hydrogel spots containing different entities.
  • Devices employing such arrays might be employed as combinatorial chemical or drug screening devices, antibody arrays, peptide arrays, cell arrays, enzymatic activity arrays, or DNA or other polynucleotide arrays that will be selective for binding to related proteins or other biomolecules.
  • encapsulated cells or biomolecules coated onto the walls of microcapillary tubes will function as flow-through devices having single or multiple channels, which might be employed as screening devices or as biosensors on systems, such as in liquid chromatography or in “lab-on-a-chip” devices.
  • Signal readout from such devices might be via binding of fluorescent proteins or of antigens, to be measured by subsequent antibody-based detection methods (possibly employing additional arrays), or via reaction with endogenous biopathways which will result in the formation of a detectable species, e.g. enzymatic conversion of a substrate to a fluorescent dye molecule, or change in the electrical properties, e.g. conductivity, of the cell and/or surrounding matrix resulting from exposure to the specific agent.
  • a detectable species e.g. enzymatic conversion of a substrate to a fluorescent dye molecule
  • change in the electrical properties, e.g. conductivity, of the cell and/or surrounding matrix resulting from exposure to the specific agent e.g. enzymatic conversion of a substrate to a fluorescent dye molecule
  • the addition of either a redox agent to the gel or the addition of an electrically conductive polymer may enable signal detection by electrical, non-photonic, means.
  • Solution A was prepared by mixing 0.075 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1.5 mL of 50 mM aqueous phosphate buffer, at pH 7.0 with 80 mM sodium chloride.
  • Solution C was prepared by mixing 100 ⁇ L of Solution B with 10 ⁇ L of goat lymphocytes in Dulbecco's phosphate-buffered saline.
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride.
  • Solution B was prepared by the same procedure as in Example 1.
  • Solution C was prepared by mixing 40 ⁇ L of Solution B with 70 ⁇ L of goat lymphocytes in Dulbecco's phosphate-buffered saline. Finally, 100 ⁇ L of Solution A was mixed with 100 ⁇ L of Solution C, and the resulting solution was microspotted using the same procedure as in Example 1. The formulation polymerized in 5 minutes, and the hydrogel spots were treated with Dulbecco's modified phosphate-buffered saline solution and incubated at 37° C. for 1 day to 3 days in RPM1640 cell media. The viability of lymphocytes was examined with a light microscope using AlamarBlue which demonstrated viable encapsulated cells.
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride.
  • Solution B was prepared by the same procedure as in Example 1.
  • Solution C was prepared by mixing 40 ⁇ L of Solution B with 70 ⁇ L of E. coli in Dulbecco's phosphate-buffered saline. Finally, 100 ⁇ L of Solution A was mixed with 100 ⁇ L of Solution C, and the resulting solution was placed into a disposable culture tube. The formulation polymerized in 5 minutes, and the hydrogel was treated with Dulbecco's modified phosphate-buffered saline solution and incubated at 37° C. for 1 day in RPM1640 cell media. Viability and growth of E. coli were confirmed by observing turbidity in the hydrogel after 1 day.
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride.
  • the slide was treated with a PBS buffer containing 1% Bovine Serum Albumin (Sigma Chemical Co.) and 0.1% Triton X-100 (Boehringer Mannheim) for 1 hour at room temperature.
  • Anti-transferrin in the hydrogel was interacted with Cy3-labeled transferrin (1 ⁇ g/ml in 1% bovine serum albumin, 0.1% triton X-100 in phosphate buffered saline) for 1 hour and then visualized; it demonstrated that there was specific binding of fluorescent dye-labeled transferrin at sites containing the anti-transferrin antibody and not at other sites containing different antibodies or no antibodies.
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer at pH 7.0.
  • Solution B without salts was prepared by the same procedure as in Example 1.
  • hydrogel microbeads may be formed which encapsulate biologics.
  • the polymer/cell (or protein) mix is added to a non-miscible liquid, such as an oil, while curing is occurring to cause microbeads of various dimensions to be formed.
  • Separation from the oil or other suspending liquid yields a slurry of beads suitable for use in bioreactors, assay devices, artificial organs, biosensors or the like.
  • multiple layers of encapsulated cells, proteins or other bioactive molecules might be used to construct complex materials having unique overall properties.
  • dyes or other agents might be added to the encapsulating polymer to facilitate subsequent identification of the encapsulated cell type if heterogeneous mixtures of cells are to be employed.
  • Such a cell identification mechanism combined with a chromaphore-based or fluorescent-based response from specific cells in response to added agents, e.g. expression of green fluorescent protein in response to specific cell signaling pathway activation by a ligand or drug, permits the screening of large populations of heterogeneous cells in a rapid and facile fashion.

Abstract

A method for encapsulating biologics within a hydrogel by using an aqueous solution of an isocyanate-functional polyurethane prepolymer which is mixed with an amount of biologics and an aqueous solution containing a dithiol crosslinking agent under physiological pH conditions to create a hydrogel. An additional bidentate crosslinking agent is optionally included. The product of such method may be a bioreactor or an assay device having a plurality of different biologics encapsulated at predetermined locations in a substrate.

Description

  • This application is a division of U.S. Ser. No. 10/398,725, which was a §371 of PCT/US2/10411, filed Apr. 2, 2002, which claims priority from U.S. Provisional Patent Application Ser. No. 60/281,268, filed Apr. 3, 2001, the disclosures of all of which are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to systems and methods for forming polyurethane hydrogels useful for encapsulating biologics, such as living cells, proteins, enzymes, antibodies and small organic molecules, and to the compositions which result therefrom. More specifically, the present invention relates to the formulation and use of a polymerization process employing biocompatible polymers and biocompatible polymerization conditions, such as neutral pH, where there is maintenance of an aqueous environment and preservation of physiologically relevant osmolarity throughout the polymerization process, as well as to bioassays utilizing such improved resultant products. This invention represents a significant development in the art of encapsulation of certain materials and a significant advancement, from certain standpoints, of the process described in U.S. Pat. No. 6,174,683, which is assigned to the assignee of this application.
  • DESCRIPTION OF PRIOR ART
  • The use of enzymes, antibodies, peptides, or other bioactive molecules, e.g. aptamers, has received increasing attention as tools for screening in the fields of bioassays and proteomics. As part of this development, the use of hydrogel supports for these bioactive materials has also gained in importance. Hydrogels are defined as water-containing polymeric matrices. In particular, hydrogels provide a support for biomaterials that more closely resembles the native, aqueous, cellular environment, as opposed to a more denaturing environment that results when proteins or other materials are directly attached to a solid support surface using other molecular scale linkages, such as coatings.
  • Certain hydrogels have previously been described as matrix supports for biomolecules and/or living cells, and these include alginates, alginates modified to permit cross-linking, acrylamide-based hydrogels, and polyethylene oxide-based hydrogels. In general, however, there is frequently difficulty in reconciling the gel polymerization and encapsulation requirements with the gentle conditions requisite for maintaining the viability or activity of live cells or certain active proteins. In addition, many of the materials suitable for these gentle conditions, e.g. alginate-based polymers, lack the structural requirements and/or biostability necessary for broad applications.
  • Alginate gels have been widely utilized for immobilization of eukaryotic cells and proteins. This form of hydrogel is generally benign and biocompatible during the encapsulation process; however, it can suffer from a lack of structural stability. Alginates are thus sometimes combined with multivalent cations to form more stable, ionically cross-linked gels. However, upon exposure to physiologically relevant buffers and environments, divalent cations tend to exchange with monovalent species, and the polymer often loses structural integrity. As a result, alginates are somewhat undesirable hydrogels for encapsulating biomolecules and living cells. Moreover, the overall manufacturability of alginate gels is difficult, further lessening the desirability and applicability of such a gel system.
  • Polyacrylamide hydrogel systems have also received considerable attention as matrices for attaching biomolecules and encapsulation vehicles. For example, Arenkov, et al. (Anal. Biochem. 278, 123-131 (2000)), describe gel pad arrays formed by photoinitiated polymerization of acrylamide/bisacrylamide mixtures using methylene blue as the photocatalyst. Proteins are then covalently linked to each gel pad following application to the micromatrix either by crosslinking with glutaraldehyde or by chemical modification of carbohydrate moieties present on select proteins to allow subsequent chemical linkage to the gel support. However, such a method of linkage can be potentially very damaging to the integrity and/or activity of the protein, and it may also require the presence of sugar residues not ubiquitously found on all proteins.
  • Alternative to the use of polyacrylamide-based hydrogels are the use of those composed primarily of polyethylene oxide (PEO) polymerization units. These polymers can offer a number of distinct advantages in the areas of biocompatibility, diffusion of small molecules and manufacturing process control. For example, the grafting of PEO onto serum albumin significantly reduces immunogencity of the native albumin (Abuchowski, et al. 1977). Hubbell, et al. (U.S. Pat. No. 5,573,934 and related patents) teach the use of polyethylene glycol polymers for encapsulating cells using a dye-based photoinitiated free radical-based polymerization process.
  • In the aforementioned polyacrylamide or PEG polymeric gels, initiation of polymerization requires the addition of a separate, photoactivatable catalyst and/or the addition of free radical-generating polymerization accelerators, separate and distinct from the polymer components or subunits. Chudzik and Anderson (U.S. Pat. No. 6,156,345) teach the use of polymer initiator groups which are pendant from the polymerizable groups and thus avoid the separate addition of initiator components.
  • Mixed polymer/alginate systems have also been devised to overcome limitations inherent in each system alone. For instance, Desai, et al. (U.S. Pat. No. 5,334,640) employ mixtures of an ionically cross-linked biocompatible component with a covalently linked component. However, the overall process remains dependent upon photoinitiated, free radical-based polymerization.
  • Use of methodologies incorporating free radicals as essential elements within such a process is a generally undesirable feature of many of the encapsulation/polymerization techniques in present use. For example, in cell encapsulation with acrylamide gels, “polymerization of acrylamide generates heat and free radicals, causing loss of in the chemiosmotic integrity and enzymatic activity of the immobilized cells” (see Poncelet De Smet, et al. in “Fundamentals of Animal Cell Encapsulation and Immobilization”, Mattheus F. A. Goosen, editor, CRC Press, Boca Raton, Fla., 1993, p. 301). It is therefore desirable to provide a polymerization process which does not use free radicals to initiate polymerization, thereby avoiding potential harm to encapsulated cells and biomolecules. It is also desirable to utilize polymers which have both structural and mechanical durability in biological situations and uses, particularly ones which are truly biocompatible, i.e. non-toxic to the encapsulated biomolecule or cell and to the surrounding media or host.
  • Wood, et al. teach the use of various cross-linking polymer systems, including a polyurethane-based hydrogel formed from isocyanate-functional prepolymers, to form a cross-linked polymer to encapsulate microbial cells (U.S. Pat. Nos. 4,436,813 and 4,732,851). Also described are methods using polyazetidine prepolymers and carboxymethylcellulose which can be crosslinked with polyvalent ions. Direct contact of isocyanates with the microbial cells which occurs in the encapsulation within such a polyurethane-based hydrogel and exposure to other potentially toxic conditions may not be suitable for the encapsulation of certain sensitive biological materials.
  • In the '683 patent, a polyurethane-based hydrogel prepolymer is used to simultaneously derivatize biomolecules, such as nucleic acid probes, within its structure during polymerization. Such a polymerization process can use PEG-based prepolymers and is advantageous from its avoidance of free radicals or other agents as a result of its employ of water to initiate polymerization. However, because organic solvents are often employed in the prepolymer formation, derivatization and/or solubilization, the process may still be toxic to certain sensitive biological materials, such as living mammalian cells.
  • In brief, there remains a particular need for truly benign, non-toxic, biocompatible and mechanically robust hydrogel polymers and associated polymerization methodology in order to encapsulate certain biologics, such as sensitive proteins, enzymes, antibodies and living cells, in a useful and economically feasible fashion, which can provide products that are well suited for assays and other applications.
  • SUMMARY OF THE INVENTION
  • It is an object of the invention to provide a method for biocompatible polymerization of isocyanate-modified biocompatible macromers to either directly or indirectly encapsulate or coat biologics, i.e. living cells, proteins, nucleic acids and other bioactive materials and compounds, including small organic molecules. The polymerization process is truly biocompatible as it employs no organic solvents. This novel process utilizes thiol-based crosslinkers which reduces the crosslinking of biomaterials within the hydrogel, thereby rendering the process capable of encapsulating and attaching such biological material in forms particularly suitable for diagnostic and therapeutic use, for example, microarrays of proteins or cells or other organic compounds for high-throughput testing.
  • The method of polymerization employs thiol-containing crosslinkers and selective reaction conditions, specifically neutral pH and aqueous buffers, to preferentially favor the reaction of sulfhydryl groups, as opposed to amines, as the preferred conjugation nucleophile where water is present during polymerization; this provides mild, non-radical reaction conditions that allow gentle encapsulation which is of particular importance to biomolecules and living cells. The porosity of the encapsulating polymer can be advantageously varied, and the encapsulation process permits deposition or dispensing, onto glass slides or other surfaces, of discrete hydrogel droplets in a desired physical form, e.g. spots or layers that encapsulate cells, proteins or other organic molecules, either directly or indirectly through binding agents, or alternatively by forming droplets or spheres that separately encapsulate such biologics. Moreover, the overall encapsulation/polymerization process of preparation comprises fewer steps than comparable methodologies, thereby simplifying and easing process development. Because the resultant hydrogel polymers can provide antibody or enzymatic arrays and viable cell encapsulation, the potential employment of such materials in bioreactors, biosensors, biochips and artificial organs is facilitated. Compositions containing such hydrogel encapsulated cells are expected to serve as a logical extension of bioassay development for complex biopathway screening, and encapsulated cells will be useful tools in bioreactors for economically generating complex therapeutic agents and materials. In addition, encapsulated living cells may potentially serve as artificial organs or biosensors, responding as needed to altered or toxic environments. Microarrays of encapsulated cells or other such biologics are also expected to be useful in high throughput biological testing.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a diagrammatic view showing a mechanism of cross-linking prepolymers.
  • FIG. 2 is a diagrammatic view, similar to FIG. 1, of an alternative cross-linking reaction embodying various features of the present invention.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • Water is often added to cure or initiate the crosslinking of isocyanate-functional prepolymers, particularly polyurethane prepolymers. This is in contrast to processes employing free radical-based methodology, e.g. UV-induced photopolymerization, that is used to generate reactive species suitable for forming covalent linkages between prepolymer units. Molecules having isocyanate-functional groups are covalently linked to a polymer of choice to provide a prepolymer, and addition of water produces an active primary amine at a certain frequency by conversion of some isocyanate moieties, based upon temperature and pH. Such primary amines subsequently react with other isocyanates attached to other prepolymer units, thereby covalently linking the prepolymer units together, as illustrated in FIG. 1; this is generally representative of certain reactions utilized in the '683 patent. This process leads to the generation of a suitable optically transparent, urea-linkage-based hydrogel, so long as reactivity of the polyurethane prepolymer and reaction conditions are controlled to prevent gas bubble formation and/or precipitation of the polymer. During such a polymerization process, various biological entities or small molecules, i.e. biologics, can be present or can be later added to create biologically active hydrogels. The term biologics, for purposes of this patent application, should be understood to include living cells, proteins, such as antibodies, other bioactive materials, both natural or synthetic, and small organic molecules which function bioactively. It can thus be seen that the size of a biologic may vary substantially, and as explained hereinafter, molecules of small size may desirably be provided with anchoring moieties.
  • The '683 patent describes the addition of primary amine-derivatized oligonucleotides to isocyanate-functional prepolymers in order to produce oligonucleotide arrays that are attached to a solid support surface. An advantageous feature of such a process is that, during the completion of the polymerization reactions between isocyanate prepolymer units, the oligonucleotides will become covalently linked to the polymer matrix. However, such a method, based upon amine conjugation, may not be suitable for certain sensitive biologics, e.g. certain proteins and living cells. Because primary amines are components of all proteins including those present on the surface of living cells, e.g. ligand receptor proteins, ion channel proteins and cell-to-cell adhesion proteins, extensive derivatization or conjugation of such amines directly to the isocyanate-functional prepolymer may lead to the protein's inactivation, denaturation or alteration of its functionality. It has now been found that this potential difficulty is minimized as a result of employing a new crosslinking approach that relies primarily upon thiol groups, instead of amines, for this purpose.
  • Thiol-based crosslinking agents serve as mediators of the cross-linking reaction between isocyanate groups on different prepolymers, as opposed to employing amine functionalities. Of course, in any aqueous environment required for forming a hydrogel, a certain percentage of the isocyanate groups will undergo hydrolysis; however, the primary amines formed as a result will have pKa values in the range of 9 to 10. By maintaining a neutral pH, i.e. about 6.5 to 7.6, the vast majority of these amines will be protonated and therefore will not participate in the polymerization process. A neutral pH of about 6.5 to 7.5 is preferred; about 6.6 to 7.1 is more preferred; about 6.9 to 7.1 is still more preferred; and approximately pH 7.0 is most preferred. The presence of such thiol-containing species will cross-link unreacted isocyanate groups so as to effectively carry out the polymerization process.
  • One advantageous result of such preferential use of thiol crosslinkers is the minimization of reactions with the biologics being encapsulated or immobilized at locations on the molecule where its attachment to the polymer matrix is undesirable. Control of the pH of the polymerization reaction, which places a restraint upon the nucleophilic reactivity of the amines but not that of the thiol groups, avoids potential creation of extensive links to proteins within the matrix. Proteins are of course composed of a variety of amino acids, some of which contain side chain primary amines that are potentially reactive during the overall polymerization process. Linking to such amine functionalities may well hinder the natural movement and conformation of the protein, and in the case of living cells, it may likely alter the pattern and responsiveness of extracellular and plasma membrane proteins. The present method avoids or substantially limits occurrence of such links and the negative aspects thereof.
  • For example, the pKa value for the side chain primary amine of the amino acid lysine is quite basic, approximately 10.5, and that for arginine is even more basic, i.e. over 12. If the pH of the polymerization mixture is maintained approximately neutral, then the proportion of free amine suitable for participating in a nucleophilic addition, such as that shown in FIG. 1, is less than 1/1000th of the total primary amine population represented by lysine side chains. In contrast, thiol groups remain nucleophilic and very reactive at neutral pH values. Although cysteine residues in proteins contain a thiol side chain, the frequency of cysteines within proteins is generally more than 3-fold lower than that of lysine, and when present, cysteines are frequently oxidized so as to form intramolecular cysteine linkages in native proteins, thereby further lowering the number of available sulfhydryl groups. The overall result is a very substantial reduction in the number of multiple, potentially denaturing links between embedded proteins or cells and the polymer matrix; thus, such thiol-mediated crosslinking of hydrogel prepolymers provides improved formulations for encapsulating sensitive biological molecules and living cells.
  • In addition, this encapsulation method, which depends upon thiol reactions, also provides a very effective way of anchoring small organic molecules, for example organic molecules having a molecular weight between 100 and 2000 and particularly those having a molecular weight not greater than about 500, in a manner so that they fully retain their effectiveness in the hydrogel. These small molecules are derivatized to place a thiol group at a location in the molecule where it will not interfere with the secondary or tertiary spatial configuration of the small molecule, for example, at one end of a generally linear molecule. Although the small molecule might be of such a size that it would not necessarily be retained in an encapsulating matrix of this type, the presence of the thiol group will result in a linking to an isocyanate group on the polymer and thus anchor the small organic molecule within or upon the gel in a manner such that it can assume its normal active configuration. In this manner, the encapsulation method can be used to create what might be termed chemical chips, as well as protein chips, cellular chips and the like.
  • Isocyanate-functional prepolymers are often prepared from relatively high molecular weight polyoxyalkylene diols or polyols that are reacted with difunctional or polyfunctional isocyanate compounds. Preferred prepolymers are ones made from polyoxyalkylene diols or polyols that comprise homopolymers of ethylene oxide units or block or random copolymers containing mixtures of ethylene oxide units and propylene oxide or butylene oxide units. In the case of such block or random copolymers, at least 75% of the units are preferably ethylene oxide units. Such polyoxyalkylene diol or polyol molecular weight is preferably from 2,000 to 30,000 and more preferably from 5,000 to 30,000. Suitable prepolymers may be prepared by reacting selected polyoxyalkylene diols or polyols with polyisocyanate, at an isocyanate-to-hydroxyl ratio of about 1.2 to about 2.2, so that essentially all of the hydroxyl groups are capped with polyisocyanate. Aliphatic, rather than aromatic isocyanates, are preferred as they provide more easily controlled polymerization. Generally, polyethylene glycol (PEG), polypropylene glycol (PPG) or copolymers thereof are preferred. The isocyanate-functional prepolymers being used preferably contain active isocyanates in an amount of about 0.1 meq/g to about 1 meq/g, and more preferably about 0.2 meq/g to about 0.8 meq/g. Should relatively low molecular weight prepolymers, e.g. less than 2,000, be used, they preferably contain a relatively high isocyanate content (about 1 meq/g or even higher). However, the polymerization rate of such smaller prepolymers may require more precise control to avoid too rapid polymerization, and thus would be less preferred for fabricating microarrays and the like. Moreover, prepolymers with a fairly high isocyanate content may have a relatively high content of free amines after polymerization, and the positive charges on such amine functionalities, at neutral pH, may increase non-specific binding of negatively charged biomolecules with the potential of resulting in higher levels of undesirable background signals. Thus, higher molecular weight prepolymers which contain a relatively low isocyanate content are preferred.
  • In order to enhance the diffusability of large biological molecules, it may be desirable to use low ratios of prepolymer (about 3-5 weight %) relative to the total volume of the ultimate formulation following the mixture of the components. Such relatively low percentages aid in producing hydrogel compositions having the desired porosity for use in assays, bioreactors and the like, and about 3.5 weight % of prepolymer in the ultimate formulation may be preferred. As mentioned above, the viability of entrapped biological molecules is enhanced through minimization of the involvement of amine groups by employing crosslinkers with thiol functions and maintaining a physiological pH of about 7.0, where a large percent of amines (pKa=10) will be present as protonated species which do not react with the isocyanate functionalities. Although such an arrangement in some instances could potentially result in incomplete curing of the prepolymer, the nucleophilic activity of thiols towards isocyanates is unaffected at such pH so curing can be completed, and the overall result is one of a significant advancement in formulating PEG and/or PPG-based hydrogels for encapsulating biomaterials.
  • Short-chain dithiol crosslinkers, such as 1,4-dithiothreitol (mw=154), produce a fairly high speed polymerization that needs to be slowed and carefully controlled to avoid precipitation. Longer dithiol crosslinkers provide formulations for hydrogel polymerization that are more easily controlled. Crosslinkers having a backbone of PEG and/or PPG units are one class of dithiols that provide biocompatibility and structural advantages, and such water-soluble crosslinkers of molecular weight between about 500 and 10,000 are preferred, with those between about 2,000 and 6,000 being more preferred and those between about 3,000 and 4,000 being most preferred. For example, PEG-(thiol)2 (Shearwater Polymers, Inc.) having a mw=3,400 and thiol groups at the ends of the chains, may be used with Hypol PreMa G-50 (Hampshire Chemical Corp., which has an aliphatic isocyanate content of 0.35 meq/g), and by varying the ratio between two such starting materials, it was found that the speed of polymerization can generally be effectively controlled at pH 7.0. The molar ratio of dithiol crosslinker to isocyanate (from the prepolymer) is preferably not higher than about 0.3 dithiol per isocyanate, and more preferably not higher than about 0.2 dithiol per isocyanate to provide a suitable reaction product.
  • Formulations having a ratio significantly lower than 0. 1 dithiol per isocyanate, e.g. 0.05 or below, might not polymerize promptly and/or completely at pH 7.0 without the inclusion of an auxiliary crosslinker. Thus, formulations having a ratio slightly less than about 0.1 dithiol per isocyanate are preferably supplied with an auxiliary water-soluble bidentate crosslinker having two different isocyanate-reactive functional groups, one of which is preferably a thiol group, e.g. cysteine which has a side chain thiol group and a less reactive primary α-amine group, which is of course more nucleophilic than the α-carboxyl under these conditions. Other bidentate crosslinkers that might be used include 2-mercaptoethanol, 2-aminoethanethiol, homocysteine, 2-mercaptopropanol and other short chain compounds having a thiol group and another nucleophilic group. Moreover, even when an adequate amount of the dithiol crosslinker is provided, it has been found that the provision of an auxiliary bidentate crosslinker can be advantageous in controlling the polymerization reaction in obtaining completion within desirable time limits and in obtaining hydrogel compositions that are stable, have a high water content and excellent structural strength. Accordingly, the employment of the combination of a dithiol crosslinker of relatively high molecular weight, e.g. about 2,000 to 6,000 mw, and a bidentate crosslinker of much lower molecular weight, preferably below about 300 mw, is preferred. It was found that this addition of a moderating bidentate crosslinker having two different reactive groups (e.g. cysteine) provides a novel and powerful means by which polymerization can be effectively controlled, and such is diagrammatically illustrated in FIG. 2, which also indicates that crosslinking in this manner eliminates a large amount of CO2 that would otherwise be created in normal crosslinking. When such an auxiliary crosslinker is used, it is generally used in a molar amount from about 1 to 3 times the molar amount of the dithiol, and preferably from about 1.5 to about 2.5 times the moles of the dithiol crosslinker, in which amount it has been found to moderate the polymerization reaction and result in satisfactory curing.
  • The inherent reactivity of prepolymers of this general type allows the use of chemically functional surfaces to also achieve covalent attachment of the polymer to a substrate during polymerization. Such surfaces may be provided upon substrates which will facilitate the handling and instrumented examination of the polymerized hydrogel and encapsulated biological matter; for example, fabrication of a microarray containing different bioactive material or biologics encapsulated into individual spots or regions of polymerized hydrogel placed in a known pattern on such a substrate.
  • Neutral pH is preferably maintained throughout the polymerization process by the use of 50 mM phosphate buffer supplemented with NaCl, typically 10 mM to 80 mM; osmotic pressure is preferably maintained at physiological levels, approximately 300 milliosmoles. It is found that such formulations can be made without using organic solvents by mixing isocyanate-derivatized prepolymers rapidly in phosphate buffer/NaCl and then rapidly adding a premixed solution of cells or proteins and water-soluble dithiol crosslinker in phosphate buffer/NaCl. The polymerization process will then generally occur within 20 to 60 minutes, typically less than 30 minutes, during which time the cells or proteins remain in a hydrated and an osmotically balanced state at physiological pH in the resultant reaction product. Preferably, pH and osmolality are maintained between 6.9 to 7.6 and between 250 to 400 mOsm/kg, respectively. Once cured, polymer sites containing the encapsulated biologics are easily washed, and manipulated.
  • Optical examination of these thiol-crosslinked hydrogels reveals optical clarity with no background fluorescence attributable to the gel formulation and generally similar optical properties to hydrogel formulations described in the '683 patent. However, in the '683 processes, it was often very important to carefully control the rate of CO2 evolution to avoid some opacity. The present process which uses dithiol crosslinkers in combination with bidentate modifiers inherently minimizes CO2 evolution, as mentioned before with respect to FIG. 2, and it can produce an optically transparent hydrogel with essentially no difficulty.
  • To show that these thiol-crosslinked polyurethane hydrogels are suitable for encapsulating a wide range of biologics, encapsulation of living eukaryotic cells was first examined. One criteria for the success of encapsulation of living cells is an assessment of continued cell viability, and typically, trypan blue exclusion is a technique frequently favored by biologists to easily determine cell viability. However, because the hydrogel absorbs a significant amount of the trypan blue dye, determination of cell color and intensity of the intracellular dye using this method was unreliable. As an alternative, AlamarBlue™ (Trek Diagnostic Systems, Inc.) was used. AlamarBlue is a dye that becomes fluorescent upon reduction by metabolic processes. Goat lymphocytes, having been determined by trypan blue exclusion to have both viable and dead cells present within the cell mixture, were chosen, and the prepolymer and cell suspension/dithiol crosslinker/bidentate crosslinker were mixed. The lymphocytes became encapsulated within the thiol-crosslinked hydrogel when droplets of the mixture were deposited as spots (approximately 300-1,000 microns in diameter, with a height equal to or greater than 20 microns) upon glass slides and then cured in a high humidity chamber at room temperature; mixing and curing took just less than 20 minutes. Preferably, the relative humidity (RH) is at least about 90% and more preferably is about 95% or higher. With the spots firmly attached to the glass slide, the slide was incubated for three hours at 37° with RPM 1640 media followed by incubation for 1.5 hours with AlamarBlue™ dye that had been mixed one part to twenty with RPM1640 media. After leaving the slide for 30 minutes in the dark, visualization of the spots using an epifluorescence microscope revealed brightly stained individual cells against a moderately fluorescent hydrogel background. Visible light revealed cells within the gel which were not brightly stained, which were presumably the aforementioned dead cells already present within the cell suspension. Hydrogel-only spots treated in an analogous fashion had no visible fluorescence. Therefore, AlamarBlue™ is felt to be a useful tool for accessing cell viability within these polyurethane-PEG hydrogels, and the hydrogel itself and the polymerization process were shown to be biocompatible by the maintenance of viable cells.
  • Encapsulation of proteins was also examined, formulating the gel essentially as just described. Protein encapsulation was demonstrated by the sequestration/encapsulation of anti-transferrin antibody within the gel matrix during polymerization. Verification of the antibody's functionality was demonstrated by the specific binding of fluorescent dye-labeled transferrin to those sites containing the anti-transferrin antibody and not at other sites containing different antibodies or no antibodies.
  • As additional embodiments, encapsulated living cells and/or proteins within such a thiol-crosslinked hydrogel might be deposited in pre-identified areas as spots or regions upon support surfaces, such as glass slides, or within microwells or microchambers, such as would be present in standard 96 well, 384 well or 1536 well microtiter plates. Distinct advantages are present with both approaches. In repeatedly depositing a number of discrete spots upon a single surface, each spot might contain a different entity, allowing a single incubation followed by the supply of wash solutions to contact all spots simultaneously. The use of individual microchambers would allow robotic handling of the plates and permit the use of low volumes of individual test solutions at each well. Combinations of these two approaches may also be used whereby individual chambers, arranged in a standard 96 well array or similar format, are each supplied with one or more hydrogel spots containing different entities.
  • Devices employing such arrays might be employed as combinatorial chemical or drug screening devices, antibody arrays, peptide arrays, cell arrays, enzymatic activity arrays, or DNA or other polynucleotide arrays that will be selective for binding to related proteins or other biomolecules. In addition, encapsulated cells or biomolecules coated onto the walls of microcapillary tubes will function as flow-through devices having single or multiple channels, which might be employed as screening devices or as biosensors on systems, such as in liquid chromatography or in “lab-on-a-chip” devices. Signal readout from such devices might be via binding of fluorescent proteins or of antigens, to be measured by subsequent antibody-based detection methods (possibly employing additional arrays), or via reaction with endogenous biopathways which will result in the formation of a detectable species, e.g. enzymatic conversion of a substrate to a fluorescent dye molecule, or change in the electrical properties, e.g. conductivity, of the cell and/or surrounding matrix resulting from exposure to the specific agent. In particular, the addition of either a redox agent to the gel or the addition of an electrically conductive polymer may enable signal detection by electrical, non-photonic, means.
  • The working examples which follow show methods of preparation that include the best mode presently known for providing formulations and encapsulation methods embodying particular features of the invention; however, they should be understood not to constitute limitations upon the scope of the invention which is of course defined by the claims that are set forth hereinafter.
  • EXAMPLE 1
  • Solution A was prepared by mixing 0.075 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1.5 mL of 50 mM aqueous phosphate buffer, at pH 7.0 with 80 mM sodium chloride. Solution B was prepared by dissolving 30 mg of PEG-(thiol)2 (mw=3,400) and 2 mg of free base cysteine (Sigma Chemical Co.) (mw=121) in 1 mL of 50 mM phosphate buffer, at pH 7.0 with 60 mM sodium chloride. Solution C was prepared by mixing 100 μL of Solution B with 10 μL of goat lymphocytes in Dulbecco's phosphate-buffered saline. Finally, 200 μL of Solution A was mixed with 50 μL of Solution C, and the resulting solution was microspotted onto amine-treated glass (Silanated Slides, Cell Associates, Inc.) with the use of 5 microliter glass microcapillary tubes. The hydrogel spots were polymerized carefully in a humidity box, at about 95% RH, to avoid dehydration. This formulation polymerized within 5-10 minutes. After polymerization, the hydrogel spots were physically stable and strongly attached to the glass slide; they were immediately treated with Dulbecco's modified phosphate-buffered saline solution and incubated at either room temperature or at 37□ for about 3 hours in RPM1640 cell media. The viability of lymphocytes was examined by means of the AlamarBlue staining method described previously. They were incubated for 1.5 hours with RPM1640 media plus the dye and then examined with a light microscope; viable, encapsulated cells were observed.
  • EXAMPLE 2
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride. Solution B was prepared by the same procedure as in Example 1. Solution C was prepared by mixing 40 μL of Solution B with 70 μL of goat lymphocytes in Dulbecco's phosphate-buffered saline. Finally, 100 μL of Solution A was mixed with 100 μL of Solution C, and the resulting solution was microspotted using the same procedure as in Example 1. The formulation polymerized in 5 minutes, and the hydrogel spots were treated with Dulbecco's modified phosphate-buffered saline solution and incubated at 37° C. for 1 day to 3 days in RPM1640 cell media. The viability of lymphocytes was examined with a light microscope using AlamarBlue which demonstrated viable encapsulated cells.
  • EXAMPLE 3
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride. Solution B was prepared by the same procedure as in Example 1. Solution C was prepared by mixing 40 μL of Solution B with 70 μL of E. coli in Dulbecco's phosphate-buffered saline. Finally, 100 μL of Solution A was mixed with 100 μL of Solution C, and the resulting solution was placed into a disposable culture tube. The formulation polymerized in 5 minutes, and the hydrogel was treated with Dulbecco's modified phosphate-buffered saline solution and incubated at 37° C. for 1 day in RPM1640 cell media. Viability and growth of E. coli were confirmed by observing turbidity in the hydrogel after 1 day.
  • EXAMPLE 4
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer, at pH 7.0 with 80 mM sodium chloride. Solution B was prepared by dissolving 30 mg of PEG-(thiol)2 (mw=3,400) and 2 mg of free base cysteine in 1 mL of 50 mM phosphate buffer, at pH 7.0 with 60 mM sodium chloride. 25 μL of Solution A, 10 μL of Solution B, 5 μL of 50% trehalose in DI water and 10 μL of anti-transferrin antibody (goat anti-human transferrin, 5 mg/ml, protein-G purified )(Calbiochem) were mixed, and the resulting solution was microspotted onto an amine-treated glass slide so as to form spots 300 μm to 1,000 μm in diameter and at least 20 μm in height. Other similar spots were created without the addition of the anti-transferrin antibody. The hydrogel spots were carefully polymerized in a humidity box at room temperature and 95% RH, and after polymerization, the hydrogel spots were found to be physically stable and well attached to the glass slide. The slide was treated with a PBS buffer containing 1% Bovine Serum Albumin (Sigma Chemical Co.) and 0.1% Triton X-100 (Boehringer Mannheim) for 1 hour at room temperature. Anti-transferrin in the hydrogel was interacted with Cy3-labeled transferrin (1 μg/ml in 1% bovine serum albumin, 0.1% triton X-100 in phosphate buffered saline) for 1 hour and then visualized; it demonstrated that there was specific binding of fluorescent dye-labeled transferrin at sites containing the anti-transferrin antibody and not at other sites containing different antibodies or no antibodies.
  • EXAMPLE 5
  • Solution A was prepared by mixing 0.1 g of Hypol PreMa G-50 (Hampshire Chemical Corp.) and 1 mL of 50 mM phosphate buffer at pH 7.0. Solution B without salts was prepared by the same procedure as in Example 1. Solution C was prepared by mixing 40 μL of Solution B with 70 μL of 234 μm L-alpha-cysteine-N-[8-(1,2,3,4-tetrahydro-acridin-9-ylamino)-octyl]-amide (mw=428.26), an acetylcholine esterase inhibitor, in 50 mM phosphate buffer at pH 7.0. Finally, 100 μL of Solution A was mixed with 100 μL of Solution C, and the resulting solution was microspotted using the same procedure as in Example 1. The formulation polymerized in 5 minutes, and the hydrogel microspots were treated with cy-3 labeled acetylcholine esterase. This testing confirmed presence and functionality of acetylcholine esterase inhibitor in the hydrogel reaction product.
  • Although the invention has been described with regard to certain preferred embodiments, it should be understood that changes and modifications as would be obvious to those having ordinary skill in this art may be made without deviating from the scope of the invention which is set forth in the claims appended hereto. The inclusion of additional polymers or modifications to the above-described polymer might permit either cell proliferation or increased viability of select cell types within the matrix. For example, peptide linkages within such a polymer may be specifically crafted to dissolve upon exposure to extracellular matrix proteases generated by the encapsulated cell, thereby dissolving the polymeric matrix as needed to permit cell expansion and growth. Alternatively, other polymers or agents, such as collagen, might be added to such a polymeric blend to aid cell viability by use of specific adhesion factors and/or binding methods between encapsulated cells and surrounding support. In contrast to spotting the hydrogel compositions onto a solid substrate or surface, hydrogel microbeads may be formed which encapsulate biologics. As one example, after mixing the prepolymer with the crosslinker and biologics, the polymer/cell (or protein) mix is added to a non-miscible liquid, such as an oil, while curing is occurring to cause microbeads of various dimensions to be formed. Separation from the oil or other suspending liquid yields a slurry of beads suitable for use in bioreactors, assay devices, artificial organs, biosensors or the like. Moreover, multiple layers of encapsulated cells, proteins or other bioactive molecules might be used to construct complex materials having unique overall properties. Alternatively, dyes or other agents might be added to the encapsulating polymer to facilitate subsequent identification of the encapsulated cell type if heterogeneous mixtures of cells are to be employed. Such a cell identification mechanism, combined with a chromaphore-based or fluorescent-based response from specific cells in response to added agents, e.g. expression of green fluorescent protein in response to specific cell signaling pathway activation by a ligand or drug, permits the screening of large populations of heterogeneous cells in a rapid and facile fashion.
  • The disclosures of all U.S. patents mentioned hereinbefore are incorporated herein by reference. Particular features of the invention are set forth in the claims which follow.

Claims (20)

1. A method of encapsulating biologics within a hydrogel comprising:
(a) providing an isocyanate-functional hydrogel prepolymer;
(b) providing an amount of biologics;
(c) providing crosslinkers having at least two thiol groups;
(d) mixing said prepolymer, said crosslinkers, and said biologics in aqueous solution to initiate polymerization under physiological pH and osmolality and create a polymerizing hydrogel; and
(e) dispensing said polymerizing hydrogel into a desired physical form.
2. The method of claim 1 wherein said amount of biologics is mixed in said aqueous solution with said thiol crosslinker prior to mixing with said prepolymer.
3. The method of claim 1 wherein said thiol crosslinkers comprise a crosslinking agent having a backbone of polyethylene glycol, polypropylene glycol, or a copolymer thereof and a molecular weight between about 2,000 and about 6,000.
4. The method of claim 1 wherein said thiol crosslinkers include a crosslinking agent having two thiol groups and a bidentate crosslinking agent having one thiol group and another different isocyanate-reactive group.
5. The method of claim 4 wherein the bidentate crosslinking agent comprises cysteine or homocysteine.
6. The method of claim 1 wherein the polymerizing hydrogel is dispensed onto a solid substrate so as to result in attachment thereto of the hydrogel-encapsulated biologics.
7. The method of claim 1 wherein the polymerizing hydrogel is repeatedly dispensed onto multiple, pre-identified areas of a substrate to form discrete spots arranged as an array.
8. The method of claim 7 wherein different of said spots at known locations contain at least two different biologics.
9. The method of claim 1 wherein said biologic has a molecular weight not greater than about 500 and contains a thiol-reactive group at a location that will not interfere with its spatial configuration for linking to an isocyanate group of the polymerizing hydrogel.
10. The method of claim 1 wherein said mixture is maintained at a pH of about 6.6 to about 7.1.
11. The method of claim 1 wherein said mixture is maintained at an osmolality of about 250 to about 400 mOsm/kg.
12. A method of preparing a hydrogel composition having a biologic encapsulated therein, which method comprises the steps of:
(a) providing an isocyanate-functional hydrogel prepolymer, a biologic, and crosslinkers having at least two thiol groups that will react with said isocyanate-functional hydrogel prepolymer,
(b) mixing said isocyanate-functional hydrogel prepolymer, said biologic and said crosslinkers in an aqueous solution at neutral pH, to provide a solution containing about 3 to about 5 weight % prepolymer, and
(c) maintaining said mixture of step (b) at neutral pH, so as to cause said biologic to be encapsulated within a polymerized hydrogel wherein said biologic remains bioactive.
13. The method according to claim 12 wherein said mixture is maintained at a pH of between about 6.5 and about 7.5.
14. The method according to claim 12 wherein said aqueous solution also contains a water-soluble bidentate crosslinking agent having two different isocyanate-reactive nucleophilic groups.
15. A hydrogel composition having a bioactive biologic encapsulated therein, which composition comprises:
the reaction product of an isocyanate-functional prepolymer, a biologic, and a crosslinker having at least two thiol groups,
said isocyanate-functional prepolymer having been crosslinked in the presence of said biologic by reaction with said crosslinker in an aqueous solution at physiological conditions and, as a result of said crosslinking, said biologic is encapsulated within a stable polymerized hydrogel in a manner so that said biologic remains bioactive.
16. The hydrogel composition of claim 15 wherein said prepolymer is a polyurethane prepolymer having a backbone of polyoxyalkylene diols or polyols having a molecular weight between about 5000 and about 30,000 which were reacted with difunctional or polyfunctional isocyanates.
17. The hydrogel composition of claim 15 wherein said crosslinker comprises a thiol-functional crosslinking agent having two thiol groups and a bidentate crosslinking agent having one thiol group and another different isocyanate-reactive group.
18. The hydrogel composition of claim 17 wherein the bidentate crosslinking agent comprises cysteine.
19. The hydrogel composition of claim 15 wherein said biologic is selected from the group consisting of proteins, nucleic acids, living cells, and organic molecules having a molecular weight between about 100 and about 2000.
20. The hydrogel composition of claim 15 wherein said thiol-functional crosslinker comprises a backbone of polyethylene glycol, polypropylene glycol, or a copolymer thereof and has a molecular weight between about 2,000 and about 6,000.
US11/670,061 2001-04-03 2007-02-01 Methods and gel compositions for encapsulating living cells and organic molecules Abandoned US20070141164A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/670,061 US20070141164A1 (en) 2001-04-03 2007-02-01 Methods and gel compositions for encapsulating living cells and organic molecules

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US28126801P 2001-04-03 2001-04-03
US10/398,725 US7172866B2 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules
PCT/US2002/010411 WO2002081662A1 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules
US11/670,061 US20070141164A1 (en) 2001-04-03 2007-02-01 Methods and gel compositions for encapsulating living cells and organic molecules

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/398,725 Division US7172866B2 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules
PCT/US2002/010411 Division WO2002081662A1 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules

Publications (1)

Publication Number Publication Date
US20070141164A1 true US20070141164A1 (en) 2007-06-21

Family

ID=23076600

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/398,725 Expired - Lifetime US7172866B2 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules
US11/670,061 Abandoned US20070141164A1 (en) 2001-04-03 2007-02-01 Methods and gel compositions for encapsulating living cells and organic molecules

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/398,725 Expired - Lifetime US7172866B2 (en) 2001-04-03 2002-04-02 Methods and gel compositions for encapsulating living cells and organic molecules

Country Status (8)

Country Link
US (2) US7172866B2 (en)
EP (1) EP1373472B1 (en)
JP (1) JP4198468B2 (en)
AT (1) ATE365792T1 (en)
CA (1) CA2443060A1 (en)
DE (1) DE60220893T2 (en)
IL (2) IL158128A0 (en)
WO (1) WO2002081662A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023076620A1 (en) * 2021-10-29 2023-05-04 Sigilon Therapeutics, Inc. Compositions for cell-based therapies and related methods

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002094900A1 (en) * 2001-05-21 2002-11-28 Dow Global Technologies Inc. Transparent polyurethane-hydrogel composition, method of making transparent polyurethane-hydrogel composition, and air-freshener application
WO2003050276A1 (en) * 2001-12-05 2003-06-19 Dow Global Technologies Inc. Method for immobilizing a biologic in a polyurethane-hydrogel composition, a composition prepared from the method, and biomedical applications
GB2407821B (en) 2002-06-05 2007-03-21 Es Cell Int Pte Ltd Stem Cells
EP3023140B1 (en) * 2003-04-10 2019-10-09 President and Fellows of Harvard College Formation and control of fluidic species
US9200245B2 (en) 2003-06-26 2015-12-01 Seng Enterprises Ltd. Multiwell plate
WO2004113492A1 (en) * 2003-06-26 2004-12-29 Molecular Cytomics Ltd. Improved materials for constructing cell-chips, cell-chip covers, cell-chip coats, processed cell-chips and uses thereof
BRPI0414004A (en) 2003-08-27 2006-10-24 Harvard College electronic control of fluidic species
US20050112650A1 (en) * 2003-10-20 2005-05-26 Ciphergen Biosystems, Inc. Reactive polyurethane-based polymers
US20060040377A1 (en) * 2004-08-17 2006-02-23 Biocept, Inc. Protein microarrays
CA2583308C (en) * 2004-10-08 2020-01-07 Georgia Tech Research Corporation Microencapsulation of cells in hydrogels using electrostatic potentials
US7439062B2 (en) 2004-12-23 2008-10-21 Biocept, Inc. Beads for capturing target cells from bodily fluid
WO2006078470A2 (en) 2005-01-18 2006-07-27 Biocept, Inc. Cell separation using microchannel having patterned posts
US8158410B2 (en) 2005-01-18 2012-04-17 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20090136982A1 (en) 2005-01-18 2009-05-28 Biocept, Inc. Cell separation using microchannel having patterned posts
WO2007030661A1 (en) * 2005-09-09 2007-03-15 Avery Dennison Corporation Hydrogel including modified cyclodextrin crosslinked with polyurethane prepolymer
EP1928918B1 (en) * 2005-09-27 2011-11-16 UNIVERSITE JOSEPH FOURIER - Grenoble 1 Hydrogel functionalized with a polymerizable moiety and their uses as biosensors or bioreactors
US8263405B2 (en) 2005-11-24 2012-09-11 Mitsuru Akashi Controllably degradable hydrogel for culturing cells to produce three-dimensionally organized cells
US7695956B2 (en) 2006-01-12 2010-04-13 Biocept, Inc. Device for cell separation and analysis and method of using
KR100849185B1 (en) * 2006-01-19 2008-07-30 서울산업대학교 산학협력단 Chitosan or Hyaluronic acid-Polyethylene oxide- and Chitosan-Hyaluronic acid-Polyethylene oxide-Based hydrogel and Manufacturing Method Therefor
US20070243549A1 (en) * 2006-04-12 2007-10-18 Biocept, Inc. Enrichment of circulating fetal dna
KR100721431B1 (en) 2006-04-19 2007-05-25 학교법인 포항공과대학교 Polymer capsule and process for the preparation thereof
EP2101975A2 (en) 2006-11-03 2009-09-23 Trustees of Tufts College Biopolymer sensor and method of manufacturing the same
CA2704304C (en) * 2006-11-03 2015-01-13 Trustees Of Trufts College Biopolymer optical waveguide and method of manufacturing the same
EP2086749B1 (en) 2006-11-03 2013-05-08 Trustees Of Tufts College Nanopatterned biopolymer optical device and method of manufacturing the same
WO2008118211A2 (en) 2006-11-03 2008-10-02 Trustees Of Tufts College Biopolymer photonic crystals and method of manufacturing the same
US20080176768A1 (en) * 2007-01-23 2008-07-24 Honeywell Honeywell International Hydrogel microarray with embedded metal nanoparticles
WO2009061823A1 (en) 2007-11-05 2009-05-14 Trustees Of Tufts College Fabrication of silk fibroin photonic structures by nanocontact imprinting
US9145540B1 (en) 2007-11-15 2015-09-29 Seng Enterprises Ltd. Device for the study of living cells
KR20100118973A (en) * 2007-12-13 2010-11-08 다니스코 유에스 인크. Compositions and methods for producing isoprene
EP2237887A2 (en) 2007-12-26 2010-10-13 Seng Enterprises Ltd. Device for the study of living cells
JP2011525254A (en) * 2008-06-18 2011-09-15 トラスティーズ オブ タフツ カレッジ Edible holographic silk products
JP5188314B2 (en) * 2008-08-04 2013-04-24 キヤノン株式会社 Biopolymer testing apparatus and method
US20100240054A1 (en) * 2008-09-22 2010-09-23 Biocept, Inc. Identification and isolation of fetal cells and nucleic acid
CA2789009C (en) 2009-02-12 2017-03-21 Trustees Of Tufts College Nanoimprinting of silk fibroin structures for biomedical and biophotonic applications
JP2012533780A (en) 2009-07-20 2012-12-27 タフツ ユニバーシティー/トラスティーズ オブ タフツ カレッジ Implantable absorptive reflector made of protein only
WO2011026101A2 (en) 2009-08-31 2011-03-03 Trustees Of Tufts College Silk transistor devices
WO2011057131A1 (en) 2009-11-09 2011-05-12 Spotlight Technology Partners Llc Polysaccharide based hydrogels
JP2013509963A (en) 2009-11-09 2013-03-21 スポットライト テクノロジー パートナーズ エルエルシー Fragmented hydrogel
US9174216B2 (en) 2013-03-13 2015-11-03 DeNovo Science, Inc. System for capturing and analyzing cells
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
EP2739587B1 (en) 2011-08-01 2020-05-27 Denovo Sciences Cell capture system
US9404864B2 (en) 2013-03-13 2016-08-02 Denovo Sciences, Inc. System for imaging captured cells
US9606102B2 (en) 2013-01-26 2017-03-28 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US9707562B2 (en) 2013-03-13 2017-07-18 Denovo Sciences, Inc. System for capturing and analyzing cells
EP2796873A1 (en) * 2013-04-25 2014-10-29 QGel SA Method for a cell-based drug screening assay and the use thereof
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US9856535B2 (en) 2013-05-31 2018-01-02 Denovo Sciences, Inc. System for isolating cells
WO2015147147A1 (en) * 2014-03-26 2015-10-01 国立大学法人北海道大学 Method for high-speed fixing of biological specimen by using hydrogel and device therefor
US9694338B2 (en) * 2014-12-18 2017-07-04 Wisconsin Alumni Research Foundation Covalently-immobilized hydrogel arrays in multi-well plates
EP3403098B1 (en) 2016-01-12 2021-09-22 BioAtla, Inc. Diagnostics using conditionally active antibodies
AU2018323449B2 (en) 2017-08-29 2020-09-03 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10633693B1 (en) 2019-04-16 2020-04-28 Celsee Diagnostics, Inc. System and method for leakage control in a particle capture system
EP3966307A4 (en) 2019-05-07 2023-10-04 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
AU2020290981B2 (en) 2019-06-14 2024-03-07 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
BR112023020933A2 (en) 2021-04-16 2023-12-12 Capra Biosciences Inc GENETICALLY MODIFIED ORGANISM, RETINOL DEHYDROGENASE GENE (RDH12), EXPRESSION VECTOR, HOST CELL, RETINOL DEHYDROGENASE 12 (RDH12), BIOFILM, BIOREACTOR AND BIOFILM REACTOR, AND METHOD FOR PRODUCING AN ISOPRENOID IN A BIOFILM BIOREACTOR
WO2024036256A1 (en) 2022-08-10 2024-02-15 Capra Biosciences, Inc. Circulating biofilm bioreactor

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4098645A (en) * 1976-02-24 1978-07-04 W. R. Grace & Co. Immobilization of proteins with polyurethane polymers
US4436813A (en) * 1982-03-16 1984-03-13 Purification Engineering, Inc. Immobilized microbial cell composition for making L-aspartic acid
US4794090A (en) * 1986-09-26 1988-12-27 W. R. Grace & Co.-Conn. Immobilization support for biologicals
US5175229A (en) * 1986-11-18 1992-12-29 W. R. Grace & Co.-Conn. Biocompatible polyurea-urethane hydrated polymers
US5403750A (en) * 1991-03-06 1995-04-04 W. R. Grace & Co.-Conn. Biocompatible, low protein adsorption affinity matrix
US5573934A (en) * 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5849368A (en) * 1995-02-01 1998-12-15 Schneider (Usa) Inc Process for hydrophilicization of hydrophobic polymers
US5962280A (en) * 1997-02-14 1999-10-05 Nippon Shokubai Co., Ltd. Microorganisms immobilized on a solid support with a quaternary polyallylamine polymer
US6174683B1 (en) * 1999-04-26 2001-01-16 Biocept, Inc. Method of making biochips and the biochips resulting therefrom
US6433134B1 (en) * 1998-07-09 2002-08-13 Biocept, Inc. Peptide nucleic acid precursors and methods of preparing same
US20030134294A1 (en) * 2001-12-05 2003-07-17 Sandford Andrew F. Method for immobilizing a biologic in a polyurethane-hydrogel composition, a composition prepared from the method, and biomedical applications
US6642046B1 (en) * 1999-12-09 2003-11-04 Motorola, Inc. Method and apparatus for performing biological reactions on a substrate surface
US6818018B1 (en) * 1998-08-14 2004-11-16 Incept Llc In situ polymerizable hydrogels

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE286746T1 (en) * 1999-01-29 2005-01-15 Beiersdorf Ag PROTEIN CONTAINING HYDROGELS
DE60137523D1 (en) * 2000-10-26 2009-03-12 Biocept Inc THREE-DIMENSIONAL FORMAT-PROOFING BIOCHIP

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4098645A (en) * 1976-02-24 1978-07-04 W. R. Grace & Co. Immobilization of proteins with polyurethane polymers
US4436813A (en) * 1982-03-16 1984-03-13 Purification Engineering, Inc. Immobilized microbial cell composition for making L-aspartic acid
US4794090A (en) * 1986-09-26 1988-12-27 W. R. Grace & Co.-Conn. Immobilization support for biologicals
US5175229A (en) * 1986-11-18 1992-12-29 W. R. Grace & Co.-Conn. Biocompatible polyurea-urethane hydrated polymers
US5403750A (en) * 1991-03-06 1995-04-04 W. R. Grace & Co.-Conn. Biocompatible, low protein adsorption affinity matrix
US5573934A (en) * 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5849368A (en) * 1995-02-01 1998-12-15 Schneider (Usa) Inc Process for hydrophilicization of hydrophobic polymers
US5962280A (en) * 1997-02-14 1999-10-05 Nippon Shokubai Co., Ltd. Microorganisms immobilized on a solid support with a quaternary polyallylamine polymer
US6433134B1 (en) * 1998-07-09 2002-08-13 Biocept, Inc. Peptide nucleic acid precursors and methods of preparing same
US6818018B1 (en) * 1998-08-14 2004-11-16 Incept Llc In situ polymerizable hydrogels
US6174683B1 (en) * 1999-04-26 2001-01-16 Biocept, Inc. Method of making biochips and the biochips resulting therefrom
US6642046B1 (en) * 1999-12-09 2003-11-04 Motorola, Inc. Method and apparatus for performing biological reactions on a substrate surface
US20030134294A1 (en) * 2001-12-05 2003-07-17 Sandford Andrew F. Method for immobilizing a biologic in a polyurethane-hydrogel composition, a composition prepared from the method, and biomedical applications

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023076620A1 (en) * 2021-10-29 2023-05-04 Sigilon Therapeutics, Inc. Compositions for cell-based therapies and related methods

Also Published As

Publication number Publication date
DE60220893D1 (en) 2007-08-09
JP4198468B2 (en) 2008-12-17
EP1373472B1 (en) 2007-06-27
US7172866B2 (en) 2007-02-06
EP1373472A4 (en) 2005-11-09
IL158128A0 (en) 2004-03-28
EP1373472A1 (en) 2004-01-02
IL158128A (en) 2009-02-11
WO2002081662A1 (en) 2002-10-17
JP2004533500A (en) 2004-11-04
DE60220893T2 (en) 2008-03-06
ATE365792T1 (en) 2007-07-15
CA2443060A1 (en) 2002-10-17
US20040029241A1 (en) 2004-02-12

Similar Documents

Publication Publication Date Title
EP1373472B1 (en) Methods and gel compositions for encapsulating living cells and organic molecules
JP2004533500A5 (en)
Griffith et al. Microdistribution of substratum-bound ligands affects cell function: hepatocyte spreading on PEO-tethered galactose
US10266652B2 (en) Cross-linked poly-E-lysine non-particulate support
US7625951B2 (en) Stimuli-responsive hydrogel microdomes integrated with genetically engineered proteins for high-throughput screening of pharmaceuticals
US20140038826A1 (en) Covalently cross linked hydrogels and methods of making and using same
KR20020026275A (en) Biochip and method for making it
WO2006023323A1 (en) Microarrays utilizing hydrogels
Lu et al. Cell encapsulation with alginate and α‐phenoxycinnamylidene‐acetylated poly (allylamine)
EP1328810B1 (en) Three dimensional format biochip
CA2435894C (en) End group activated polymers with oligonucleotide ligands
US20060084159A1 (en) Method for manufacturing of three dimensional composite surfaces for microarrays
Frey et al. Surface-immobilized biomolecules
CN113621551B (en) Dynamic cell microenvironment simulation platform and preparation method thereof
AU2002246918B9 (en) Three dimensional biochip
AU2002246918A1 (en) Three dimensional biochip

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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