CA2371011C - Polyionic coatings in analytic and sensor devices - Google Patents

Polyionic coatings in analytic and sensor devices Download PDF

Info

Publication number
CA2371011C
CA2371011C CA002371011A CA2371011A CA2371011C CA 2371011 C CA2371011 C CA 2371011C CA 002371011 A CA002371011 A CA 002371011A CA 2371011 A CA2371011 A CA 2371011A CA 2371011 C CA2371011 C CA 2371011C
Authority
CA
Canada
Prior art keywords
peg
pll
functionalized
copolymer
polyionic
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.)
Expired - Lifetime
Application number
CA002371011A
Other languages
French (fr)
Other versions
CA2371011A1 (en
Inventor
Marcus Textor
Donald L. Elbert
Stephanie Finken
Laurence Ruiz-Taylor
Nicholas D. Spencer
Rolf Hofer
Jeffrey A. Hubbell
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.)
Eidgenoessische Technische Hochschule Zurich ETHZ
Universitaet Zuerich
Original Assignee
Eidgenoessische Technische Hochschule Zurich ETHZ
Universitaet Zuerich
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 Eidgenoessische Technische Hochschule Zurich ETHZ, Universitaet Zuerich filed Critical Eidgenoessische Technische Hochschule Zurich ETHZ
Publication of CA2371011A1 publication Critical patent/CA2371011A1/en
Application granted granted Critical
Publication of CA2371011C publication Critical patent/CA2371011C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B5/00Measuring for diagnostic purposes; Identification of persons
    • A61B5/145Measuring characteristics of blood in vivo, e.g. gas concentration, pH value; Measuring characteristics of body fluids or tissues, e.g. interstitial fluid, cerebral tissue
    • A61B5/14546Measuring characteristics of blood in vivo, e.g. gas concentration, pH value; Measuring characteristics of body fluids or tissues, e.g. interstitial fluid, cerebral tissue for measuring analytes not otherwise provided for, e.g. ions, cytochromes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B5/00Measuring for diagnostic purposes; Identification of persons
    • A61B5/145Measuring characteristics of blood in vivo, e.g. gas concentration, pH value; Measuring characteristics of body fluids or tissues, e.g. interstitial fluid, cerebral tissue
    • A61B5/1455Measuring characteristics of blood in vivo, e.g. gas concentration, pH value; Measuring characteristics of body fluids or tissues, e.g. interstitial fluid, cerebral tissue using optical sensors, e.g. spectral photometrical oximeters
    • A61B5/1459Measuring characteristics of blood in vivo, e.g. gas concentration, pH value; Measuring characteristics of body fluids or tissues, e.g. interstitial fluid, cerebral tissue using optical sensors, e.g. spectral photometrical oximeters invasive, e.g. introduced into the body by a catheter
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/28Materials for coating prostheses
    • A61L27/34Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • 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
    • 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
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • G01N33/54373Apparatus specially adapted for solid-phase testing involving physiochemical end-point determination, e.g. wave-guides, FETS, gratings
    • 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
    • G01N33/54393Improving reaction conditions or stability, e.g. by coating or irradiation of surface, by reduction of non-specific binding, by promotion of specific binding
    • 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
    • G01N33/551Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being inorganic
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/814Enzyme separation or purification
    • Y10S435/815Enzyme separation or purification by sorption
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/961Chemistry: molecular biology and microbiology including a step of forming, releasing, or exposing the antigen or forming the hapten-immunogenic carrier complex or the antigen per se
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/969Multiple layering of reactants
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/819Multifunctional antigen or antibody
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/823Immunogenic carrier or carrier per se

Abstract

Multifunctional, polyionic copolymers are synthesized on/or applied to substrate surfaces for analytical and sensing purposes, the coatings being particularly useful for suppression of non-specific interaction, adsorption or attachment. Groups can be coupled to, integrated into or absorbed to the multifunctional polymer that are able to recognize, interact with and bind specifically to target analyte molecules. These materials can also be used to modulate biological interactions upon substrate surfaces for use as selective implant surfaces that resist cell attachment and may optionally promote the attachment of specific cell types or induce a particular cellular behaviour. The multifunctional polymer coatings typically include brush copolymers based on a polycationic or polyanionic backbone with side chains that control interaction with the environment, such as poly(ethylene glycol) or poly(ethylene oxide)-based side chains that decrease cellular adhesion, and analyte-specific side chains.

Description

POLYIONIC COATINGS IN ANALYTIC AND SENSOR DEVICES

Background of the Invention This application is in the general area of polymeric coating materials which can be applied to surfaces of substrates used in analytical and sensing devices to promote specific recognition of the target analyte while minimizing non-specific adsorption of other molecules in the sampling solution.

There is a need to improve the selectivity and sensitivity of bioaffinity and diagnostic sensors, especially for use in screening assays and libraries for DNA/RNA and proteins. A common approach to diagnostic sensor design involves the measurement of the specific binding of a particular component of a physiological sample. Typically, physiological samples of interest (e.g. blood samples) are complex mixtures of many components that all interact to varying degrees with surfaces of diagnostic sensors. However, the aim of a diagnostic sensor is to probe only the specific interaction of one component ~vi-hile minimizing all other unrelated interactions. In the case of sensors in contact with blood, proteins, glycoproteins and/or saccharides, as well as cells, often adsorb non-specifically onto the sensor surface. This impairs both selectivity and sensitivity, two highly important performance criteria in bioaffinity sensors.

A variety of materials and surfaces is used in analytical and sensor devices, such as glass, silicon wafers. metals or metallized surfaces and metal oxides as bulk or coating materials. The choice of the materials is closely related to the particular sensing technique used. If surface plasmon resonance is used as an analytical or sensing method, the chips will consist of a substrate coated with a metal such as gold. In the case of analytical or sensor chips to be used in combination with specific optical detection techniques, such as fluorescence spectroscopy, optical waveguide techniques or a combination of the two, optically transparent substrates and/or coatings are often needed. Metal oxides or metal oxide coatings are particularly suitable in such cases, in view of their stabilitv, inertness and optical transparency. Polymeric materials are traditionally used for applications in lateral flow assays or multiwell plate assays. Glass or silicon based materials are often used for capillary electrophoresis applications. Both polvmeric and glass type materials are used in fiberoptics.

Independent of the analytical or sensing technique used, there is a basic need for surface modification of the chips, as these materials and surfaces do not have the necessary properties in terms of controlled adsorption phenomena in a given detection or sensing application. There is a i o need for simple, cost-effective treatment materials and methods that effectively reduce non-specific interactions at or adsorption onto metal oxide-based sensor surfaces while introducing a specific binding interaction with the target analyte. Such specific recognition, binding and detection are often achieved through key-lock type of chemical or biological interactions such as antibody-antigen interactions.

The application of multifunctional polymers in implants can be employed to control biological interactions that determine a bioresponse to an implant. Here the same principles of recognition, specificity and suppression of nonspecific adsorption are relevant. An example is the selection of attaching cells in a biological response, where suppression of nonspecific adsorption of proteins from the body fluids can suppress the nonspecific attachment of cells and the inclusion of biospecific recognition ligands, such as adhesion peptides, can enhance the attachment of a specific cell population or subpopulation.

The use of functional polymers and copolymers is an approach often chosen in the biomaterial area to modify surface properties or cell-to-cell interactions. For example, U.S. Patent Nos. 5,573,934 and 5,626,863 to Hubbell et al. disclose hydrogel materials containing a water-soluble region such as polyethylene glycol and a biodegradable region, including various biodegradable polymers such as polylactide and polyglycolide, terminated with photopolymerizable groups such as acrylates. These materials can be applied to a tissue surface and polymerized, for example, to form tissue coatings. These materials are adhered to tissue surfaces by polymerizing the photopolynlerizable groups on the materials after they have been applied to the tissue surface. U.S. Patent Nos. 5.462,990 and 5,627,233 to Hubbell et al. disclose multifunctional polymeric materials for use in inhibiting adhesion and immune recognition between cells and tissues. The materials include a tissue-binding component (polycation) and a tissue non-binding component (polyanion). In particular. Hubbell discloses various PEG/PLL
copolymers, with molecular weights greater than 300, with structures that include AB copolymers, ABA copolymers, and brush-type copolymers.

These polymers are being commercially developed for use as tissue sealants and to prevent surgical adhesions. NN'O 98/47948 `Multifunctional Polymeric Tissue Coatings' by Hubbell et al. describes another class of polymer, grafted polyionic copolymers that are able to attach to biological and non-biological samples in order to control cell-surface and cell-cell and tissue-surface interactions in biomedical applications. These materials have not been used to coat devices or devices for implantation, however, especially devices having metal oxide or other surfaces which may not adhere well to receptors or other ligands used for sensing or analysis.

It is therefore an object of the present invention to provide materials which can be used to control and modify cell-surface and cell-cell interactions, especially on the surface of devices and implants.

It is a further object of the present invention to provide methods for application of these materials to a substrate which is rapid, inexpensive, and flexible.

It is another object of the present invention to provide a stable polymeric material that can be applied simply, quickly and cost-effectively to charged surfaces of analytical devices and sensor chips for applications where it is essential to establish a specific binding interaction while preventing the unwanted non-specific interactions that typically occur at surfaces in contact with physiological and other types of samples.

analytical or diagnostic devices, culture surfaces, sensor chips, and implants.
Summary of the Invention Multifunctional, polyionic copolymers with molecular architectures and properties optimized for specific applications are synthesized on/or applied to substrate surfaces for analytical and sensing purposes. The coatings are particularly useful for suppression of non-specific interaction, adsorption or attachment of molecular or ionic components present in an analyte solution. Chemical, biochemical or biological groups can be coupled to, integrated into or absorbed to the multifunctional polymer that are able to recognize, interact with and bind specifically to target molecules in the i o material containing the analyte to be detected. These multifunctional polymer coatings are compatible with a variety of different established methods to detect, sense and quantify the target molecule in an analyte. These materials can also be used to modulate biological interactions upon substrate surfaces for use as selective implant surfaces that resist cell attachment and may optionally promote the attachment of specific cell types or induce a particular cellular behavior.

The multifunctional polymer coatings typically include brush copolymers based on a polycationic or polyanionic (jointly referred to herein as `polyionic') backbone with side chains that control interaction with the environment, such as poly(ethylene glycol) or poly(ethylene oxide)-based side chains that decrease cellular adhesion (referred to herein as "non-interactive" side chains or polymers), and analyte-specific side chains.
Examples of polyionic backbones are poly(amino acids) such as poly(lysine) and poly(arginine) with positive charges at physiological pH and poly(glutamic acid) and poly(aspartic acid) with negative charges at physiological pH. The poly(ethylene glycol) (PEG) chains are highly water soluble and highly flexible. PEG chains have an extremely high motility in water and are essentially non-ionic in structure. They are well known for their weak interaction with both molecules and ions and, if attached to the surface in suitable form (molecular weight, density, orientation), they decrease adhesiveness or adsorption to the surface, such as protein resistance in contact with blood or serum. The choice of positively charged (cationic) or negatively charged (anionic) backbones of such PEO-grafted backbones is related to the fact that surfaces often possess a positive or negative charge when exposed to an aqueous environment. In particular, metal oxides or metal oxide coatings exposed to an aqueous analyte spontaneously acquire a negative charge at pH above the isoelectric point (IEP) and positive charges at pH below the isolectric point of the particular oxide chosen. At pH of 7 (neutral solution) for example, niobium oxide. tantalum oxide or titanium oxide will be negatively charged, while aluminium oxide at pH 7 is positively charged. In other cases, such as a noble metal surface, the surface may not be (sufficiently) charged to allow for complete polymer adsorption through electrostatic interaction. In such cases the surface may be treated to introduce positive or negative charges. For example. carboxylate groups may be introduced through self-assembly of carboxy-terminated long-chain alkanethiols on gold or silver to induce a positive charge at a pH above 4.

Alternatively, amino groups that are positively charged at pH below 9 may be introduced on gold or silver surfaces, e.g. through self-assembly of amino-terminated alkanethiols. The PEG-grafted polyionic copolymer can be further modified by introducing functional groups at or near the terminal (free end) position of the PEG chains. These groups allow further functionalization and incorporation of adhesive sites or recognition entities that serve as the capture moiety in an analytical or sensing assay.
Non-modified and modified copolymers can be used singly, consecutively or as a mixture. They can be used to pattern the surfaces into non-adhesive and specifically adhesive areas by applications of known techniques such as microfluidic or contact printing techniques.

Although described primarily with reference to substrates or surfaces of analytical or sensing devices, where the material the copolymer is applied to is a metal or metal oxide, polymer substrates can also be used, especially when patterning is not involved in the bioanalytical concept. For example, many surfaces employed in ELISA systems and in cell culture systems are polymers. Polystyrene is a very convenient such polymer, because of its optical clarity, its ease of processability, and its low expense. Polystyrene can be readily functionalized to introduce surface charge, either negative or positive, to create the appropriate charged substrate. For example, exposure of polystyrene to air or oxygen plasmas is well known to create stable modified surfaces that include anionic aroups, and exposure to ammonia plasnlas is well known to create stable modified surfaces that include cationic groups at typical operating pH. Thus, it is readily possible to employ such polymers, and also many other polymers either with anionic or cationic groups that are intrinsic to their polymer structure or that are introduced by a surface modification procedure such as a plasma process, in both bioanalytical systems based on noncellular and cellular approaches.
Such noncellular approaches include ELISA systems, where the multifunctional polymer is adsorbed to the polymer substrate and an antibody is then chemically coupled to the multifunctional polymer.
Likewise, in cell-mediated systems, the polymer may be adsorbed and then coupled to a cell-interacting biomolecule, such as an adhesion peptide, or alternatively the cell-interacting biomolecule may be coupled prior to adsorption. In both ELISA systems and in cell-culture systems, simple and inexpensive polymer substrates may be advantageous.

Both metal and polymer substrates may also be useful in the context of medical implants. For example, a number of important implants are constructed from metals, such as titanium or stainless steel, including orthopedic implants and cardiovascular implants. Likewise, a number of important implants are constructed from polymers, such as silicone rubber and polyesters, both of which can be treated by plasma processes or other processes to introduce surface charge. The polymeric materials can also be advantageous when used to coat these materials.

Brief Description of the Figures Figures 1 A-1 D are the chemical structures of polymers described herein. Figure lA is the chemical structure of the polycationic poly(L-lysine)-g-poly(ethylene glycol) (PEG-g-PLL) for surface modification of negatively charged surfaces; Figure 1B is a chemical structure of a PEG-g-PLL polymer, functionalized with biotin at the terminus of part of the PEG

side chains; Figure 1C is the chemical structure of the polyanionic poly(L-glutamic acid)-g-poly(ethylene glycol) (PEG-g-PLG) for surface niodification of positively charged surfaces; and Figure 1 D is a chemical structure of a (PEG-g-PLG) polymer functionalized with biotin at the terminus of part of the PEG side chains.
Figure 2A is a schematic 6f adsorbed multifunctional polymers to render surfaces protein- and cell-resistant. Graft copolymers (a) and block copolymers (b) are formed from cationic components (heavy line) and poly(ethylene glycol) (light line). Specific peptides are attached to the tips of the poly(ethylene glycol) chains (dots).
Figure 2B is a schematic drawing of a bioaffinity assay based on a chip surface coated with a copolymer-monolayer. The recognition molecule is immobilized by interaction with the terminal function of the polymer-grafted PEG. The surface can be used for the specific binding of target molecules (left), while the PEG-layer prevents non-specific adsorption of biological and non-biological components of the analyte.
Figure 3 is a graph showing adsorption curves of PLL(375)-g[5.6]-PEG(5) on three metal oxide surfaces as measured by the grating coupler technique. [10 mM HEPES Z1 (pH 7.4). 1 mg/mL polymer, I mL/hr, T=26 C].
Figure 4 is a graph showing the dependence of the adsorbed amount of PLL(375)-g[5.6]-PEG(5) on the isoelectric point of the metal oxide surfaces as measured by the optical grating coupler technique[10 mM
HEPES Zl (pH 7.4), 1 mg/mL polymer, 1 mL/hr, T=26 C.]
Figure 5 is a graph of serum adsortpion to either an unmodified or a PLL(375)-g[5.6]-PEG(5)-modified Sio.6Ti0,402 surface as measured by the optical grating coupler technique. [10 mM HEPES Z 1(pH 7.4), 1 mg/mL
polymer, T=26 C.] (The baseline was achieved under 10 mM HEPES Z1, and the spikes are due to temporary flow rate related pressure changes.) Figure 6 is a graph of the adsorbed areal mass of serum and HSA
onto PLL(375)-g[5.6]-PEG(5)-modified and unmodified Sio.6Tio,402, Ti02 and Nb,O; surfaces as measured by the optical grating coupler technique. [10 mM HEPES Zl (pH 7.4), 1 mg/mL polymer, 1 mL/hr, T=26 C.]

Figure 7 is a graph of the long-term stability of the protein-adsorption suppression of a PLL(375)-g[5.6]-PEG(5)-modified Si0.6Ti0.40, surface as measured by the optical grating coupler technique. [Polymer adsorption carried out under 10 mM HEPES Z1, 1 mg/mL polymer, 1 mL/hr, T=26 C;
serum adsorption, 1 mL/hr, T=26 C.]

Figure 8 is a graph of the effect of polymer architecture on the adsorbed areal density of the copolymer and subsequent serum on Si0.6Ti0.40, surfaces as measured by the optical grating coupler technique.
[Polymer adsorption carried out under 10 mM HEPES Z1, 1 mg/mL
polymer. I mL/hr, T=26 C; serum adsorption, I mL/hr, T=26 C.]

Figure 9 is a graph of the activity of horseradish peroxidase: Titanium oxide coated with different PLL-g-PEG derivatives. The streptavidin-conjugated enzyme is immobilized via biotin, which acts as the functional group of the PLL-PEG-biotin copolymer. The non-specific binding of the enzyme onto the uncoated metal oxide as well as onto the non-functionalized PLL-g-PEG is investigated in parallel.

Figure 10 is the time-resolved selective sensor response of the biotin-derivatized PLL-g-PEG-biotin (adsorbed onto a waveguide chip) to streptavidin exposure.

Figure 11 is a graph of the amount of PLL-g-PEG-biotin adsorbed onto a SixTi 1-x 02 waveguide chip surface and the amount of streptavidin subsequently adsorbed onto the PLL-g-PEG-biotin covered chip surface for three different biotin concentrations in the PLL-g-PEG-biotin molecule (see Table 5).
Figure 12 is a graph of the amount of streptavidin detected by the optical waveguide technique as a function of biotin concentration in the polymer on a chip surface formed of SixTi 1-x 02 with adsorbed PLL-g-PEG-biotin.
Detailed Description of the Invention Polycationic and polyanionic copolymers that adsorb to (oppositely) charged surfaces of substrates and devices ('chips') for analytical and sensing applications are disclosed. These serve three functions: (1) charged sites in the backbone attach the polymer to substrate surface via interactions between oppositely charged groups on the substrate and the backbones; (2) grafted side chains that form a dense structure such as a brush make the surface non-interactive, i.e., prevent the adsorption of molecules or ions out of an analyte onto the surface (referred to herein as "non-interactiveness", "non-adhesiveness", or "resistance to non-specific adsorption") and (3) functional groups allow the incorporation of recognition units for specific detection and sensing of low concentrations of analytes (referred to herein as "fiulctional recognition sites"). Although described primarily with reference to chips, these materials can also be used to coat polymeric materials used in immunoassay and combinatorial libran, screening techniques, usually polymers such as polystyrene or polycarbonate, or implants such as bone prosthetics, screws, rivits, and vascular grafts, which are typically formed of metals and/or polymeric materials.

Treatment of the surface or substrate materials with the copolymers makes the surface completely or nearly completely resistant to (unwanted) non-specific adsorption. Suitable functional, reactive or interactive groups can also be introduced into the copolymers. As used herein, these functional or reactive or interactive copolymers are referred to as `modified copolymers'. These reactive groups can be used for the further functionalization of the polymer, i.e. the adsorption or attachment of specific molecules such as antibodies, antigens, enzymes, oligonucleotides, single-stranded DNA or RNA moieties. The latter molecules immobilized at the surface in a sea of non-interactiveness are able to sense specifically target molecules using established analytical or sensing techniques. The multifunctional copolymers that include the specific recognition unit or functional molecules are referred to herein as `functionalized polymers'.
Procedures for treatment of the substrates and surfaces with the non-modified, modified and/or functionalized polyionic copolymers are also disclosed.

Preferred analytical or sensing detection procedures and applications of the chips modified and functionalized are described in detail below.

1. Compositions of Non-Interactive Polymer-grafted Polyionic S Copolymers.

Block copolymers are defined as copolymers in which a polymeric block is linked to one or more other polymeric blocks. This is distinguished from random copolymers, in which two or more monomeric units are linked in random order to form a copolymer. Brush copolymers (as in a = i o bottlebrush) are copolymers, which have a backbone of one composition and bristles of another. These copolymers are also known as comb copolymers.
The terms brush and comb are used interchangeably. Dendritic polymers, also known as dendrimers or starburst polymers, are polymers which include a core molecule which is sequentially reacted with monomers with three or I i more reactive groups, such that at each sequential coupling step, the number of reactive groups at the ends of the polymer increases, usually exponentially. A dendron is a subunit of a dendrimer, the cone shaped structure resulting from sequential reactions starting with a core containing only reactive group. As used herein. molecular weight refers to weight-2o average molecular weight, unless otherwise specified.

The preferred non-interactive polymer is poly(ethylene glycol), or "PEG". For ease of description, the 2eneral description of the graft copolymers refers to PEG rather than "non-interactive polymer".

The co-polymers can be brush copolymers (as in a bottlebrush, with a 25 backbone of one composition and bristles of another) with a charged polymeric backbone. such as a poly(aniino acid). The first example refers to poly-L-lysine (PLL) and bristles of polyethylene glycol (PEG) (see Figure 1 A). The molecular weight of the PLL is between 1,000 and 1,000,000, preferably greater than 100,000, more preferably, between 300,000 and 800,000. The 30 molecular weiglit of the PEG is between 500 and 2,000,000, more preferably between 1,000 and 100,000. Various surfaces binding polyionic polymers WO 00/65352 PCT/[1S00/1 1708 can be substituted for PLL, and various non-interactive polymers can be substituted for PEG.
A. Noir-Iirteraetive Polti-mers The term 'non-interactive' is used here in the context of the application of such molecules for coating analytical and sensor devices and means that the non-interactive polymer in the surface-adsorbed copolymer reduces the amount of (non-specific) adsorption of molecules such as inorganic ions, peptides, proteins, saccharides and other constituents contained in typical analytes of biological or non-biological origin.

Alternatives to the wording non-interactive are non-adhesive, adsorption-resistive or adsorption-repulsive in the context of non-specific adsorption.
PEG is a preferred material as the non-interactive polymer. The choice of the grafting ratio (number of'PEG chains to number of monomers in the polymeric polyionic backbone) is important as it determines, when i 5 adsorbed onto the device surface, the degree of the desired non-adhesiveness.
For PEG with a MW of 5000, the optimal graft ratio is between I PEG chain for every 3 to 10, preferably 4 to 7, lysine subunits for analytical or sensing applications, and may be adjusted based on desired properties. The optimum grafting ratio however depends on the MW of the PEG as well as on the specific applications. For example, if the MW of the PEG chains to be grafted onto the PLL backbone is 2000. the ratio of PEG units to PLL units should be between 2 and 8, preferentially between 3 and 5. The properties of sensor surfaces coated with PLL-PEG of different architecture are discussed in Example 1.
Suitable non-interactive polymers include mixed polvalkylene oxides having a solubility of at least one gram/liter in aqueous solutions such as some poloxamer nonionic surfactants, neutral water-soluble polysaccharides, polyvinyl alcohol, poly-N-vinyl pyrrolidone, non-cationic .
poly(meth)acrylates, many neutral polysaccharides, including dextran. ficollTM

3o and derivatized celluloses, polyvinyl alcohol, non-cationic polyacrylates, such as poly(meth)acrylic acid, and esters amide and hydroxyalkyl amides thereof, and combinations thereof.

B. Polyioiiic Backbone Polymers Suitable polycationic blocks include natural and unnatural polyamino acids having net positive charge at or close to neutral pH, positively charged polysaccharides, and positively charged synthetic polymers. Representative polycationic blocks include monomeric units selected from the group consisting of lysine, histidine, arginine and ornithine. Representative positively charged polysaccharides include chitosan, partially deacetylated chitin, and amine-containing derivatives of neutral polysaccharides.
Representative positively charged synthetic polymers include polyethyleneimine, polyamino(meth)acrylate, polyaminostyrene, polyaminoethylene, poly(aminoeth),l)ethylene, polyaminoethylstyrene, and N-alkyl derivatives thereof. Representative polycationic materials include natural and unnatural polyamino acids having net positive charge at neutral pH, positively charged polysaccharides. and positively charged synthetic polymers. Examples of suitable polycationic materials include polyamines having amine groups on either the polymer backbone or the polymer sidechains, such as poly-L-lysine and other positively charged polyamino acids of natural or synthetic amino acids or mixtures of amino acids, including poly(D-lysine), poly(ornithine), poly(arginine), and poly(histidine), 2o and nonpeptide polyamines such as poly(aminostyrene), poly(aminoacrylate), poly (N-methyl aminoacrylate), poly (N-ethylaminoacrylate), poly(N,N-dimethyl aminoacrylate), poly(N,N-diethylaminoacrylate), poly(aminomethacrylate), poly(N-methyl amino-methacrylate), poly(N-ethyl aminomethacrylate), poly(N,N-dimethyl aminomethacrylate), poly(N,N-diethyl aminomethacrylate), poly(ethyleneimine), polymers of quaternary amines, such as poly(N,N,N-trimethylaminoacrylate chloride), poly(methylacrylamidopropyltrimethyl ammonium chloride), and natural or synthetic polysaccharides such as chitosan. Polylysine is a preferred material.

In general, the polymers must include at least five charges, and the molecular weight of the polyionic material must be sufficient to yield the desii-ed dearee of binding to the surface of the analytical or sensing device, having a molecular weight of at least 1000 g/mole.
For example. PEG reacted .vith polyethylene imine with a molecular weight greater tlian 10.000 will have approximately the saine physical ; properties as the PEG/PLL copolymers described herein. Polyhydroxyethyl methacrylate can be reacted with a suitable stoichiometric ratio of a reagent such as tresyl or tosyl chloride (an activating agent), which converts some of the hydroxy groups to leaving groups. These leaving groups can be reacted xvith polycationic polymers, for example, polyaminoethyl methacrylate with iu a molecular weight greater than 10,000. to yield a high-molecular-weight polymer. A suitable stoichiometric ratio is one mole activating agent per mole of polyhydroxyethyl methacrylate. and one mole activated polyhydroxyethyl methacrylate per every 3 to 9, preferably 5 to 7 moles of reactive groups on polyaminoethyl methacrylate. Suitable cationic polymers 15 are those that, when combined with a suitable non-interactive polymer, have roughly the same physical properties as the PEG/PLL copolymers described lierein.
Suitable polyanionic blocks include natural and synthetic polyamino acids having net negative charge at neutral pH. A representative polyanionic 2o block is poly(glutamic acid), which contains carboxylic acid side chains with a negative charge at pH 7 (see Figure 1 C). Glycolic acid is just one example.
It may be replaced by other natural or unnatural monomers that can be polymerized and contain a side functional group with negative charge at or near neutral pH, for example, any polymer having carboxylic acid groups attached as pendant 25 groups. Suitable materials include alginate. carrageenan, furcellaran, pectin, xanthan, hyaluronic acid, heparin, heparan sulfate, chondroitin sulfate, dermatan sulfate, dextran sulfate, poly(meth)acrylic acid, oxidized cellulose, carboxymethyl cellulose and crosmarmelose, synthetic polymers and copolymers containing pendant carboxyl groups, such as those containing 3o maleic acid or fumaric acid in the backbone. Polyaminoacids of predominantly negative charge are particularly suitable. Examples of these materials include polyaspartic acid, polyglutamic acid, and copolymers thereof with other natural and unnatural amino acids. Polyphenolic materials such as tannins and lignins can also be used. Preferred materials include alginate, pectin, carboxymethyl cellulose, heparin and hyaluronic acid.

In general, the molecular weight of the polyanionic material must be sufficiently high to yield strong adhesion to the positively charged analytical or sensor device surface. The lengths of the polycationic and polyanionic materials which would result in good blockage of adhesive interactions may be determined by routine experimentation. It should be understood that "good" is a word that must be defined by the requirements of the particular situation at hand, e.g., how long bindinQ is required and how complete a non-interactivness is required by the particular sensing application.

Apart from carboxylate end groups, sulfate, sulfonate, phosphate or groups that apart from being negatively charged have cation-chelating properties such as ethylendiamintetraacetic acid (EDTA) or nitrilotrisacetic acid (NTA) can be used as well. The term `cation-chelating' refers to the ability of such a functional group to strongly coordinate to cations contained in the surface of the sensing device, preferentially metal cations such as Ti(IV), Zr(IV), Hf(IV), V(V), Ta(V), Nb(V), Hf(V), Cr(III), Cr(VI), Mo(VI) or W(VI).

The amine groups in copolymers are the primary amines of lysine residues, but other groups can be used. For example, the polymer can be prepared using arginine or histidine. resulting in guanidino or imidazoyl cationic groups, respectively. Likewise. more than one PEG group can be provided, for example, by using as a starting material a small molecule with at least two carboxyl groups and at least two amino groups, for example, the dipeptide Glu-Lys.

For all embodiments, the molecular weight and number of PEG
blocks per lysine block is determined such that the resulting copolymer has the properties of both the PLL and the PEG. If the proportion of PEG is too high, the adhesion of the polymer to the analytical or sensing substrate (chip) is reduced. If the proportion of PLL is too high, the ability of the PEG to reduce the non-specific adsorption of ions and molecules out of the analyte is insufficient. The polymers must have sufficient PEG character to minimize molecular interaction with the analytical or sensor chip. Polymers with too few PEGs per PLL are less suitable for minimizing these interactions. The polymers must also have sufficient PLL character to adequately bind to a chip surface. Polymers with insufficient PLL character fail to bind adequately. The polycationic polymer can be any polycation that provides a sufficient amount and density of cationic charges to be effective at adhering to the analytical or sensing substrate.

C. PEG/po[yionic poly ier dendrimers.

- 0 The PEG/poly(amino acid) dendrimers are copolymers where one or more linear PEG polymeric blocks are covalently linked to the focal point of a cationic dendrimer, for example. dendrimerically polymerized polylysine, such that the dendi-imer fans out from the PEG. Preferably, the PEG is linked to the central point of the dendrimer, which is grown from the PEG as described in detail below. The particular utility of the dendritic construction is the ability to precisely control the mass of the resulting copolymer, the geometrical relationship between the polymeric blocks, and the degree of substitution. For instance, in the examples shown, there is exactly one PEG
for a defined number of positive charges. In contrast, grafting preformed PEG molecules onto a polycationic backbone normally results in a random positioning of the PEG groups on the backbone.

The dendrimer preferably contains between 16 and 128 reactive amine groups, which correlates to a dendrimer of between generation 4 and generation 7. The molecular weight of the PEG is between 500 and 2,000,000, preferably between 5,000 and 100,000.
The dendrimeric PLL allows the formation of a compact structure, with a high charge density. These PEG-lysine dendrons are effective in preventing cell spreading when adsorbed on a simple anionic surface if the polymer contains about 8 or more positive charges (generation-3 dendron).

Other non-lysine-based dendrimers can also be prepared and are intended to be within the scope of the PEG/PLL dendrimers described herein.
For example, the dendrimers can include polycationic groups other than amines. for example, quaternarv ammonium salts. Further, synthetic, non-amino acid based cations can be included. Cationic amino acids such as ui-nithine can also be incorporated into the dendrimers.

D. Analyte-specific Li;ands The first requirement for an analytical or sensing device. which allows specific detection or recognition, is the resistance of the device surface towards non-specific adsorption. This requirement can be fulfilled by the copolymers described above, as well as in Example 1. The second requirement is the introduction of functional groups (called ligands) that Ie allow specific interaction with select components of the analyte. Such ligands can be introduced into the PEG-grafted polyionic polymer.
Preferentially such ligands are introduced at the free end of the PEG chain.
The tvpe of ligand is chosen accordina to the needs of the analytical or sensing task. Examples are:

15 Ligands that induce physico-chemical adsorption of a molecule as i-ecognition unit for the subsequent analytical or sensing detection. Examples of the recognition unit are monoclonal or polyclonal antibodies, that will specifically bind complex molecules to be analyzed during the sensing step (called target molecules) such as peptides, proteins, oligonucleotides, DNA

20 or RNA (fragments). The interaction of the recognition unit and the sensing device surface will be through electrostatic and/or van der Waals' forces.
Examples for the ligands are hydrophobic moieties such as methyl or trifluoromethyl groups, positively charged groups such as amines (carrying a positive charge below pH of 9), a quaternary amine (positively charged 25 across the whole pH range), or negatively charged ligands such as carboxylate (negatively charged above pH 4).

Ligands that are able to bind covalently to functional groups of the recognition unit. Examples are ester, maleimide, succinimidyl, vinylsulfone, conjugated C=C double bonds, epoxy, aldehyde, ketone, silane or siloxane 30 functionalities. These ligands can react with functional groups of the recognition unit, in particular with accessible amine, hydroxy or thiol groups.

Ligands that are able to bind to receptors on the surfaces of cells. In the approach of the immediately preceding paragraph, a recognition unit may be bound to the ligand directly, by reaction. An example is the reaction of a cysteine-containing peptide to a vinvlsulfone ligand. Specific reaction i between the tliiol on the cysteine residue and the vinylsulfone ligand then results in immobilization of the peptide to the substrate, or to the polymer that is then adsorbed to the substrate. This coupling may be done with the polymer still in fi=ee solution, and thus the ligand definitionally becomes the peptide. or it may be done after the polymer with the cysteine-reactive ligand io is adsorbed upon the substrate. Both approaches have advantages. In the first case, of the coupling of the peptide ligand to the polymer before adsorption to the substrate, the material may be more readily characterized.
In the latter case, of coupling of the peptide ligand after adsorption of the polymer upon the substrate, the separation of the non-coupled peptide is very 15 easy, being reduced to the process of rinsing. The case of the peptide ligand binding to receptors on the surface of a cell can be particularly interesting, e.g. in analysis of cellular behavior or in the therapeutic manipulation of cell behavior in a culture system or upon an implant.
Ligands that are able to bind specifically to a bioactive recognition 20 niuiet). Examples of such recognition units include antibodies in general, antigens. proteins. enzymes, oligonucleotides, c-DNA fragments and other groups or molecules provided they are able to interact specifically with the analyte in the subsequent analytical or sensing assay. These surface-attached recognition units must possesses further specific interaction sites, which can 25 be used for specific recognition and binding of a particular analyte molecule.
An example is the use of a polycationic PLL-g-PEG, where a fraction of the PEG chains are functionalized at the terminal position of the PEG chain with bi,,tin (see Figure IB). The functionalized PLL-g-PEG-biotin is adsorbed onto an analytical or sensing device surface, followed by the adsorption of streptavidin onto the 30 biotin sites. Such a sensing surface is able to recognize and bind specifically biotinylated target molecules during the analytical or sensing assay. Alternatively, the adsorbed.
Diotinylated PEG-grafted polyionic copolymer can directly react with analytes that are streptavidin-conjugated. Details are given in Example 2.

Ligands that are able to interact with an intermediate reactive bifunctional molecule (called crosslinker). Firstly, one end of the homo- or ~ (preferentiallv) hetero-functional cross linker is attached to the ligand of the copolymer. In a second step the other (free) end of the crosslinker is reacted with the recognition unit.

Ligands that are able to form stable complexes with a cation. In a second step the cation will form a complex either with the recognition unit directly through a suitable functionality or alternatively with a bifunctional crosslinker (in the latter case followed by procedure 4). Examples for the ligand include carboxylate, amide, phosphate. phosphonate, nitrilo triacetic acid and other known groups that are able to chelate cations. Examples for the cations include Mg(II), Ti(IV), Co(III), Co(VI), Cu(II), Zn(II), Zr(IV), Hf(IV), V(V), Nb(V), Ta(V), Cr(III), Cr(VI), Mo(VI) and other cations known to form stable complexes with chelating ligands.

Examples of suitable recognition units include antibodies in general, proteins, enzymes, lectins, oligonucleotides, c-DNA fragments and other groups or molecules provided they are able to interact specifically with the 2o analyte in the subsequent analytical or sensing assay.

Many interesting ligands in the bioanalysis of cellular responses are peptides.
In such cases, the peptides may be coupled to the multifunctional polymers, e.g. to the termini of the polymer chains that are used to resist nonspecific adsorption, either before or after the polymer is adsorbed to the substrate.

The peptide may be bound to the multifunctional polymer through a number of means, including reaction to a cysteine residue incorporated within the peptide. Cysteine residues are rarely involved in cell adhesion directly. As such, few cell adhesion peptides comprise a cysteine residue, and thus a cysteine residue that is incorporated for the purpose of coupling of the peptide will be the unique cysteine residue for coupling. While other approaches are possible, the preferred method is coupling of the peptide to the multifunctional polymer through a cysteine residue on the polymer.

Othei- bioactive features can also be incorporated. e.g. adhesion proteins, growth factor proteins, cytokine proteins. chemokine proteins, and the like.
Funutionalized surfaces can be used in bioanalytical systems involving cells, in which sonle affecter of cell function is the measured feature. A test fluid mav contain an analyte, to which the response of cells is sought. The cellular response may be used in as a measure of the presence or the activity of the analyte. Alternatively, the cellular response per se may be the knowledge that is sought, e.g. the migration response of a particular cell type to a growth factor, when the cells are migrating upon a particular adhesive substrate. The collection of such scientific information is of significant value in the screening of the activity of drug candidates, particularly when higher order cellular responses such as adhesion. migration. and cell-cell interactions are tar<,reted.

Functionalized surfaces can be used in therapeutic systems involving cells, in which cells are cultured for later therapeutic use. In current therapeutic systems, cultured cells are sometimes used. Examples are in the culture of chondrocytes for transplantation in articular cartilage defects in the knee or in the culture of endothelial cells for transplantation in vascular grafts. In such cases, modulation and manipulation of the phenotype of the cells is of prime interest. For example. it may sometimes be of interest to stimulate cells into one phenotype (such as a proliferative phenotype), and later to stimulate them into a second phenotype (such as a differentiated phenotype). Alternatively, it may be desirable to maintain them in one particular phenotype, e.g. to maintain chondrocytes in a chondrocyte phenotype, rather than have them dedifferentiate into cells that more closely resemble fibroblasts. Surfaces and substrates may be useful in this. It is known that extracellular matrix interactions play an important role in regulating cell phenotype, and such interactions may be readily modified through the use of the multifunctional polymers of this invention.

Functionalzed surfaces can be used in therapeutic systems involving cells, in which the cells are cultured and used in contact with the surface. As an example of this situation, bioreactors are used in some extracorporal therapeutic systems, such as cultured hepatocytes used to detoxify blood in acute liepatic failure patients. In such cases, one wants to maintain the hepatocytes in the reactor in a functional, differentiated state. The adhesive interactions between the cells and their substrate are thought to play an important role in these interactions. and thus the technology of this invention provides a means by which to control these responses.

FLulctionalized surfaces can be used in therapeutic systems involving cells, in which the functionalized surfaces is a component of an implant. The interactions between cells in an implant environment and the surface of an implant may play a controlling role in determining the biocompatibility of an implant. For example, on the surface of a stent implanted within the coronary artery, the presence of blood platelets is not desirable and may lead to in-stent restenosis. As such, it would be desirable to prevent the attachment of blood platelets to the stent surface. The attachment of all cells is not necessarily bad, however. For example, endothelial cells are thought to play a very positive role in preventing restenosis. As such, it would be desirable to treat the stent substrate with a functional coating that would prevent the nonspecific adsorption of proteins and also presents adhesion Iigands that are capable of binding to endothelial cells but not blood platelets.
The peptide REDV, based on an alternatively spliced domain in fibronectin, provides one potential such target, in that it has been reported that this sequence does not bind to receptors present on the blood platelet but does bind to receptors that are present on the endothelial cell (J. Biol. Chem 1992, 267 p.14019-26).
D. Surfaces or Substrates ("chips") The type of substrates and surfaces of the analytical or sensing device suitable for the envisioned use depends on both the type of application, the type of analytical or sensing technique used, as well as on the suitability for binding the polyionic copolymer.
All types of substrates or substrate surfaces that are used in the area of analytical or sensing tasks which can be combined with any technique for the detection of the target molecules or analytes.

Suitable substrates or surfaces which include metals, metal oxides and/or polymeric materials are also discussed below in conjunction with the section on detection of analyte binding. Other substrates or surfaces include tissue and cell culture substrates, and means for immunoassay, which are typically formed of a polymer such as polystyrene or polycarbonate. Other supports or substrates include medical devices or prosthetics which are fornled of inetals (such as stainless steel). nylon. degradable and non-degradable biocompatible polymers such as poly(lactic acid-co-glycolide).
Examples include bone implants and prosthetics, vascular grafts, pins, screws, and rivits. The most common substrate material for a stent is a metal.
In many such implants, used in dentistry and in orthopedic surgery, metal implants are used. In other cases, polymeric implants may be more usefttl. It is possible to adsorb the multifunctional polymer to either polymeric or inorganic substrates, depending only upon the surface charge of the substrate.

Substrate and surface functionalization for use in other analytical assay platforms is also an important application of the technology. One of the largest areas in bioanalytical assays is the use of enzyme linked immunosorbent assays (ELISA) or linked immunosorbent assays (LISA). In these applications, a binding or recognition element is bound typically to a multiwell plate and then blocked with a protein based molecule to occupy surface area not containing the recognition element. The derivatized surface is then exposed to a solution containing analyte. The surface is then exposed to a second recognition element that is tagged with a molecule that can be assayed via any of the conventional spectroscopic or other methods. The above description is for a typical "sandwich" type assay, and this basic format can be modified in a variety of standard ways. For example, an antigen can be directly coupled to a surface and used to assay the level of antibody produced from immunological response to the antigen and present in biological fluids or tissues. This is a common technique used for determination of human disease or illness or even for determination of biocompatability of a drug or implant in which the degree of antibody production is measured as a function of drug level or dose.

One of the key issues with the assays is the blocking step that prevents non-specific binding. This step is time consuming and variable, and can generate false positive. Conventional methods for blocking of sites not uccupied by the antigen or antibody include blocking with proteins, for example bovine serum albumin (BSA).
Functionalization of substrates and substrate surfaces for the analysis and control of cellular interactions in an important use, having application in culture-based assays, in therapeutics based on cell and tissue culture and bioreactors, and based on implants. Functionalized surfaces can be used in bioanalytical systems involving cells. in which the cellular response is the measured feature. In the field of cell biology. the response of a cell to a substrate is an important issue. and analysis of this response is an important hioanalvtical task. For example, the response of a cell to extracellular matrix components is an important issue in cell adhesion and migration and is important in issues such as cancer metastasis and wound healing. As such, bioanalytical systems, including as a key component of them bioanalytical surfaces and substrates, are useful in measurement of such responses.
Useful substrates are polymeric or inorganic. Modified polystyrene is a common cell culture substrate, modified so as to render the polystyrene anionic. Such as substrate alone has limited usefulness in bioanalysis of cellular behavior. It supports cell adhesion via proteins that spontaneously adsorb or are adsorbed from purified solutions. These proteins are subject to remodeling by cellular activities. As such, technology that would provide a well defined culture substrate would indeed be useful. In such usefulness, qualities such as the ability to resist nonspecific adsorption, to present biospecific adhesion ligands, and to remain stably adsorbed during extensive periods of culture.
11. Synthesis of Polyionic Copolymers grafted with Non-Interactive Polymers and Analyte Specific Ligands.

A. Synthesis of Graft Copolymers.

PEG may be bonded to the a-amines of lysine residues of poly(L-lysine) as follows. Poly(L-lysine) (PLL) can be reacted with a PEG with one end protected (i.e., a protected monomethoxy PEG), the terminal hydroxyl of whicli has been previously activated with carbonyldiimidazole (CDI). The PLL and the activated PEG can be mixed in an aqueous solution buffered at pH 9 and allowed to react for 48 hours at room temperature. The number of PEG chains grafted per PLL chain may be controlled by adjusting the ratio of moles of activated PEG added per mole of added PLL. The reaction may not proceed to completion. i.e., the mole ratio of PEG to PLL in the reaction mixture may not be identical to that in the PEG-g-PLL product, but higher ratios of PEG to PLL will produce higher amounts of PEG in the PEG-g-PLL
product.

The cationic domains tend to be highly reactive, and efforts must be made to control the extent of addition of PEG to PLL. Executing the reaction in the absence of water reduces deactivation of PEG and allows better stoichiometric control. For example, unprotected poly-L-lysine can be - 5 dissolved in water, then added to dimethylformamide (DMF) to make a solution that is 5% aqueous. The poly-L-lysine can then be reacted with CDI
mono-activated PEG in stoichiometric amounts, followed by evaporation of solvent under vacuum yielding a PEG/PLL copolymer. Alternatively, unprotected poly-L-lysine can be dissolved in water and precipitated by adding NaOH. The precipitated polymer can then be added to anhydrous DMF and then reacted with CDI mono-activated PEG in stoichiometric amounts, yielding an (A)x-b-(B)y copolymer. When the reaction is performed in the absence of water, side reactions involving the activated group can be reduced (i.e., deactivation is reduced), and at long reaction times the ratio of mole PLL to PEG in the polymer product more closely resembles than in the reactant mixture.

Solution polymerization of PLL may be carried out using monomers containing different epsilon protecting groups, which allows strict control over the degree of substitution of PEG onto PLL. N-carboxy anhydrides of various amino acids may be synthesized and polymerized into copolymers, as in the following example. N,N'-dicarbobenzoxy-L-lysine (Z,Z-lysine) can be reacted with phosphorus pentachloride to yield a,N-carbobenzoxy-a,N-carboxv-L-lysine anhydride. u.N-carbobenzoxy-a,N-tert-butyloxycarbonyl-L-IN,sine (Z.boc-lysine) can be reacted with sodium methoxide to yield the sodium salt of Z.boc-lysine. The sodiuni salt of Z.boc-lysine can be reacted xvith phosphorus pentachloride to yield a.N-tert-butyloxycarbonyl-a,N-~ carboxy-L-lysine anhvdride. Z.Z-1}'sine anhvdride can be added to Z,boc-lysine anliydride. and the two monomers can be polymerized by the addition ot' sodium rnethoxide as an initiator. A copolynier results, poly((X boc-lysine)-co-(a Z-]ysine). The boc tiroups can be removed by addition of the polymer to trifluoroacetic acid for fifteen minutes.

= 10 The salt fonn can be converted to the free base by reaction with a reactant such as pyridine. The free amines on the polymer can then be reacted with CDI PEG in DMF. The Z groups can then be deprotected by adding the polymer to HBr in acetic acid for fifteen minutes, yielding an (PEG)x-b-(PLL)y copolymer, where the ratio of PEG to PLL in the final 15 product can be controlled by the initial ratio of boc protected lysines.

It may be desirable to produce versions of the polymer.that are not of a brush structure. This may be facilitated by not deprotecting the epsilon amines of PLL, so that the only reactive groups are the amine and carboxyl termini. For example. reaction of CDI activated PEG with poly a,N-2o carhobenzoxv-L-lysine in DMF yields an (A)x-(B)y copolymer. Activation of the carboxyl terminus of the (A)x-(B)y copolymer with 2-Succinimido- 1, 1, 3, J-tetramethyluronium tetrafluoroborate TSU followed by reaction with mono-amino PFG
in DMF yields an (A)x-(B)y-(A)z copolymer.

B. Synthesis of Funetionalized Graft Copolymers.

25 The synthesis of PEG-grafted polyionic copolymers, for which all or part of the PEG side chains are functionalized with a particular group at the terminal or close to terminal position. follows the same procedures as outlined above. However, instead of using a one end protected PEG (e.g.
methoxy terminated PEG), a functionalized PEG is used. Either only 30 functionalized PEG is used or (preferentially) a mixture of functionalized and non-functionalized PEG. The functional group at the end of the PEG chain are chosen according to the needs and concept of the analytical assay. The i-atio of tunctionalized PEG to non-functionalized PEG depends again on the requirements of the analytical or sensor assay. It has to be optimized for a given type of application. An example of the optimization of this ratio is given in Example 2.

The details of a particular synthesis are given in Example 2 for the case of biotinylated PEG/PLL to be used in PEG-biotin-streptavidin-biotinvlated type of assay. An alternative but related approach is provided in Example 3.

Non-charged, hydrophilic side chains other than PEG can be used as long as they impart the property of non-interactiveness or adsorption resistance to the treated surface 111. Methods for Surface Modification with Graft Copolymer.
A. Methods for modifying Chip Surfaces or Substrates Six typical general protocols for the modification of a chip surface intended to be used for bioaffinity sensing using the optical waveguide technique with or without fluorescence detection are described.
ProiocolA:

1) Pretreatment of the chip surface comprising a glass slide on which a metal oxide layer as waveguiding layer has been deposited: a) ultrasonic cleaning in an organic solvent; b) plasma treatment of the surface in an oxygen atmosphere 2) Spontaneous adsorption of a monolayer of the cationic PLL-g-(PEG),(PEG-biotin),_., out of an aqueous solution. 1-x denotes the fraction of ftinctionalized PEG chains in comparison to the total number of PEG chains.
3) Rinse 4) Exposure of the chip surface with an aqueous solution of streptavidin, binding specifically to the surface accessible biotin ligands.

5) Rinse of the surface to remove loosely (non-specifically) bound streptavidin.

6) Specific adsorption of an analyte solution containing biotin-conjugated target molecules to the surface-immobilized streptavidin sites.

7) Rinsing of the surface to remove weakly bound (non-specifically acisoi-bed) inolecules.

8) Detection of the specifically bound target molecule using the optical waveguide technique. This is accomplished by e.g. varying the incoupling angle until the conditions for incoupling are met. The determination of the > effective index through measurement of the electric and magnetic mode (TE
and TN,) then allows determining quantitatively the mass of the (specifically) adsorbed biotin-conjugated target molecule.

Alternatively to 8), the biotin-conjugated recognition molecule (antibody) is adsorbed to the streptavidin-functionalized chip surface. The target molecule - o is specifically immobilized at the chip surface, followed by reaction with a tracer molecule (fluorescently labeled antibody to the target molecule). The evanescent field generated at the surface of the waveguide is then used to excite fluorescence of the tracer molecule. The fluorescence is detected u5ing a fluorescence detector situated on the side opposite to the liquid flow 15 cell, making use of the very high detection sensitivity of the fluorescence technique in combination with the advantage that practically only the labeled molecules attached to the surface are fluorescence-excited, with minimal contribution from `bulk' fluorescence.

Protocol B

20 1) Pretreatment of the chip surface comprising a glass slide on which a metal oxide layer has been deposited. as a waveguiding layer: a) ultrasonic cleaning in an organic solvent; b) plasma treatment of the surface in an oxygen atmosphere 2) Spontaneous adsorption of a monolayer of the cationic PLL-g-25 (PEG),(PEG-vinylsulfone),_x out of an aqueous solution. 1-x denotes the fraction of functionalized PEG chains in comparison to the total number of PEG chains 3) Attachment of single stranded c-DNA fragments containing a thiol group that adds to the vinylsulfone function via a nucleophilic additions.

30 4) Rinse of the surface to remove weakly bound (not covalently linked) c-DNA.

5) Specific adsorption of the complimentary DNA strand (target molecule) that has been previously labeled with a fluorescence tag to the c-DNA
recognition ligand.

6) Rinse to remove weakly bound (non-specifically attached) molecules.
~ Detection of the specifically bound DNA fragments upon determination of the intensity of the fluorescence excited in the evanescent field of the ~~-aveguide.
Prolocol C

1) Cleaning of a gold-coated chip using organic solvents.

2) Self-assembly of an c,o-amino-terminated alkanethiol out of alcoholic solution to form an ordered self-assembled monolayer (SAM) through interaction of the thiol group with the gold substrate. This SAM will acquire positive charges when in contact with an aqueous environment at pH below 9 (e.g. at neutral pH) as a consequence of the terminal amino groups being partly protonated.
3) Spontaneous adsorption of the polvanionic (negatively charged) polymer poly(glutamic acid)-g-(PEG),(PEG-succinimid),_, onto the positively charged Au-SAM.
4) Reaction of the succinimid groups accessible at the surface of the polymer-adsorbed chip with a primary amine group of an antigen.

5) Rinse to remove loosely bound (non-specifically adsorbed) antigen.
Specific reaction of the surface-bound antigen with the corresponding antibody in the analyte solution monitored in situ by the Surface Plasmon Resonance (SPR) technique.

Protocol D
1) Pretreatment of the chip surface comprising a glass slide on which a metal oxide layer has been deposited as a waveguiding layer: a) ultrasonic cleaning in an organic solvent; b) plasma treatment of the surface in an oxygen atmosphere.
2) Microfluid patterning of the waveguide chip surface with an aqueous solution of PLL-g-PEG using a polymer stamp with a micron-sized pattern at the stamp surface. This process produces a patterned (e.g. a type of line pattern) surface consisting of regions that have been PLL-g-PEG treated (along the microfluidic channel network) and regions that have not been treated (where the stamp protected the surface).

3) Treatment of the patterned waveguide chip surface in an aqueous solution of the cationic PLL-g-(PEG),(PEG-vinvlsulfone),_,. This step will fill the uncovered regions in step 2) with the functionalized PLL-g-(PEG),(PEG-~-inNlsulfone),., producing a pattern with non-flulctionalized and fiuictionalized PLL-g-PEG.
4) Rinse -0 5) Adsorption or attachment of different recognition molecules in different regions with adsorbed functionalized PLL-g-PEG using techniques that allow localized recognition molecules deposition such as ink-jet techniques, printing or fluidic patterning.

6) Rinse Application of a solution containing multiple fluorescently labeled analytes and two-dimensionally resolved, parallel detection of the specific binding of different analyte molecules at spatially separated, different recognition sites by 2-dimensionally resolved detection of the fluorescence excited by the evanescent field of the optical waveguide.

?0 Pi'otoc'nl E
1) Pretreatment of the chip surface comprising a glass slide on which a metal oxide layer has been deposited as a waveguiding layer: a) ultrasonic cleaning in an organic solvent; b) plasma treatment of the surface in an oxygen atmosphere 2) Spontaneous adsorption of a monolayer of PLL-g-(PEG)X(PEG-R'-OPOA_, out of an aqueous solution. 1-x denotes the fraction of functionalized PEG chains in comparison to the total number of PEG chains.
R' is a crosslinker used to attach the phosphate group at the terminal position of the PEG chain in the course of the synthesis of the modified PEG-g-PLL.
3) Rinse 4) Exposure of the chip surface with an aqueous solution of a soluble zirconium Zr(IV) salt such as zirconium (IV) nitrate. Zirconium (IV) cations \Nill bind to the surface accessible phosphate groups of the modified PEG-g-PLL polymer.

5) Rinse of the surface to remove loosely bound, not phosphate-coordinated Zirconium (IV) cations 6) Specific adsorption of phosphate or phosphonate-conjugated antibody molecules to the zirconium (IV) sites. The phosphate or phosphonate group of the antibody will coordinatively attach to the immobilized zirconium(IV) cations via formation of a complex.

7) Rinsing of the surface to remove NN-eakly bound (non-specifically adsorbed) molecules.

8) Exposure of the functionalized chip to a solution that contains an Lulknown inixture of different proteins. Detection of the protein molecules that interact specifically with the surface accessible, immobilized antibodies on the chip surface upon determination of the intensity of the fluorescence excited in the evanescent field of the waveguide.
Protocol F
1) Pretreatment of the chip surface comprising a suitable substrate such as silicon on which an electronic circuit suitable for the use as a nerve cell-based sensor has been produced.

2) Microfluidic patterning of the chip surface with an aqueous solution of PLL-g-PEG using a polymer stamp with a micron-sized pattern at the stamp surface that is compatible with the electric network of the sensor surface.
This process produces a surface consisting of regions that have been PLL-g-PEG treated (along the microfluidic channel network) and regions that have not been treated (where the stamp protected the surface).

Treatment of the patterned chip surface in an aqueous solution of the cationic PLL-g-(PEG)x(PEG-peptide),_,. This step will fill the uncovered regions in step 2) with the functionalized PLL-g-(PEG)X(PEG-peptide),_X, producing a pattern with non-functionalized and functionalized PLL-g-PEG.
The oligopeptide is e.g. YIGSR known to be a specific sequence in the protein laminin that induces the attachment and migration of nerve cells.

4) ExposLu=e of the patterned surface to a cell-culture solution containing nerve cells. Attachnient and spreading of the nerve cells in the protein- and ceil-adhesive areas.

Use of the living-cell-based sensor for sensing (via monitoring the electrical signals of the attached nerve cells) environmentally relevant compounds such as gaseous or liquid components that are toxic to nerve cells ur unkno~%-n components in order to test their potential toxicity.

B. General Protocols for the Preparation and Use of Non-Chip Substrates - 10 The substrates can be modified in a variety of ways. Non-modified and niodified copolymers can be adsorbed onto suitable surfaces either in pure form or as mixtures. The optimum choice depends on the type and concentration of the ligand molecules, the tareeted receptor, and the substrate properties. Examples of substrates to which the copolymer can be applied include bioanalytical devices such as ELISA or LISA plates, lateral flow assay substrates such as nitrocellulose, nylon, or Polyvinyldiene fluoride PVDF
membranes, polymeric microparticles or inorganic particles, capillaries for electrophoresis, fiber optics, or "lab on a chip" devices.

"1`he materials described herein are particularly suited for these ?o applications in that the polyionic copolymers allow easy and efficient post production modification of the suface. tailored to the desired application, with non-interactive properties and of specific interaction properties. The adsorbing domain of the multifunctional polymer should have opposite charge as that of the substrate prior to adsorption of the polymer, leading to stable and strong adsorption. The considerations for the domains of the copolymer that resist nonspecific adsorption are also similar to those described above. Nonionic water-soluble polymers such as poly(ethylene glycol) are especially preferred. On the termini of these chains may be placed either reactive groups or directly binding groups, to provide for attachment device surfaces. Alternatively, no biological recognition element may be used, leading to a surface that resists nonspecific attachment but does not promote biospecific attachment, or the surface may be pretreated with a reactive moietv and the free surface area that is not derivitized with the reactive moiety subsequently can be "filled in" with either the polymer with no biologicial element that resists nonspecific attachment, or alternatively, with a second protocol.

From the viewpoint of the surface treatment, there are two basic niethods:

I ) In Li type of batch pi-ocess where the surface is functionalized. In a fluid manifold. one or several analytes and reagents are locally applied to the surface. After awaiting the completion or near completion of the bioaffinity reaction (incubation step), the surface is washed in a buffer.

2) In a continuous process where the surface to be functionalized is part of a gaseous or liquid cell or flow-through system. The conditioning of the surface can be done in a continuous and continuously monitored process within that liquid or flow-through cell, followed by in situ monitoring of the signal due to the specific interaction and adsorption or attachment of the specific target molecule in the analyte solution. The original surface may afterwards be restored/regenerated aQain and conditioned for the immediately following next bioaffinity assay. This may be repeated many times.

B. General protocols for the modification of a surface intended to be used for bioanalytical devices other than for screening and detection.
Protocol A

1) Pretreatment of the surface to generate charged surfaced if not inherent to material as obtained.

2) Spontaneous adsorption of a monolayer of the cationic PLL-g-(PEG)jPEG-Y),_, out of an aqueous solution. 1-x denotes the fraction of PEG chains functionalized by reactive molecule Y in comparison to the total number of PEG chains.

3) Rinse 4) Exposure of the surface with an aqueous solution of reactive molecule, binding specifically to the surface ligands.

5) Rinse of the surface to remove loosely (non-specifically) bound reactive molecule.

Subsequent methodology according to standard protocol for the analytical technique.

Pi-otocol B

1) Pretreatment of the surface to generate charged surface if not inherent to nlaterial as obtained.

2) Reaction of the cationic PLL-g-(PEG),(PEG-Y),_, in solution with ligand reactive with molecule Y. 1-Y denotes the fraction of PEG chains functionalized with molecule Y in comparison to the total number of PEG
chains.
3) Spontaneous adsorption of a monolayer of the derivatized cationic PLL-g-(PEG),(PEG-ligand),_, out of an aqueous solution.

4) Rinse 5) Subsequent methodology according to standard protocol for the analytical technique.

Protocol C
1) Pretreatment of the surface to generate charged surface if not inherent to material as obtained.
2) Spontaneous adsorption of a monolayer of the derivatized cationic PLL-g-(PEG) out of an aqueous solution.

3) Rinse 4) Subsequent methodology accordina to standard protocol for the analytical technique.

Protocol D
1) Treatment of the surface with reactive moiety of interest.
2) Rinse Spontaneous adsorption of a monolayer of the derivatized or non-derivatized cationic PLL-g-(PEG) out of an aqueous solution. (as per Protocol A, B, or C) 4) Rinse 5) Subsequent methodology according to standard protocol for the analytical technique.

Examples of various applications of these materials and techniques are provided.

Polystyrene, used for example as microwell plates, can be rendered negatively charged at physiological pH. This material generally supports the adsorption of a wide variety of proteins solution for assay. Microwell plates can be purchased commercially with negatively charged surfaces.

The negatively charged 96 microwell plate is exposed to PLL-g-(PEG),(PEG-VS),_, where VS denotes a vinylsulfone functionality, and coupled to IgG or IgG fragment as in Protocol A. The amount of IgG thus displayed in the microwell may be controlled readily through the quantity x, x = 0 leading to a large amount of exposed IgG and the quantity x = I
leading to no exposed ligand. The amount of antigen contained in a solution niay then be analyzed via conventional ELISA methods. For example, subsequent steps for a standard "sandwich" assay would be incubation of the IgG derivatized microwell plate to the antigen solution for 2 hours to overnight, rinsing three times with PBS or other buffer solution, incubation with a horseradish peroxidase labeled IgG. rinsing with buffer again to remove non-specifically bound HRP labeled IgG, and then detection of bound antigen through a peroxidase generated adsorbing precipitate.

The amount of IgG may also be readily varied by adding stoichiometrically limited amounts of the IgG, and then passivating the other reactive groups if desired by exposure to cysteine or beta mercaptoethanol.

The amount of IgG may also be readily varied by adding the ligand simultaneously with another reactant, such as cysteine or beta mercaptoethanol. The fraction of the VS groups that react with the IgG will be determined by the molar ratios of the two agents and the relative reactivities of the two agents.

Two or more biorecognition elements may be added to the VS sites, either in parallel or in series.

Of importance is that the PEG component of the coating acts as a pre-applied blocking agent, eliminating the need for subsequent blocking steps normally required for microwell plate or other analytical type methods. This ~~

is significant in that it can reduce the time required for the assay by many hours and reduces uncertainty in assay performance which arises from incomplete or variable blocking often observed using conventional blocking agents suc11 as bovine serum albumin.

It is also possible to adsorb the biorecognition element directly to the microwell plate surface, and then subsequently coat the free surface area with a dcrivatized or underivatized PLL-g-PEG following Protocol A or B or D.
Alternatively, if the biorecognition element is not active in the attached form, it can be derivatized with a peptide or other tag that will allow reaction with i o the reactive ligand of the derivatized PLL-g-PEG. For example, a thiol containing tag will react with the vinylsulfone group of (PEG)X(PEG-VS),_x.
The ability to control the binding site of the antigen or antibody can confer higher surface activity.

In another example, lateral flow assay substrates which are commonly formed of nylon 6,6 are modified with the graft copolymer. As with other bioanalytical assays, it is of considerabe interest to limit nonspecific adsorption to the membrane surface. One use of lateral flow assays is for determination of antibody concentrations in a biological solution which are produced in response to exposure to an antigen. Nylon 2o 6,6 is purchased commercially with a negatively charged surface that is stable from pH 3 to 10. The surface is then exposed in the detection region to PLL-g-(PEG),(PEG-VS),_, and rinsed. The remaining material is then exposed as appropriate to PLL-g-PEG with no reactive group to block adsorption to areas outside the detection region and any uncoated regions within the detection region. The PLL-g-(PEG)~(PEG-VS),_X in the detection region is then coupled to an antigen. as in Protocol A. Assay for an antibody to this antigen is performed by flowing a solution to be analyzed through the substrate which results in attachment of the antibody to the bound antigen.
The amount of bound antibody/antigen complex is then determined by exposure of the complex to a labeled anti-IgG and quantitated spectroscopically.

Alternatively, the lateral flo,,v assay surface could be coated with a pre-derivatized PLL-g-PEG as in Protocol B or derivatized with multiple antigens or antibodies. The amount of antigen or antibody present can be varied to optimize the coupling reaction and subsequent detection.
C. General Protocols for the Preparation and Use of Substrates for use in Cell-Based Systems The analytical or tlierapeutic surfaces and substrates may be formed by a number of ways and for a number of purposes, as described below.
From the general perspective of the choice of multifunctional copolymers, io the same principles hold as described above for chip-based devices. The adsorbing domain of the multifunctional polymer should have an opposite charge to that of the substrate prior to adsorption of the polymer, leading to stable and strong adsorption. The considerations for the domains of the copolymer that resist nonspecific adsorption are also similar as to those described above. Nonionic water-soluble polymers such as poly(ethylene glycol) are especially preferred. On the termini of these chains may be placed either reactive groups (as in Protocol A below) or directly binding groups (as in Protocol D below), to provide for attachment to cell surfaces.
Alternatively, no biological recognition element may be used, leading to a ?o surface that resists nonspecific attachment but does not promote biospecific attachment (as in Protocol C below).

Protocol A:
Adsorption of multifunctional polymers onto bioanalytical or biomedical materials may be accomplished as follows.

1) Pretreatment of the polymer or metal surface comprising cleaning under conditions such that the native surface charge is displayed, e.g. the native surface charge at the operating pH of an underlying polymer layer or the surface charge of a metal oxide coating atop a metal substrate. Such cleaning may be accomplished by rinsing, by washing with a detergent and then rinsing to remove detergent, or, for stable materials such as metals, by cleaning in a reactive plasma. Some polymers may be pre-treated as well by plasma processing, especially to introduce surface charge. For example, so-called tissue culture polystyrene is often produced by processing polystyrene in an air or an oxygen plasma, which both cleans the substrate as well as introduces modifications into the surface that are negatively charged at neutral pH. Most metal surfaces, such as stainless steel, titanium and tantalum, are negatively charged at neutral pH, due to the characteristics of the metal oxide overlayer. For the remainder of this protocol, it is assumed that the substrate is negatively charged.

2) Spontaneous adsorption of a nionolayer of the cationic PLL-g-(PEG),(PEG-VS),_c out of aqueous solution. 1-x denotes the fraction of reactively functionalized PEG chains in comparison to the total number of PEG chains. VS denotes a vinylsulfone function, which will react with thiols at neutral or basic pH. A number of other reactive groups can be used, such as maleimides, acrylates and quinines - the only condition is that these groups must not be highly reactive with amines (e.g., on the lysine) at adsorption pH. Amine reactive groups can be used, however, if the positively charged sites on the backbone are substituted with a nonreactive group, such as would be obtained by the use of poly(arginine) rather than PLL.

3) Rinse 2o 4) Exposure of the material surface to peptide comprising a cysteine residue in aqueous solution at a neutral or basic pH, e.g. from pH 7.5 - 9.
The rate of reaction is dependent upon the pH of the solution, being more rapid at basic pH than at neutral pH.

5) Rinse Protocol B
Adsorption of multifunctional polymers upon substrates that are positively charged, such as an amino-containing or amino-modified polymer or aluminum oxide upon aluminum, can be obtained exactly as described above, except by substituting an anionic polymer, such as polyglutamic acid or polyaspartic acid or polyacrylic acid for the PLL.
Protocol C

Adsorption of multifunctioiial polymers lacking a specific recognition element can also be used, when it is desired to simply block nonspecific interactions and not to promote specific interactions. This can be achieved exactl_y as obtained in Protocols A or B. using polymers where x = 1.

Protocol D

The biolo(yical recognition element can be attached to the nlultiftulctional polymer prior to adsorption to the substrate, as described below.

Adsorption of multifunctional polymers onto bioanalytical or biomedical materials may be accomplished as follows.

1) Pretreatment of the polymer or metal surface comprising cleaning under conditions such that the native surface charge is displayed, e.g. the native surface charge at the operating pH of an underlying polymer layer or the surface charge of a metal oxide coating atop a metal substrate. Such cleaning may be accomplished by rinsing, by washing with a detergent and then rinsing to remove detergent, or, for stable materials such as metals, by cleaning in a reactive plasma. Some polymers may be pre-treated as well by plasma processing, especially to introduce surface charge. For example, so-called tissue culture polystyrene is often produced by processing polystyrene in an air or an oxygen plasma, which both cleans the substrate as well as introduces modifications into the surface that are negatively charged at neutral pH. Most metal surfaces, such as stainless steel, titanium and tantalum, are negatively charged at neutral pH, due to the characteristics of the metal oxide overlayer. For the remainder of this protocol, it is assumed that the substrate is negatively charged.

2) Spontaneous adsorption of a monolayer of the cationic PLL-g-(PEG),(PEG-RE),_, out of aqueous solution. 1-x denotes the fraction of functionalized PEG chains in comparison to the total number of PEG chains.
RE denotes a recognition element function, such as a peptide or protein or saccharide or polysaccharide.
3) Rinse These techniques are useful for example, with cell culture substrates, which are typically formed of polystyrene. The polystyrene is or can be surface modified so as to render it negativelv charged at physiological pH.
This material generally supports the adsorption of a wide variety of proteins ~ fi-om cell culture medium, including fibronectin and vitronectin from serum-containing mediunl. a situation which prevents the investigation of biospecific interactions between cells and their substrate.

Tissue culture polystyrene is exposed to PLL-g-(PEG),(PEG-VS),_T
and coupled to the adhesion peptide GCGRGDSPK, as in Protocol A. The io amount of peptide thus displayed upon the culture substrate may be controlled readily through the quantity x, x = 0 leading to a large amount of exposed peptide and the quantity x = I leading to no exposed peptide. Cell behavior on the bioanalytical substrate may be measured, e.g. adhesion, spreading, growth, migration, or higher order functional indicators, such as 15 the production of growth factors, cytokines and chemokines, the expression of matrix proteins, and so forth.

The amount of grafted peptide may also be readily varied by adding stoichiometrically limited amounts of the peptide GCGRGDSPK, and then passivating the other reactive groups if desired by exposure to cysteine or 20 beta mercaptoethanol.

The amount of grafted peptide may also be readily varied by adding the peptide GCGRGDSPK simultaneously with another reactant, such as cysteine or beta mercaptoethanol. The fraction of the VS groups that react with the peptide biorecognition element will be determined by the molar 25 ratios of the two agents and the relative reactivities of the two agents.

Two or more biorecognition elements may be added to the VS sites, either in parallel or in series.

Nonpeptide biorecognition elements may also be added to the VS
groups, e.g. thiol-containing sugars or polysaccharides. Proteins may readily 30 be added to the VS groups, e.g. by exposure to a protein that contains one or more free cysteine residues.

In anotlier example. the cell phenotvpe in therapeutic bioreactors can be nianipulated by application of the graft copolymer to the cell culture substrate. The use of cells as therapeutic agents has been extensively explored. An example is the use of hepatocytes in extracorporal bioreactors as a liver support device in patients -ith liver failure. Problems in such approaches have been encountered in preventing the overgrowth of unwanted cell sub-populations and in maintaininci the proper phenotype of the cells, both in this and other examples. This may be minimized as follows.

Anionic microcarrier beads are treated by exposure to PLL-g-~ o (PEG),(PEG-VS),_, and then grafting -,vith a thiol-containing galactose moiety, as in Protocol A. Hepatocytes bind to galactose via the asialoglycoprotein receptor, which is absent on most other cells, such as tibi-oblasts that may be present in a clinical preparation of hepatocytes intended for therapeutic use in a bioreactor. It is also known that adhesion of hepatocytes to substrates via galactose binding to the asialoglycoprotein receptor can maintain the hepatocyte phenotype more effectively than, for example, adhesion to collagen.

Hepatocytes are seeded on the microcarriers and allowed to attach in a spinner flask as usual. The microcarriers are then placed in a bioreactor, e.g. within the shell space of a hollow fiber bioreactor, with human plasma perfused from the patient, via a blood cell separator, to remove toxins from the plasma.

In still another example, a coronary artery stent, formed from stainless steel, is exposed to PLL-g-PEG as in Protocol C, to block nonspecific protein adsorption and thus blood platelet adhesion upon the surface. The stent may be coated either prior to mounting on a deployment catheter or after, and it may be dried after the final rinse procedure or not.
It may thus be coated either in a manufacturing facility or intraoperatively.
The treated stent is deployed within the coronary artery as usual in the treatment of coronary artery disease.

A coronary artery stent, formed from stainless steel, is exposed to PLL-g-(PEG)x(PEG-VS),_, and then to the endothelial cell-specific peptide GCGREDVG. as in Protocol A. The treated stent is deployed in the coronary artery as usual. In this case. the PEG component serves to reduce the adsorption of plasma proteins, including fibrinogen. and the corresponding in-stent thrombosis and restenosis that may result therefrom.

~ The biological recognition element GDGREDVG serves to promote the attachment and overgrowth of endothelial cells, to provide for a more natural biological covering of the stent surface after deployment and endothelial cell migration over the stent.

The stent may also be treated with the multifunctional polymer io already grafted with the biological recognition element. A coronary artery stent, formed from stainless steel, is exposed to PLL-g-(PEG),(PEG-GC.GREDVG),_, , as in Protocol YYD, with x = 0.75.

IV. Means for Detection and/or Quantitation of Analyte Binding.
A preferred technique is the use of optical waveguide techniques, 15 where the evanescent field is able to sense the analyte that has specifically interacted with the recognition unit in the adsorbed layer of the functionalized PEG-grafted polyionic copolymer. The advantage of the technique is the fact that it only senses analyte molecules close to the surface (i.e. adsorbed or attached to the surface) and not the analyte molecules in the 20 bulk of the solution to be analyzed. A second advantage is the high sensitivity of the technique, which is typically of the order of of the order of 1 ng/cm2 of adsorbed analyte for sensors based on changes of the effective refractive index (such as grating coupler sensors). The detection sensitivity can be further increased if fluorescently labeled target molecules are used 2i and the fluorescence signal excited by the evanescent field is used. As a consequence. substrates, chips or devices are preferred which comprise an optical waveguide with an optically transparent waveguiding layer.
"Optically transparent" in this context shall mean transparency at least at the wavelength of an adequate excitation light source and optionally of the 30 fluorescence or more general luminescence generated by the excitation, typically in the visible or near infrared, for interrogation of the analyte molecules.

The optical waveguides can be designed as self-supporting single- or multilayer systems. Glass plates or glass or transparent polymeric fibers without cladding are examples of self-supporting single-layer systems. For purposes of enhanced sensitivity. however, substrates, chips or devices are s preferred which comprise an optical thin film waveguide comprising a first opticalkti-ansparent layer (a) deposited on a supporting second layer (b) with lower refractive index than layer (a). Especially in case of support materials (second layers (b)) which are only partially or non-transparent or are fluorescent themselves, it is preferred, if the substrates, chips or devices comprise another optically transparent layer (b') with lower refractive index than layer (a) and a thickness between 5 nm and 10000 nm, preferably between 10 nm and 1000 nm, which is located between layers (a) and (b) adjacent to layer (a).

In general, however, using optical waveguides with or without fluorescence detection, it is preferred that the substrates (layer (b)) are optically transparent, such as glass or transparent polymers, have to be used.

Suitable waveguiding layers that are deposited onto the transparent substrate are high-refractive-index transparent materials. Examples include oxides of titanium. silicon. tantalum. niobium. hafnium, zirconium, or mixtures thereof. An additional possibility is to coat the waveguiding layer with an additional thin coating that is particularly suited to attach the polyionic polymer backbone. The only requirements are that this additional coating has to be transparent and thin enough not to adversely affect the optics of the waveguiding process. Other examples are metal nitrides, oxynitrides, carbides, or borides.

Such inorganic layers are generally deposited by physical or chemical vapor deposition or by wet-chemical techniques such as sol-gel techniques onto a suitable substrate such as glass or polymer. It is preferred that the substrates, chips or devices include one or more optical coupling elements for incoupling the excitation light into the optically transparent layer (a).
These optical coupling elements can be selected form the group of prism couplers, evanescent couplers comprising joined optical waveguides with overlapping evanescent fields, end face couplers with focusing lenses, preferably cylindrical lenses, located adjacent to the waveguiding layer (a), and grating couplers. Preferred grating couplers are diffractive gratings selected from the group of relief gratings of any profile, such as rectangular, ti-iangular or sinusoidal profile. and phase gratings or volume gratings with a periodical modulation of the refractive index in the optically transparent layer (a). For ease of production and its reproducibility, diffractive relief gratings with a rectangular profile are the most preferred optical coupling elements.

Most of the metal oxides (and other metal compounds) carry a positive or negative charge at or near to neutral pH. In particular, titanium oxide, tantalum oxide and niobium oxide, with isoelectric points (IEP) below 7, spontaneously acquire a negative charQe in contact with aqueous environments at neutral pH and are therefore suitable to be used in combination with the polycationic polymers. Another possibility is to artificially impose a positive or negative charge onto the waveguiding surface, e.g. through introduction of functional groups such as carboxylate, sulfonate or phosphate or alternatively of positive charges through introduction of fiinctional groups such as amines. There are a variety of techniques known that are suitable for such a pretreatment. Examples include silanization with amino-terminated silanes such as aminopropyltriethoxysilane (APTES) or other silanes carrying a positive or negative charge upon contact with an aqueous environment.

A further sensing technique is the surface plasmon resonance (SPR) technique, which uses an evanescent field generated in thin metal films. If this metal film spontaneously forms a passive oxide film or can be artificially oxidized, this oxide film with positive or negative charge in contact with an aqueous medium can again be used directly to attach polyionic polymers of the right charge. Since noble metal surfaces such as gold often used in SPR

are generally not covered by an oxide film, the surface has first to be treated by chemical or electrochemical techniques in order to generate positive or negative charges. A preferred pretreatment technique is the use of self-assembled monolayers of alkane thiols well known to form ordered nlonolayers on gold and silver surfaces. If bifunctional thiols are used carrying a functional group in the c,)-position. which will acquire a positive or negative charge in contact with aqueous environments, such pretreated metal stu-faces can again be used in combination with polyionic copolymers in the saune or similar way as discussed above for the metal oxide surfaces with spontaneously formed charges. Examples include thiols with a terminal amino, quartenary ammonium, carboxylate or phosphate group.

Independent of the type of substrates and surfaces used, cleaning of i o the surface is an important aspect of this technology. Controlled surface cleanliness is necessary to ensure an optimum degree of adsorption or attachment of the polyionic copolymer to the positively or negatively cllarged device surface. In the case of the metal oxides discussed above, several cleaning procedures have been tested and optimized for the intended use of the polyionic copolymers in the analytical and sensing area. These include organic solvent cleaning, acid treatment, UV/ozone treatment, glow discharge, plasma cleaning and combinations (see Examples 1 and 2).
Plasma cleaning can be done in an oxygen, water vapor, nitrogen or argon gas at reduced pressure. The cleaning of the oxide surface is important to 2o reduce the natural hydrocarbon concentration and or to increase the number of available hydroxy groups at the oxide surface. The hydroxy groups are known to be essential in the formation of charges in contact with an aqueous environment. Plasma treatment in water vapor is preferred if the aim is to increase the number of hydroxyl groups at the surface.

Alternative excitation and detection methods are commonly known and include fluroescence excitation via direct interaction with an excitation source (not through evanescent field excitation), chemiluminescent, adsorption and radioisotope labeling. In all spectroscopic cases, the wavelength used is dependent on the absorbing or emitting molecule as well as the substrate, and can range for example from the near Infra-red to the visible region of the spectrum. Adsorption and fluorescence are commonly used in assays such as lateral flow assays, fiber optic based assays, or multiwell plate based assays in which either transmission or reflection mode are employed. ln cellular assays. techniques include fluorescence based microscopy and two photon techniques.

V. Fabrication of Patterned Surfaces Two-dimensionally patterned surfaces are an important aspect of modei-n analytical and sensor devices such as biochips used today or in the future in DNA, RNA or protein microarray and immunoassay technologies.
The technique allows the parallel detection of many different target molecules in one analyte of complex composition through specific surface fiulctionalization and recognition site immobilization on a local scale. The three basic prerequisites are a surface technology that allows one to reproducibly fabricate geometrically patterned surfaces, a technique to immobilize i-ecognition units on a localized scale and detection techniques that are compatible with the local scale of the individual assay.

PEG-grafted, polyionic copolymers are suitable to fabricate patterned surfaces on a millimeter to micron scale. A preferred way is to use the established microfluid patterning technique. A typical application involves a polymeric stamp that has a three-dimensionally structured surface, e.g. a number of channels in one or in two directions. This stamp is brought in close contact with an analytical or sensor chip surface forming an array of channels between the stamp and the chip surface. The non-functionalized PEG-grafted polycationic copolymer solution is introduced into the stamp/chip fluidic channel network either by capillary forces or by a forced flow using a pump. After having adsorbed the polyionic copolymer A

according to the invention onto the chip surface along the channels, the cliannels are rinsed with pure water and the stamp and chip separated. In a next step, the whole surface can be dipped to attach another polyionic copolymer B such as a functionalized polyionic copolymer. The product is a surface with an array of lines A/B/A/B/ etc. The lines of such an array may have width from the mm scale to the micron or submicron range.
Alternatively, the fluidic patterning can be applied once more with a different stamp pattern or with the same pattern but at a different angle, e.g. at right anole to the first pattern, forming a rectangular or square array. In a subsequent step, the lines B that have reactive functional groups can be reacted with a suitable recognition molecule such as an antibody for proteins or a single stranded DNA or RNA fragment. This can be done by precision ink _jet techniques, by stamping or - again - by microfluid patterning using a different or differently oriented pattern.

Another technique that allows the formation of specific patterns using polyionic copolymers is micro contact printing ( cP). A polymer stamp with a three-dimensionally structure surface is impregnated with the polyionic 1 o copolymer and transferred in a stamping process to the flat chip surface.
A third alternative is to start with a patterned sensor chip surface fabricated e.g. by a lithographic technique. The pattern has regions of positively and negatively charged surfaces. An example is a surface with square islands made from niobium oxide in a sea of aluminium oxide. If this surface is brought into contact with an aqueous solution of a polyanionic copolymer according to the invention at neutral or near neutral pH, only the positively charged aluminum oxide surface (and not the negatively charged niobium oxide pattern) will be coated by the negatively charged polymer.
The opposite situation holds for a polycationic copolymer.

?o A fourth technique is mechanical patterning where the polymer is locally applied through spotting techniques using instruments such as pencils or capillaries.

These techniques can be used to make both types of patterns: islands formed by modified or functionalized polymer in a sea of non-modified polymer or vice versa. Also, the locally adsorbed polymers may be combined with other type of molecules to backfill empty space at the surface.
VI. Applications The materials described here have a variety of applications in the area of substrates or devices (called `chips' in the general sense) for analytical or sensing purposes. In particular, they are suited for the surface treatment of chips intended to be used in analytical or sensing applications where the aim is specific detection of biologically or medically relevant molecules such as peptides. proteins. oligonucleotides. DNA or RNA fragments or generally any type of antigen-antibody or key-lock type of assays. Particularly if the analyte contains a variety of molecules or ionic species, and if the aim is either to specifically detect one molecule or ion out of the many conlponents or several molecules or ions out of the many components, the invention provides a suitable basis for producing the necessary properties of the chip surface_ 1) the ability to withstand non-specific adsorption and 2) the ability to introduce in a controlled way a certain concentration of recognition eiitities. which will during the analytical or sensing operation interact specifically with the target molecules or ions in the analyte (see Figure 2B
for illustration). If combined with suitable analytical or sensor detection methods, the invention provides the feasibility to produce chips that have both high specificity and high detection sensitivity in any type of analytical or sensing assay, in particular in bioaffinity type of assays.

The materials described here additionally have a variety of applications in the area of substrates or devices which are not "chip" based applications. In particular, for use in analytical or sensing applications where the aim is specific detection of biologically or medically relevant molecules such as peptides, proteins. oligonucleotides, DNA or RNA fragments or 2o generally any type of antigen-antibody or key-lock type of assays.
Particularly if the analyte contains aN-ariety of molecules or ionic species, and if the aim is either to specifically detect one molecule or ion out of the many components or several molecules or ions out of the many components, the materials and methods described herein provide a suitable basis for producing the necessary properties of the chip surface: 1) the ability to withstand non-specific adsorption and 2) the ability to introduce in a controlled way a certain concentration of recognition entities, which will during the analytical or sensing operation interact specifically with the target molecules or ions in the analyte. If combined with suitable analytical or sensor detection methods, the methods provides the feasibility to produce assay surfaces that have both high specificity and high detection sensitivity in any type of analytical or sensing assay. in particular in bioaffinity type of assays.
In substrates and surfaces that interact with cells, whether in vitro or in vivo, the interaction between the cell and its substrate plays an important role in controlling its behavior. As in the other bioanalytical systems, the ~ dual issues of prevention of nonspecific interactions and presentation of specific ligands is relevant. In this case. the nonspecific adsorbates of concern are adhesion-promoting proteins and polysaccharides, as well as other biopolymeric materials present in biological fluids. For example, the number of adhesion proteins present in blood plasma or serum is very high, j o and as such it is difficult to obtain a biospecific response in the absence of biospecific interactions. A multifunctional polymer may be used to suppress the nonspecific adsorption of these components in biological fluids.
Moreover, a specific ligand or group of ligands can be incorporated into the multifunctional polymer, preferably at or near the termini of the chains that 15 prevent the nonspecific interactions. Such ligands can the bind to a more limited set of cell-surface receptors than would otherwise be achieved with the spontaneously and nonspecifically adsorbing components. This can be useful in bioanalytical cell culture systems, in extracorporal therapeutic systems, and in implanted devices.

20 The methods can be applied to chips for any type of qualitative, semiquantitative or quantitative analytical or sensing assay. Particularly suitable detection techniques to be combined with chips include:

1) The optical waveguide technique, where the evanescent field is used to interact with and detect the amount of target molecules adsorbed to the chips 25 surface. The technique relies on incoupling white or monochromatic light into a waveguiding laver through an optical coupling element, preferably a diffraction grating or holographic structure. The light travels within the waveguiding layer through total internal reflection. The evanescent wave, extending out of the plane of the waveguide by typically 50 to 200 nm 30 interacts with molecules that are close (e.g. adsorbed or otherwise immobilized) to the surface. In the configuration of grating coupler sensors, typically the incoupling angles a(TE) and/or a(TM) for electric and magnetic inodes are measured, which are altered upon molecular adsorption or desorption onto/from the incoupling grating, due to the resulting changes of the so-called effective refractive index. The effective refractive indices N(TE). N(TM) are calculated for the observed coupling angles on the basis of incoupling condition. Supposing that N(TE), N(TM) has been calculated and the optical parameters of the waveguide laver (nF,dF), of the substrate (nS), of the covering medium (nC) are known, the refractive index (nA) and the thickness (dA) of the added layer can be calculated. Using the model that the refractive index in the adsorbed layer linearly depends on the concentration of the adsorbed material, the mass per area of the adsorbed material can be calculated.

2) Fluorescence spectroscopy or microscopy where fluorescently labeled target molecules are quantitatively analyzed by measuring the intensity of the fluorescence light.

3) Combination of 1) and 2), where the evanescent optical field is used to excite the fluorescence tags of target or tracer molecules adsorbed onto the chip surface modified. The fluorescence is detected using a fluorescence detector situated on the side opposite to the liquid flow cell. This technique is particularly suitable in view of the very high detection sensitivity of the fluorescence technique in combination with the advantage that practically only the labeled molecules attached to the surface are fluorescence-excited, with minimal contribution from `bulk' fluorescence. The combination of the surface treatment of the chip with this analytical procedure based on evanescent field excited fluorescence detection allows detection with extreme analytical sensitivity and selectivity down to the level of only a few hundred molecules or so.

Therefore, a preferred detection technique is the specific detection of target molecules of one or more analytes in a liquid sample, wherein analyte-specific capture or recognition molecules are immobilized at the polyionic copolymer treated surface of the chip comprising an optical waveguide, fluorescently labeled analyte or analyte analogue or tracer molecules are brought into contact with this surface, excitation light is launched into the waveguiding layer (a), and the fluorescence from the fluorescent labels and generated in the evanescent field of the optical waveguide is detected.
Spectrally selective optical components, such as interference filters or bandpass filters (e.g. longpass filters). or combinations thereof in the optical emission path is helpful to improve the sensitivity and improve the disci-imination of background light or scattered excitation light.

4) The Surface Plasmon Resonance Technique (SPR) where the interaction of surface plasmons in thin metal films resonance condition, i.e., the resonant incidence angle for the escitation of a surface plasmon in a thin 1 o metal film, is changed upon molecular adsorption or desorption into/from the metal film, due to the resulting change of the effective refractive index.

5) Ultraviolet or Visible (UV/VIS) Spectroscopy where the adsorption at a particular characteristic wavelength is used to quantitfy the amount of target molecules adsorbed or attached to the modified surface.
6) Infrared Techniques such as Fourier Transform Infrared (FTIR) Spectroscopy, where the excitation of atomic or molecular vibrations in the infrared region is used to detect and quantify target molecules that have previously been adsorbed or attached to the surface modified chips. Surface oi- interface sensitive forms of IR spectroscopy such as Attenuated Total Reflection Spectroscopy (ATR-FTIR) or Infrared Reflection-Adsorption Spectroscopy (IRAS) are particularlv suitable techniques.

7) Raman Spectroscopy (RS) to detect specific vibrational levels in the molecule adsorbed or attached onto the modified chip surface. Surface- or interface-sensitive types of RS are particularly suitable, e.g. Surface Enhanced Raman Spectroscopy (SERS).
8) Electrochemical techniques where for example the current or charge for the reduction or oxidation of a particular target molecule or part of that molecule is measured at a given potential.

Chip based devices can also be assayed with standard fluorescence or adsorption techniques in which excitation is through light reflected off the substrate surface as opposed to the evanescent field interaction.

Otlier analytical or bioanalytical device surfaces can be used for qualitative, semiquantitative or quantitative analytical or sensing assays.

Non "chip" based substrates also includes fiberoptic substrates. In the case of fiberoptics, techniques as described for "chip" substrates are applicable.

For other non "chip" based substrates vvhich do not support evanescent field excitation or are not a"chip", suitable techniques are described below.

1) Fluorescence spectroscopy or microscopy where fluorescently labeled target molecules are quantitatively analyzed by measuring the intensity of the fluorescence light. The fluorescence is detected using standard detectors i0 positioned either for transmission, or more preferably, for reflection based detection methods. This technique is particularly suitable in view of the very high detection sensitivity of the fluorescence technique for fluorphores with hi'oh emission and excitation cross sections in combination with the advantage that practically only the labeled molecules attached to the surface are fluorescence-excited, with minimal contribution from `bulk' fluorescence. Sensitivity may be additionally improved by conventional methods such as the use of filters to eliminate scatter from the excitation source into the detector or the use of higher sensitivity detectors such as cooled charge coupled devices (CCDs).

2) Adsorption spectroscopy where the adsorption at a particular characteristic wavelength is used to quantitfy the amount of target molecules adsorbed or attached to the surface modified according to the invention through reflection or transmission techniques. For simple assay formats such as lateral flow assays, the detection by visual inspection of a color change in the assay region. The adsorption can cover the wavelength range from the near Infrared to the visible region of the spectrum. The signal can be generated by either a tagged molecule or, as in the case of enzyme linked immunosorbent assays (ELISA) through an enzymatic reaction that generates a colored precipitate. In the latter case, the amount of analyte can be correlated to the precipitate adsorption.

3) Infrared Techniques such as Fourier Transform Infrared (FTIR) Spectroscopy, where the excitation of atomic or molecular vibrations in the infrared region is used to detect and quantify target molecules that have previously been adsorbed or attached to the modified chip surface. Surface or interface sensitive forms of IR spectroscopy such as Infrared Reflection-Adsorption Spectroscopy (IRAS) are particularly suitable techniques.

~ 4) Electrochemical techniques where for example the current or charge tor the reduction or oxidation of a particular target molecule or part of that molecule is measured at a given potential.

The analytical or sensor chips can be used in a variety of ways.
Non-modified and modified copolymers can be adsorbed onto suitable surfaces either in pure form or as mixtures. The optimum choice depends on the type and concentration of the target molecules and on the type of detection technique. Furthermore. the technique is particularly suited for the modification of chips to be used in assays where multiple analytes are determined on one chip, either sequentially or simultaneously."

Examples are microarrays for multipurpose DNA and RNA
bioaffinity analysis `Genomics Chips', for protein recognition and analysis based on sets of antibody-antigen recognition and analyze (Proteomics Chips). Such techniques are particularly efficient for the analysis of a nniltitude of components on one miniaturized chip for applications in biomedical, diagnostic DNA/RNA, or protein sensors or for the purpose of establishing extended libraries in genomics and proteomics. The suitability of these materials for the production of such chips results from the fact that polyionic copolymers can be used for easy and efficient patterning of the chip surface into areas with non-interactive properties and areas of specific interaction. Since the above mentioned multipurpose, array type of techniques often make extensive use of microfluidic techniques and networlcs, the surface modification and functionalization using the copolymers describes herein is particularly suitable to be combined with the microarray approach. The fluidic network can be used to specifically treat localized areas of the chip within the fluidic network, since the technique of modifying the chip surface a simple, fast, cost-effective, spontaneous adsorption process out of an aqueous solution and therefore fully compatible with the microfluidic analytical and sensing concepts.

From the viewpoint of the detection step, there are two basic alternatives:

1) In a type of batch process where the chip is functionalized. In a fluid manifold. one or several analytes and reagents are locally applied to the chip Surl"icc. After mvaiting the completion or near completion of the bioaffinity reaction (incubation step), the chip is washed in a buffer and analyzed using one or a combination of the methods described above.

2) In a continuous process where the chip is functionalized and is part of a gaseous or liquid cell or flow-through cell. The conditioning of the surface can be done in a continuous and continuously monitored process within that liquid or flow-through cell, followed by in situ monitoring of the signal due to the specific interaction and adsorption or attachment of the specific target molecule in the analyte solution. The original surface of the chip may afterwards be restored/regenerated again and conditioned for the immediately following next bioaffinity assay. This may be repeated many times.

In a related but different area. the surface treatment of chips has applications in biosensors, where the aim is to attach and organize living cells in a defined manner on such chips. Since protein adsorption and cell attachment is closely related, this opens the possibility to organize cells on chips in defined way. This can be done by patterning a chip surface into areas that are non-interactive to proteins and therefore cells (e.g. areas with adsorbed non-functionalized PEG-grafted polyionic copolymer) and areas which interact with several or one cell-adhesive protein and where therefore cell attachment, will occur. The latter can be areas without adsorbed non-functionalized PEG-grafted polyionic copolymer or with adsorbed functionalized PEG-grafted polyionic copolymer whereby the function is one that induces interactions with the cell (membrane) e.g. via specific peptides that interact with integrin receptors in the cell membrane.

The detection of specific areas of the pattern can be localized to the specific areas, or can be performed for multiple specific areas simultaneously. In general, an important aspect is the sequential or simultaneous determination of multiple analytes in one or more liquid samples, where the patterned surface is used in microarray assays for the determination of analytes of the group formed of peptides, proteins, antibodies or antigens, receptors or their ligands, chelators or "histidin tag ~ components". oligonucleotides, polynucleotides. DNA, and RNA fragments, enzymes. enzyme cofactors or inhibitors. lectins, carbohydrates.

The one or more liquid samples may comprise body fluids, such as blood, serum, plasm, lymph, urine or tissue fluids or egg yolk.

They also may comprise optically opaque fluids, surface water, soil or plant extracts, bio or process broths, or samples from biological tissue.

In summary, the materials and methods described herein can be used in many application areas, e.g., for the quantitative or qualitative determination of chemical, biochemical or biological analytes in screening assays in pharmacological research, combinatorial chemistry, clinical or preclinical development, for real-time binding studies or the determination of kinetic parameters in affinity screening or in research, for DNA and RNA
analytics and the determination of genomic or proteomic differences in the genome, such as single nucleotide polymorphisms, for the determination of protein-DNA interactions, for the determination of regulation mechanisms for mRNA expression and protein (bio)synthesis, for toxicological studies and the determination of expression profiles, especially for the determination of biological or chemical markers, such as mRNA, proteins, peptides or lowmolecular organic (messanger) compounds, for the determination of antigens, pathogens or bacteria in pharmacological product research and development, human and veterinary diagnostics, agrochemical product research and development, symptomatic and presymptomatic plant diagnostics, for patient stratification in pharmaceutical product development and for the therapeutic drug selection, for the determination of pathogens, harmful compounds or germs, especially of salmonella, prions, viruses and bacteria, especially in nutritional and environmental analytics.

The present invention will be further understood by reference to the following non-limiting examples.

Example 1. PLL grafted with monomethoxy PET, PLL-g-PEG;
Application to Wave Guide Substrate.
The following describes the use of PLL grafted with non-modified monomethoxy PET. PLL-g-PEG, for which the polymer architecture has heen optiniized to achieve optimum or close to optimum protein resistance for application in analytical or sensinQ tasks. For clarity, the following system of abbreviations will be used when referring to the various polymers discussed in this paper: PLL(mol. wt. PLL)-g[graft ratio]-PEG(mol. wt.

t o PEG) signifies that the graft copolymer has a PLL backbone of molecular weight of (moi. wt. PLL) in kD. a graft ratio of lysine-mer/PEG side chain and PEG side chains of molecular weight (mol. wt. PEG) in kD.

.ti:11171hesi,, uJ'I'LL---PE(y Table 1 shows the details of the masses and solvents that were used to synthesize the different polymers according to the following procedure. PLL
of mol. wt. 20,000 or 375,000 (Sigma, St. Louis, MO, USA) was dissolved in 50 mM sodium borate buffer (SBB), pH 8.5. The solution was filter sterilized (0.2 m pore-size filter). Monomethoxy PEG-nitrophenyl carbonate. mol. wt. 5000 (Shearwater Polymers. Huntsville, AL, USA) or hydroxysuccinimidyl ester of inethoxypoly(ethylene glycol) proprionic acid mol. wt. 2000 (SPA-PEG, Shearwater Polymers Europe, Inc., NL) was either quickly dissolved with stirring in 2.5 mL 50 mM SBB, pH 8.5 or taken as a solid and added to the dissolved PLL. The reaction was allowed to proceed for 6 hr at room temperature, after which the reaction mixture was dialyzed (SpectraTM-Por, mol. wt. cutoff 12-14,000; Spectrum, Houston. TX, USA) for 24 hr. first against phosphate buffered saline (PBS; 0.2 g/L KCI, 0.2 g/L
KH,POõ 8 g/L NaC1, 1.15 g/L anhydrous Na,HPO41pH 7.4 t 0.1, 285 mOsm/kg HZO t 5%), and subsequently against deionized water. The product mixture was freeze-dried and stored at -20 C under Ar. PLL(375)-g(5.6)-PEG(5) -'H-NMR (D,O): ppm - 1.35, 1.60, 1.68 (-CH2-); 2.88 (-CH2-N-); 3.55 (PEG); 4.20 (-N-CHR-COO-). By NMR, the areas of the lysine side chain peaks were compared with the area of the PEG peak to determine the ~~ratt ratio of the conib copolymer.

A dendron copolymer (Dendron-5) was prepared according to the following procedure. Monomethoxy PEG of molecular weight 20.000 (PEG
20K: Shearwater Polymers, Huntsville_ AL. USA) was dried by azeotropic distillation from benzene. The hydroxvi terminus of the PEG was first esterified with 9-fluorenylmethyloxycarbonyl Fmoc-Gly. The anhydride of Fmoc-Gly was produced by reaction of Fmoc-Gly (1.78 g; 8 equiv.; Novabiochem, San Diego, CA, USA) with diisopropylcarbodiimide (0.469 mL; 4 equiv.; DIPCDI; Aldrich) in dimethylformamide (DMF; 10 mL: anhydrous, Aldrich) and io dicliloromethane (DCM; 4 mL; anhydrous, Aldrich) for 30 min, room temperature with stirring under argon. The PEG (15 g; I equiv.) was dissolved in DCM (30 mL). Dimethylaminopyridine (91.6 mg; I equiv., Novabiochem) in DCM (5 mL) was added to the dissolved PEG, and this was added to the Fmoc-Gly anhydride. The vessel that contained the PEG

- 5 was washed with 5 mi DCM, which was added to the reaction mixture. The reaction proceeded for 6 hr at room temperature with stirring under argon.
The reaction mixture was filtered through paper under vacuum and then precipitated in cold, rapidly stirring ether, and collected by vacuum filtration.
This resulting product constituted the zeroth-generation dendron copolymer.

2o The following reaction sequence was repeated five times to yield, Dendron-5, a fifth generation dendron copolymer.

Subsequent Fmoc-protected amino acids were added to the PEG as follows. Fmoc groups on the coupled lysine residues were removed by dissoiving the Fmoc-protected. lysine-grafted PEG in 20% piperidine in 25 DMF (Perseptive Biosystems, Framingham, MA, USA; 4 ml/g PEG 20K), lieating with swirling at 45 C until dissolved, then allowing the mixture to stand at room temperature for 30 min, followed by precipitation in cold, stirred ether, vacuum filtration and drying under vacuum, with 500 mg of each PEG-amine product retained. The Fmoc-amino acid (3 equiv.; Fmoc-L-30 Lys(Fmoc)-OH; Bachem, King of Prussia, PA. USA) and hydroxybenzotriazol HOBT (3 Equiv.; Novabiochem) were dissolved in DMF (3 ml/g amino acid), and DCM (2 ml/g amino acid). DIPCDI (3 equiv.) was added, and after 15 min the PEG

WO 00/65352 PCT/[JS00/11708 (2ni1 DCM/g PEG) was added (the vessel that contained the PEG solution washed with 5 ml DCM. 2 times). The reaction proceeded with stirring at room temperature for at least 6 hr under argon. The reaction mixture was then precipitated in stirred cold ether. vacuum filtered, and dried under vacuum.

The dendron product was analN=zed by gel-permeation chromatocraphy (GPC) at 1% in DMF (Polvmer Laboratories PL-EMD 950 evaporative mass detector; columns - Polvmer Laboratories 5 pm Mixed D
300 x 7.5 and Polymer Laboratories 5 m 500 A 300 x 7.5 in series, Polymer io Laboratories, Amherst, MA, USA). By comparing the chromatograms from the mass detector, which allows a measurement of the mass of polymer, with the UV adsorption at 300 nm (Fmoc absorption ,oo = 6558 L/cm mol), the percentage of coupling was calculated.
Pluronics' F-108 NF and F-68 NF were obtained from BASF (Mount Olive, NJ, USA).

Sensor Chips All of the waveguides used in this study were purchased from Microvacuum, Ltd. (Budapest. Hungary) and comprise a 1-mm-thick AF45 glass substrate, covered with a waveguiding layer with the following specifications: waveguide material (Sol-Gel technique): Si,Ti,_x O,, where x=0.25 0..5, refractive index (nF)=1.77t0.03, thickness (dF)=170-220 nm, Substrate glass slide, length (L) = 12 mm, width (w) =16 mm, thickness (H)=0.55 mm, refractive index (ns) = 1.53, grating periodicity: 2400 lines/mm (0.4166 m).

For experiments involving titanium and niobium oxide surfaces, an additional 14-nm-thick oxide layer was sputter coated in a Leybold dc-magnetron Z600 sputtering unit onto the waveguiding layer. All of these surfaces were characterized by X-ray photoelectron spectroscopy XPS, Atomic Force Microscopy AFM and Time of Flight Secondary Ion Mass Spectroscopy ToF-SIMS.
Before each experiment, the waveguides were cleaned according to the following procedure: sonication in 0.1 M HCI for 10 min, extensive rinsing with ultrahigh purity waler and drying under nitrogen, followed by 2 min of oxygen plasma cleaning ir, i::arrick Plasma Cleaner/Sterilizer PDC-32G

instrument (Ossining, NY, USA).

Grating Coupler System The technique involves the incoupling of excitation light beam from a He-Ne laser into a planar waveguide that allows for the direct online monitoring of macromolecule adsorption. The method is highly sensitive (i.e. -i ng/cm') up to a distance of 100 nm above the surface of the waveguide. The technique allows for the in situ, real time study of adsorption kinetics. Areal adsorbed mass density data were calculated from the thickness and refractive index values derived from the mode equations 1 o according to Feijter's formula. A value of 0.182 cm'/g for dn/dc was used for the protein adsorption calculation, and a value of 0.202 cm;/g as determined in a Raleigh interferometer was used for the PLL-g-PEG
adsorption calculations. All grating coupler experiments were conducted in a BIOS-I instrument (ASI AG, Switzerland) using a Kalrez (Dupont, USA) flow-through cell as described previously. The flow though cell was used for studying both PLL-g-PEG adsorption and protein adsorption. The flow rate and wall shear rates were 1 mL/hr and 0.83 s', respectively. The calculation of adsorbed masses was done according to the following procedure:

Protocol of the Adsorption Experiments Protoc=ol for the adsorption of PLL-g-PEG on waveguide surface.
The samples were ultrasonically cleaned in 0.1 M HCl for 10 min, extensively rinsed with ultrahigh purity water and dried in a nitrogen stream, followed by 2 min of oxygen-plasma cleaning as described earlier. PLL-g-PEG modified surfaces were prepared by dip coating for 10 min in a 1 mg/mL solution of PLL-g-PEG in 10 mM HEPES (4-(2-hydroxyethyl)piperazine-l-ethanesulfonic acid, adjusted to pH 7.4 with 1M
NaOH solution). This buffer solution will be referred to as HEPES Z1 hereafter. Subsequently, the modified waveguides were rinsed immediately with ultrapure water and dried under nitrogen. Some samples were analyzed and used without a proper cleaning step as described in the sample preparation procedure to test the effect of surface contamination on the adsorption and performance of PLL-g-PEG.

Protocol for optical gi-ating coupler experiment crnd saniple preparalion Samples with adsorbed PLL-g-PEG layers were prepared as described above and dried in flowin(i nitrogen. Their protein and serum adsorption performance was subsequently measured in the grating coupler svstenl as described below.

Samples modified with PLL-g-PEG in situ were initially placed in HEPES Z1 immediately following the cleaning procedure and allowed to soak overnight. Prior to assembling the flow-through cuvette in the grating 1 o coupler instrument, the samples were rinsed with ultrapure water and dried under nitrogen. These presoaked samples equilibrated and reached a flat baseliile in HEPES Z1 in less than 1 hr. Then. the samples were exposed in Situ to the PLL-g-PEG solution (1 mg/mL in HEPES Z1). The adsorption was subsequently monitored for 30 min. The polymer solution was then replaced with HEPES Z1, and the protein and serum adsorption performance was measured as described later.

Protocol fot- grating coupler experiment and sample preparation Samples with adsorbed PLL-g-PEG layers were prepared as described above and dried in flowing nitrogen. Their protein and serum 2u adsorption performance was subsequently measured in the grating coupler system as described later. Samples modified with PLL-g-PEG in situ were initially placed in HEPES Z1 immediately following the cleaning procedure and allowed to soak overnight. Prior to assembling the flow-through cuvette in the grating coupler instrument, the samples were rinsed with ultrapure water and dried under nitrogen. These presoaked samples equilibrated and reached a flat baseline in HEPES Z1 in less than 1 hr. Then, the samples were exposed in situ to the PLL-g-PEG solution (1 mg/mL in HEPES Z 1).
The adsorption was subsequently monitored for 30 min. The polymer solution was then replaced with HEPES Z 1, and the protein and serum adsorption performance was measured as described later.

Si0.6Tio 40, waveguides were used for the pH-dependence measurements and were prepared as described in the immediately preceding procedure. However, in this case. the solution was 10 mM HEPES titrated to the predetermined pH by the addition of either 1 M NaOH or 1 M HCI. The polymer concentration was 0.1 mg/ml in the same pH-adjusted HEPES
solution. After 1 hr of polymer adsorption and 30 min of washing with the pH adjusted solution, the solution was changed to the pH 7.4 buffer (HEPES
Z 1). and the protein adsorption performance was measured as described later in this section. The same procedure was used for the ionic-strength-dependence experiments, except that NaC1 was used to generate the solution of predetermined ionic strength.

i o Protocol of the protein adsorption experiments The waveguides were exposed to a solution of human serum (Control Serum N, Art.# 07 3711 9, US# 42384 and human y-globulin, Roche (Switzerland)) for 1 hr at a temperature of 25 C and subsequently washed for 30 min in HEPES Z 1. Human serum albumin (HSA), human fibrinogen and human fibronectin were obtained from Sigma Chemical Co. (USA); and their related antibodies were obtained from Dako A/S (Denmark). In the case of the single protein adsorption experiments, the waveguide was exposed to a I mg/mL solution of the appropriate protein in HEPES Z1 for 1 hr at a temperature of 25 C and subsequently washed for 30 min in HEPES
Z l .

In some cases after the serum exposure, the waveguide was tested against solutions of 0.1 mg/mL rabbit anti-HSA and 0.28 mg/mL rabbit anti-human fibronectin for 30 min at a temperature of 25 C and subsequently washed for 30 min in HEPES Z1. Adsorption of human fibrinogen was tested separately by exposure to a 1 mg/mL solution of fibrinogen for 1 hr at 25 C followed by a 0.1 mg/mL solution of rabbit anti-human fibrinogen for min.

PLL(375)-g[5.6J-PEG(5) adsorption and protein resistance study All of the following measurements were carried out in a flow-through 30 cell, and both the PLL-g-PEG pretreatment and the protein-adsorption tests were carried out in situ and consecutively without an intermittent drying stage unless otherwise noted. The results from grating coupler experiments (see Figure 3) indicate that the PLL-g-PEG polymer spontaneously adsorbed out of a pH 7.4 buffered aqueous solution onto metal oxide surfaces. The example shown in Figure 3 involved the adsorption of the PLL-g-PEG onto three different metal oxide surfaces: titanium, niobium and silicon/titanium.

This adsorption process occurred rapidly and resulted in the formation of a laver of adsorbed polvmer on the surface. Typically for Si,,.,Tio a0, surfaces, a laver with an adsorbed areal density of approximately 125 ng/cm' formed, and 95% of the final observed mass .vas reached within the first 5 min.

Similar behavior was observed for the other two metal oxide surfaces investigated (i.e. niobium pentoxide, titanium dioxide). Although the adsorption kinetics were quite similar. the resulting amount of PLL-g-PEG

adsorbed to the surface was different and depended on the characteristic isoelectric point of the metal oxide. as shown in Figure 4 and Table 2.
Subsequent protein adsorption experiments revealed that PLL-g-PEG

modification of the metal oxide surfaces resulted in sharply reduced protein adsorption. Typically, the exposure of a metal oxide surface to serum produces a layer of adsorbed protein with an areal density between 150 and 250 ng/cm' after the washing step. However, waveguides pre-coated with a self-assembled layer of PLL-g-PEG show a drastic reduction in subsequent ?u serwn protein adsorption. An example of this is shown in Figure 5. This experiment involved modified and unmodified titanium dioxide waveguides that were prepared according to the procedure described above. Generally, PLL(375)-g[5.6]-PEG(5) pretreatment, regardless of the preparation procedure, caused an order of magnitude decrease in the areal density of adsorbed protein after serum exposure on titanium dioxide, silicon/titanium dioxide and niobium oxide surfaces (see Figure 6). Similarly, the adsorption of human serum albumin (1 mg/mL, 10 mM HEPES buffered solution, pH
7.4) was decreased by two orders of magnitude following the PLL-g-PEG
pretreatment (see Figure 6). The quantitative protein adsorption data for all surfaces studied are listed in Table 3.

The residual material remaining on the surface after serum exposure was tested against antibodies of several common serum proteins, such as anti-albumin, anti-f7bronectin, and anti-y-globul.in. These antibodies exhibited adsorbed areal density loNN-er than that detectable by the grating coupler technique (i.e. < 2 ng/cm'). -,1-hich suggests that none of the proteins are present in their active conformations on the modified surfaces. It is likely that serum components other than proteins are picked up by the modified surfaces. Since serum is fibrinogen-depleted, fibrinogen was also tested separately from a 5 mg/mL solution in HEPES Z I, followed by an anti-fibrinogen assay. For PLL(375)-g[5.6]-PEG(5)-modified silicon/titanium dioxide surface, 85 ng/cm' and 150 ng/cm' of adsorbed areal density were l o observed for fibrinogen and anti-fibrinogen, respectively. The antibody results are not shown.

Perfornzance and long-term stabilit}, of the adsorbed PLL(375)-g[5.6]-I'ECi(5) laver Once established on the surface, the adsorbed layer of PLL(375)-g[5.6]-PEG(5) was found to be stable (i.e. <5% loss in mass of the adsorbed layer after one week) and resistant to protein adsorption over 24 hr at 37 C
under a flowing HEPES Z 1 solution, as shown in Figure 7. This experiment involved the in situ deposition within the first hour of a PLL(375)-g[5.6]-PEG(5) layer on the surface of a silicon/titanium dioxide waveguide. Two subsequent exposures to serum produced less than 20 ng/cm2 of surface adsorbed protein. Eighteen hours later, two additional serum exposures similarly produced less than 20 ng/cm'. Similar performance was observed when PBS was used as the buffer instead of HEPES Zl.

PLL(375)-g[5.6]-PEG(5) modified waveguides that were stored dry were found to retain their protein resistant properties after more than three months, and those stored in HEPES-buffered solution were found to retain their protein-resistant properties after more than one month.

Pluronics', diblock copolymers consisting of a poly propylene oxide flanked by two polyethylene oxide chains, are commonly used to immobilize PEG onto hydrophobic surfaces. However, Pluronics F-108 and F-68 were found not to adsorb onto any of the metal oxide surfaces investigated here and as a result did not show any protein adsorption suppressing properties.

PLL(375)-g[5.6]-PEG(5) was also found to adsorb onto a pre-adsorbed layer of serum proteins. After a typical adsorption of serum protein (i.e. about 260 ng/cm'), subsequent exposure to a I mg/mL solution of PLL(375)-g[5.6]-PEG(5) effected an additional layer of polymer with a surface areal density of approximately 90 ng/cm'.

Effect of preconlamination on PLL(375)-g[5.6]-PEG(5) adsorption Metal oxide surfaces that exhibited large amounts of hydrocarbon surface contamination nevertheless adsorbed a layer of PLL(375)-g[5.6]-PEG(5) that suppressed subsequent serum adsorption. Titanium dioxide waveguides that were not cleaned according to the procedure described above exhibited substantial hydrocarbon surface contamination. However, these XPS data also indicate that an additional layer of PLL(375)-g[5.6]-PEG(5) does indeed adsorb onto this contaminated surface. Furthermore, optical grating coupler experiments showed that the typical adsorbed areal density of 120 ng/cmz forms on contaminated titanium dioxide waveguides and that this adsorbed layer of polymer suppresses subsequent serum protein adsorption by about 95%. That is, the adsorption and performance characteristics of the PLL(375)-g[5.6]-PEG(5) are identical in the case of both contaminated and cleaned titanium dioxide surfaces.

Effect of polymer architecture on protein interactions Alternative architectures of the polymer were explored for the suppression of protein adsorption performance. These architectures included brush-like graft copolymers with differing PEG side-chain length, PLL
backbone length and PEG grafting ratio (see Table 1). The inverted tree-like dendrimeric PLL having a single PEG side chain attached at the base was also investigated.

Effect oJ the backbone (PLL) structure on subsequent protein adsorption The graft copolymer architecture was found to influence the subsequent serum adsorption suppression. All of the polymers investigated demonstrate adsorption on silicon/titanium dioxide surfaces in the areal density range of about 150 ng/cm', as shown in Figure 8. Of the two comb copolymers with PEG side chain mol. wt. of 5000, the PLL(375)-g[5.6]-PEG(5) demonstrates only a slightly more pronounced suppression of serum adsorption, decreasing the observed adsorption by about 90% to 30 ng/cmz.

The graft copolymer with the lower mol. wt., (PLL(20)-g[6.0]-PEG(5)), decreases the observed protein adsorption from serum exposure by about 85% to 50 ng/cm2. Dendron-5, consisting of a 20,000 mol. wt. PEG with a terminus consisting of a five generation lysine dendrimer, was also found to adsorb onto silicon/titanium dioxide surfaces in the areal density range of 120 to 150 ng/cm2. Furthermore, this adsorbed polymer layer was found to lo decrease the subsequent protein adsorption due to serum exposure by about 75% to 80 ng/cm2.

Effect of the copolymer grafting ratio on subsequent protein adsorption The grafting ratio was also found to influence the subsequent serum adsorption performance. To explore the effect of the grafting ratio on the polymer adsorption and subsequent protein resistance behavior, PLL(20)-g[3.5]-PEG(2) and PLL(20)-g[5]-PEG(2) were synthesized. These two polymers differ only in their respective grafting ratios of 3.5:1 and 5.0:1 (i.e.
lysine monomer : PEG side chain). As shown in Figure 8, PLL(20)-g[3.5]-PEG(2) and PLL(20)-g[5]-PEG(2) exhibited adsorbed mass densities of 170 and 190 ng/cmZ, respectively. However, PLL(20)-g[3.5]-PEG(2) demonstrated far greater suppression of protein adsorption (i.e. a 99%
decrease vs. an 80% decrease for the PLL(20)-g[5]-PEG(2)). In fact, the PLL(20)-g[3.5]-PEG(2) demonstrated the greatest suppression of protein adsorption from serum of all of the polymers investigated in this study. This amount of adsorbed mass is approximately of the same magnitude as the detection limit of the grating coupler technique (i.e. -1 ng/cm2) (see Table 3).

Table 1 Details of synthesis of the different types of PLL-g-PEG. (SBB

is sodium borate buffer (pH=8.5), and STBB is sodium tetraborate buffer (pH=8.5).) All of the molecular weights are in kD.

Polymer PLL PEG
PLL(mol. wt. PLL)-g[Lysine/PEG side chain]-PEG(mol. wt. PEG) PLL(375)-g[5.6]-PEG(5) 500 mg in 10 mL SBB 2.0 g in 2.5 mL SBB
PLL(20)-g[6.0]-PEG(5) 500 mg in 10 mL SBB 2.0 g in 2.5 mL SBB
PLL(20)-g[3.5]-PEG(2) 83.6 mg in 1.05 mL STBB 215.7 mg solid PLL(20)-g[5.5]-PEG(2) 106.8 mg in 1.34 mL STBB 193.2 mg solid E
c~e W N ^-~ x a ~ o 0 x v1 ~,, Q o a an u v v N
c ~

6~ O O ^ p O
~
o O Q O u N ~ O~
ci"
O r.
E
b~A x p Q ~ '~ ~ C
cj~ =O ~ O I~ ~-.

O CC R N ~, CC ~
14) ~..~ H vOi `+ O O ~n O c~d c~n M N V~
y~. CL ~ cNC ~_ E

O bA u -- -- -~
~
U
v a W ~ w ci y ~ s.+ y ,~ ~ ~n O
Z CL U

~ o c ~ C O G1, ~ ~ N
~ O
O
Z
z Table 3 The observed adsorbed areal density of polymer, serum, and fibrinogen on surfaces modified -tvith various PLL-g-PEG copolymers.
[Polymer adsorption carried out under 10 mM HEPES Z1, 1 mg/mL
polymer, 1 mL/hr, T=26 C; serum adsorption, 1 mL/hr, T=26 C;
fibrinogen adsorption, 10 mM HEPES Z1, 1 mg/mL, 1 mL/hr, T=26 C.]
Surface Adsorbed mass of Adsorbed mass of Adsorbed mass of PLL-g-PLL serum [ng/cmz] fibrinogen [ng/cmz]
polymer [ng/cm']

Unmodified - 270 451 SiõaTiO6O, PLL(375)-g[5.6]- 133 30 85 PEG(5) PLL(20)-g[6]-PEG(5) 152 50 3 PLL(20)-g[3.5]- 169 3 15 PEG(2) PLL(20)-g[5]-PEG(2) 194 63 1 Example 2: Application of Biotin-modified PEG-grafted PLL

This example refers to the use of biotin-modified PEG-grafted PLL
for application on chips for bioaffinity sensor assays using the biotin-streptavidin-biotin recognition assay.
Synthesis of the Functionalized, Biotinylated Copolymer PLL of mol. wt. 20,000 (Siama, CH) was dissolved in 50 mM sodium tetraborate buffer, pH 8.5 at a concentration of 40 mg/ml. The solution was filter sterilized (0.2 m pore-size filter). Solid n-hydroxysuccinimidyl ester of methoxypoly(ethylene glycol) proprionic acid (mol. wt. 2,000) (SPA-PEG, Shearwater Polymers Europe, Inc., NL), and -biotin, -hydroxysuccinimidyl ester of poly(ethylene glycol)-carbonate (mol. wt.
3,400) (NHS-PEG-biotin, Shearwater Polymers Europe, Inc., NL) in the desired stochiometric ratio were added to the dissolved PLL solution. The reaction was allowed to proceed for 6 hr at room temperature, after which the reaction mixture was dialyzed (Spectra/Por, MWCO 6-8000, Spectrum, Socochim, CH) against phosphate buffer saline (PBS) for 24 hrs and subsequently against deionised water for 24 hrs. The dialyzed solution was lyophilized for 48 hrs and stored under nitrogen at -25 C.

Four PLL-r-PEG polymers PLL(20)-g[3.5]-,(PEG(2)},iPEG(3.4)-biotin; .,, with a Lys:PEG ratio of 3.5:1, yielding the greatest suppression of protein adsorption from serum as shown from Example 1, and different amounts of biotin 0% (x=1),l0%a and 10%b (2 polymers) (x=0.9) and 50%
(x=0.5) of the PEG chains are biotimflated) were synthesized. In the case of PLL-g-PEG-Biotin 10%b, the NHS-PEG-biotin was added first and reaction allowed for l hr before adding the SPA-PEG. Table 4 reports the quantity of 1 o material used.
Size Exclusion Chromatography Size exclusion chromatography analysis of the polymers was performed in 0.2 M sodium carbonate/bicarbonate buffer solution pH 10 with a ShodexT"' Ohpak column, SB-894HQ (Alltech, Deerfield, IL-USA).
Molecular weights of the different polymers with Lys:PEG of 3.5:1 were all in the range of 70 kD. Molecular weights of PLL-g-PEG with Lys:PEG of 2:1 and 5:1 were approximately 100 kD and 55 kD, respectively.
Comparative and absolute measurement of the enzymatic activity of streptavidin-horseradish peroxidase conjugate immobilized on the 2o biotinylated PLL-g-PEG polymers Substrates For this assay, substrates consisted of 20 nm of pure titanium dioxide (TiO,) produced by physical vapor deposition onto a silicon wafer. The Root-Mean-Square RMA roughness of the Ti02 coating deteirnined via AFM (Biosc_ ii)e.
Digital Instruments) was 0.3 nm (scanned surface: 5 x 5 m`). The wafer was sampled into pieces of 1 cm'' and these were ultrasonically cleaned in acetone (uvasolTM, Merck) and 2-propanol (uvasol, Merck) for 5 min. each, dried under nitrogen (5.0) and subsequently cleaned by oxygen plasma (plasma cleaner/sterilizer PDC-3?G, Harrick) for 3 min.

PLL-g-PEG self-assembly Solutions of PLL-g-PEG-OCH,, PLL-g-PEG-Biotin(10 1oa), PLL-g-PEG-Biotin( I 0%b) and PLL-g-PEG-Biotin(50%) were prepared in 10 mM

HEPES buffer pH 7.4, at a conceniration of I mg/mi and filter-sterilized (0.22 m DuraporeTM Millex, Sigma-CH. Self-assembly of the different polymers on TiO2 (3 samples per polN=mer type) proceeded for 90 min.
Samples were then rinsed extensively with filter-sterilized HEPES buffer pH

7.4 and dried under nitrogen (5.0). Three bare TiO, samples were also preserved as references.

Sirepiavidin horseradish peroxidase adsorption The samples were distributed in a 6-wells plate and covered with 4 ml of a solution of streptavidin-horseradish peroxidase conjugate in PBS pH 7.4, io diluted to 1/1000 in HEPES buffer (pH 7.4). (horseradish peroxidase (HRP) to streptavidin molecular ratio is 5:1; 400 ng streptavidin / 1200 ng horseradish peroxidase). Samples were left for incubation at room temperature for 60 min. They were then rinsed extensively with fresh HEPES
buffer and subsequently with sodium acetate/citric acid buffer pH 6Ø

4uantification of the enzymatic activity of the chemisorbed streptavidin horseradish peroxiduse The samples were immediately transferred into individual wells of a 24-wells plate and incubated at room temperature under gentle shaking in 1.6 mi of a 0.42 mM solution of 3,3'-5,5'-tetramethylbenzidine (TMB, Merck-CH) in sodium acetate/citric acid buffer (pH 6.0, containing 0.004 % (v/v) H,O, and 1% DMSO, previously used to dissolve the TMB). A dilution series (5k, 10k, 100k, 500k, 1 M, 5M, l OM, 50M) of the streptavidin-HRP
PBS solution in sodium acetate/citric acid was also prepared and incubated simultaneously under the same conditions to serve as calibration curve.
After 26 min, as the solutions had visibly turned blue under the enzymatic activity of the peroxidase, the solid samples were removed from the solution and the reaction stopped by adding 100 l of 2 M H2SO4. The solution was subsequently transferred into disposable cuvettes, and the absorption measured at 450 nm (JASCO 7800 UVJVis Spectrophotometer).

Non-speciJic protein adsorption of PLL-g-PEG 2:1, 3.5:1, 5:1; mass of polymer adsorbed and choice of optimum polymer As can be seen from Figure 8, when the PEG interchain spacing is smaller or equivalent to the PEG radius of gyration, non-specific protein adsorption is efficiently prevented. On the contrary, an interchain spacing laruer than the PEG radius of gyration leads to protein adsorption levels co-nparable to the bare TiO2 surface. In addition, one can also note the ~ difference in the polymer mass adsorbed, which is higher for the PLL(20)-g[5]-PEG(2') campared to the PLL(20)-g[3.5]-PEG(2). This is probably related to the fewer anchoring NH3+ groups available in the PLL(20)-g[3.5]-PEG(2) polymer since a larger number of these side chains have been derivatized with PEG. Although the mass polymer adsorbed at the surface of the waveguide is lower, the density of PEG chains is probably high = ~~ enough to lead to chain overlapping and hence sufficient to yield a protein repellent PLL-g-PEG-methoxy interface. Therefore, the PLL-g-PEG-methoxy polymer with a I,ys:PEG
ation of 3.5:1 seems to be an optimum in our PLL-g-PEG polymer series.

As can be seen from Figure 9, activity of the peroxidase on the different surfaces ranks as follows: PLL-g-PEG-methoxy TiO, < PLL-g-15 PEG-Biotin 50% < PLL-g-PEG-Biotin 10 loa < PLL-g-PEG-Biotin 10%b. In parallel, from the grating coupler results, the mass of streptavidin adsorbed ranks as follows: PLL-g-PEG-methoxy (mass below the detection limit) <
PLL-g-PEG-Biotin 10 oa (mass < 33ng/em') < PLL-g-PEG-Biotin 10 lob (niass < 99 ng/cm2) < TiO2 (mass < 160 ng/cm') < PLL-g-PEG-Biotin 50%
20 (mass < 283ng/cm2). The extremely low peroxidase activity level on the PLL-g-PEG-methoxy (close to background noise, see Figure 4) shows that almost no streptavidin adsorbed on the surface.

As the biotin content increases, the activity levels of the peroxidase is significantly increased, as can be seen for PLL-g-PEG-biotin 10%a and -25 10%b. Further increase in the biotin content (-50%) leads to a marked decrease in the peroxidase activity, although the mass of streptavidin adsorbed is higher. At this stage, the high content of biotin at the surface probably drives the unfolding of streptavidin, thus leading to lower activity.

Alternatively steric hindrance of the enzyme molecules as a consequence of too high biotin density may be the cause for this observation. Finally, the bare surface shows less streptavidin adsorption in comparison to the 50%
biotin case, and also lower peroxidase activity, implying that the non-specifically adsorbed streptavidin is also unfolding at the surface of the Ti02 chip thus lowering the activity of the enzyme.

Specific delection ofstreptavidin on PLL-g-PEG-biotin modifaed chips using the grating coupler technique mM HEPES (pH 7.4) buffer solution and SiO,-TiOZ waveguides 10 (Microvacuum Ltd, H) were used for all of the experiments (details: see section 6.1.2). The modified waveguides were prepared in situ by exposing the waveguides to 50 g/ml PLL-g-PEG-biotin polymer solution for twenty minutes in a flow cell apparatus, followed by rinsing with buffer. Then the PLL-g-PEG-biotin modified waveguides were exposed to Control Serum N

(human) (Roche, CH) for thirty minutes and then washed in buffer. At the end of the measurement selectivity was tested with I mg/mi streptavidin (SIGMA, USA) for 20 minutes followed by a rinse with buffer. Mass data were calculated from the thickness and refractive index values derived from the mode equations (see section 5.2). All experiments were conducted in a BIOS-I instrument (ASI AG, Zurich, CH).

The first part of the experiment, shown in Figure 10 demonstrates the protein resistant behavior of the treated oxide surface. Waveguides coated with the PLL-g-PEG-biotin exhibit protein adsorption of less than 2 - 3 ng/cm' after exposure to human blood serum. This value is two-hundred-fold lower than the adsorption seen on untreated waveguides. The second part of the experiment demonstrates the specificity of the sensor. A
monolayer of streptavidin quickly and irreversibly adsorbs onto the same PLL-g-PEG-biotin functionalized surface after exposure to a 1 mg/ml streptavidin solution. Figure 11 shows the amount of PLL-g-PEG-biotin adsorbed onto the chip surface and the amount of streptavidin subsequently adsorbed onto the PLL-g-PEG-biotin covered chip surface for three different biotin concentrations in the PLL-g-PEG-biotin molecule (see Table 5). It is obvious that the modified polymer adsorbs onto the chip surface and shows an excellent specificity in the subsequent streptavidin sensing assay.
Furtherinore. the amount of streptavidin specifically detected is related to the degree of biotinylation. The relation between the amount of streptavidin detected and the biotin surface concentration is linear within the experimental error and in the range of biotin concentrations tested, as shown in Figure 12.

Table 4. PLL quantity and stoichiometric amounts of solid NHS-PEG-biotin and SPA-PEG.

Polymer PLL PEG-CH3 PEG-biotin Lys:PEG
(MW 20'000) (MW 2000) (MW 3400) expected PLL-g[2]-PEG-OCH3 27.2 mg; 1.36 mol 123 mg; 65.1 mol - 2 PLL-g[3.5]-PEG-OCH3 83.6 mg; 4.18 mol 215.7 mg; 114 mol- 3.5 PLL-g[5]-PEG-OCH3 106.8 mg; 5.34 193.2 mg; 102 mol- 5 mol PLL-g[3.5]-PEG-Biotin 39.5 mg; 1.98 mo1 92 mg; 48.7 mol 18.6 mg 5.4 mol 3.5 10%a PLL-g[3.5]-PEG-Biotin 39.5 mg; 1.98 mo1 91.9 mg; 48.7 mol 18.6 mg 5.4 mo13.5 10%b PLL-g[3.5]-PEG-Biotin 32.3 mg; 1.62 mol 2 mg; 22.2 mol 76 mg; 22.1 mo13.5 50%

Table 5. Integration values for m-PEG and PEG-biotin. The PLL unit to PEG chains ratio is estimated from the ratio of the integration value of the methyn proton (N-CHR-CO) of the lysine monomer unit to the sum of the proton-weighted integration values of the terminal methoxy protons (O-CH3) from the m-PEG chains and the methylene protons (-CH,-C(O)N-) from the biotin-PEG. The relative amounts of m-PEG and PEG-Biotin are estimated from the same respective integration.

Polymer Lys:PEG PEG-CH3 content [%] Biotin content [%]
PLL-g[2]-PEG-OCH3 2.06 100 % 0 PLL-g[ 3.5]-PEG-OCH3 3.43 100 % 0 PLL-g[5]-PEG-OCH3 5.48 100 % 0 PLL-g[3.5]-PEG-Biotin 3.9 87 - 90% 10 - 13%
10%a PLL-g[3.5]-PEG-Biotin 3.4 86 - 98% 11 - 14%
10%b PLL-g[3.5]-PEG-Biotin 50% 3.1 45 - 49% 51 - 55%

Claims (42)

CLAIMS:
1. A method for reducing non-specific adsorption comprising applying to or coating onto a substrate or surface of a biosensing or analytical device, a polyionic multifunctional copolymer in an aqueous solution, wherein the polyionic multifunctional copolymer comprises non-interactive polymer sidechains grafted onto a charged polyionic polymeric backbone which has an anionic charge at a pH greater than 4, wherein the substrate or surface has a charge that is opposite the charge of the polyionic polymeric backbone and comprises a material selected from the group consiting of metals, metal oxides, and charged polymers, wherein some or all of the non-interactive polymer sidechains are partially or fully functionalized at or near the free terminal position of the non-interactive polymer sidechains with a functional molecule, and wherein the polyionic multifunctional copolymer is applied to or coated onto a substrate or surface to reduce the non-specific adsorption of inorganic ions, peptides, proteins, or saccharides.
2. The method of claim 1 wherein the substrate or surface is in, is on, or forms: a device or material for use in cell culture or purification of materials from cell culture; a material for performing an assay; a device or prosthetic for implantation into a patient;
or an analytical or sensing device.
3. The method of claim 1 or 2 wherein the anionic backbone comprises a polymer selected from the group consisting of polymers comprising amino acids containing pendant charged groups imparting a negative charge to the backbone at pH
greater than 4, polysaccharides, and charged synthetic polymers with pendant negatively charged groups.
4. The method of claim 3 wherein the anionic backbone comprises one or more units selected from the group consisting of polyaspartic acid, polyglutamic acid, alginate, carrageenan, furcellaran, pectin, xanthan, hyaluronic acid, heparin, heparan sulfate, chondroitin sulfate, dermatan sulfate, dextran sulfate, poly(meth)acrylic acid, oxidized cellulose, carboxymethyl cellulose and crosmarmelose, maleic acid polymers, and fumaric acid polymers.
5. The method of any one of claims 1 to 4 wherein the non-interactive polymer sidechains are selected from the group consisting of poly(alkylene glycols), poly(alkylene oxides), neutral water-soluble polysaccharides, polyvinyl alcohol, poly-N-vinyl pyrrolidone, phosphoryl choline derivatives, non-cationic poly(meth)acrylates and combinations thereof.
6. The method of claim 1 wherein the non-interactive polymer sidechains are fully or partially modified at or near the terminal end of the polymers with reactive groups which are suitable for further functionalization.
7. The method of claim 6 wherein the reactive groups are selected from the group consisting of hydroxy (-OH), carboxy (-COOH), ester (-COOR), thiol (-SH), N-hydroxy-succinimidyl, maleimidyl, quinone, vinylsulfone groups, and combinations thereof.
8. The method of claim 1 wherein the functional molecule is selected from the group consisting of protein ligands, polynucleotides, carbohydrate or sugar ligands, simple organic molecules, and combinations thereof.
9. The method of claim 8 wherein the functional molecule is biotin.
10. The method of any one of claims 1 to 9 wherein the material has a positive charge and is selected from the group consisting of oxides that have an isoelectric point (IEP) above 5, and oxides or metals that have been treated to impose a positive charge onto the surface at the pH of greater than 4.
11. The method of claim 10 wherein the material is selected from the group consisting of tantalum, niobium, titanium, hafnium, silicon, iron and chromium oxides;
iron, chromium, steel, tantalum, niobium, titanium, hafnium, gold, and silver which either carry a positive charge at the pH used due to the presence of native oxide films or which have been treated to induce a positive charge in the material.
12. The method of claim 1 comprising exposing the copolymer to analyte which binds to the functional molecule in a fluid phase followed by absorption of the copolymer entity onto a substrate or surface.
13. The method of claim 1 comprising exposing the copolymer to analyte which binds to the functional molecule, wherein the copolymers are adsorbed onto a substrate or surface before or after binding the analyte.
14. A method for making a biosensing or analytical device comprising applying to or coating onto a substrate or surface of the biosensing or analytical device polyionic multi-functional copolymers in an aqueous solution, wherein the polyionic multi-functional copolymers are brush copolymers comprising non-intractive side chains grafted onto a charged polyionic polymeric backbone, wherein some or all of the non-interactive polymer side chains are partially or fully functionalized at or near the free terminal position of the non-interactive polymer side chains with a functional molecule, wherein the device has a charge opposite to the charge of the copolymer; and wherein some of the non-interactive polymer side chains are functionalized with a functional moiety and some of the non-interactive polymer side chains are not functionalized.
15. The method of claim 14, wherein the substrate or surface comprises a material selected from the group consisting of glass, metals, metal oxides, and charged polymers.
16. The method of claim 14 wherein the functionalized and non-functionalized copolymers are applied sequentially.
17. The method of claim 16 wherein the functionalized copolymers are adsorbed to the substrate or surface and exposed to analyte.
18. The method of claim 14 wherein the functionalized copolymer is attached to the substrate or surface by physisorption or chemisorption.
19. The method of claim 14 wherein the functionalized copolymer is attached to the substrate or surface by covalent chemical binding.
20. The method of claim 14 wherein the surface is patterned with areas (a) with polyionic copolymers which are at least one of adsorbed non-modified and non-functionalized, and areas (b) with polyionic copolymers which are at least one of adsorbed modified and functionalized.
21. The method of claim 20 wherein the patterns have dimensions parallel to the surface in the millimeter, micrometer or submicrometer range.
22. The method of claim 20 wherein the patterning is done by fluidic, microfluidic, stamping or microcontact printing.
23. The method of claim 20 wherein the polyionic copolymer is applied to the substrate or surface, then functional molecule is bound to the copolymer in a defined surface pattern by fluidic, microfluidic, stamping, microcontact printing or ink-jet techniques.
24. A biosensing or analytical device comprising a coating on a substrate or surface, wherein the coating comprises a polyionic multi-functional copolymer, wherein the polyionic multi-functional copolymer is a brush copolymer comprising non-interactive polymer side chains grafted onto a charged polyionic polymeric backbone, wherein some or all of the non-interactive polymer side chains are partially or fully functionalized at or near the free terminal position of the non-interactive polymer side chains with a functional molecule, wherein the substrate or surface has a charge that is opposite the charge of the copolymer.
25. A device as claimed in claim 24 wherein some of the non-interactive polymer side chains are functionalized with a functional moiety and some of the non-interactive polymer side chains are not functionalized.
26. A device as claimed in claim 24 or 25 wherein the substrate or surface comprises a material selected from the group consisting of glass, metals, metal oxides, and charged polymers.
27. A device as claimed in any one of claims 24 to 26 wherein the functionalized copolymer is attached to the substrate or surface by physisorption or chemisorption.
28. A device as claimed in any one of claims 24 to 27 wherein the functionalized copolymer is attached to the substrate or surface by covalent chemical binding
29. A device as claimed in any one of claims 24 to 28 wherein the surface is patterned with areas (a) with polyionic copolymers which are at least one of adsorbed non-modified and non-functionalized, and areas (b) with polyionic copolymers which are at least one of adsorbed modified and functionalized.
30. A device as claimed in claim 29 wherein the patterns have dimensions parallel to the surface in the millimeter, micrometer or submicrometer range.
31. The use of the device of claim 24 for qualitatively or quantitatively measuring or detecting analyte.
32. The use of claim 31 wherein the analyte is selected from the group consisting of polynucleotides, peptides, proteins, carbohydrates, sugars, and glycoproteins.
33. The use of claim 31 wherein the amount of analyte immobilized at the polyionic copolymer coated substrate or surface is determined by measuring or using electromagnetic radiation or using a method selected from the group consisting of absorption, scattering, emission, thermal phenomena or combinations thereof.
34. The use of claim 33 wherein the amount of analyte is determined by using waveguides.
35. The use of claim 34 wherein the waveguides comprise one or more optical coupling elements for incoupling excitation light to the waveguides.
36. The use of claim 35 wherein the optical coupling elements are selected from the group consisting of prism couplers, evanescent couplers comprising joined optical waveguides with overlapping evanescent fields, end face couplers with focusing lens and gating couplers and combinations thereof.
37. The use of claim 35 wherein the optical coupling elements are diffractive gratings selected from the group consisting of relief gratings with a profile selected from the group consisting of rectangular, triangular or sinusoidal profile, and phase gratings or volume gratings with a periodical modulation of the refractive index in the waveguide and combinations thereof.
38. The use of claim 37 wherein one or more of the optical coupling elements are diffractive relief gratings with a rectangular profile.
39. The use of claim 33 wherein analyte is detected using a continuous type of process.
40. The use of claim 33 wherein the functionalized polyionic copolymer is applied in a pattern, and detection is localized to specific areas of the pattern.
41. The use of claim 40 wherein the pattern includes regions of different functional molecules for detection of multiple analytes.
42. The use of the device of claim 24 to modify cell to surface interactions on a metal or metal oxide surface.
CA002371011A 1999-04-28 2000-04-28 Polyionic coatings in analytic and sensor devices Expired - Lifetime CA2371011C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US13140299P 1999-04-28 1999-04-28
US13139199P 1999-04-28 1999-04-28
US60/131,391 1999-04-28
US60/131,402 1999-04-28
US18461600P 2000-02-24 2000-02-24
US60/184,616 2000-02-24
PCT/US2000/011708 WO2000065352A1 (en) 1999-04-28 2000-04-28 Polyionic coatings in analytic and sensor devices

Publications (2)

Publication Number Publication Date
CA2371011A1 CA2371011A1 (en) 2000-11-02
CA2371011C true CA2371011C (en) 2009-12-22

Family

ID=27384152

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002371011A Expired - Lifetime CA2371011C (en) 1999-04-28 2000-04-28 Polyionic coatings in analytic and sensor devices

Country Status (8)

Country Link
US (1) US6884628B2 (en)
EP (1) EP1190252B1 (en)
JP (1) JP4647792B2 (en)
AT (1) ATE419528T1 (en)
AU (1) AU769571B2 (en)
CA (1) CA2371011C (en)
DE (1) DE60041255D1 (en)
WO (1) WO2000065352A1 (en)

Families Citing this family (241)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6048734A (en) 1995-09-15 2000-04-11 The Regents Of The University Of Michigan Thermal microvalves in a fluid flow method
AU5785400A (en) 1999-07-02 2001-01-22 Symyx Technologies, Inc. Polymer brushes for immobilizing molecules to a surface or substrate, where the polymers have water-soluble or water-dispersible segments and probes bonded thereto
US20030059809A1 (en) * 1999-12-17 2003-03-27 Todor Vujasinovic Biochips,preparation and uses
US7163712B2 (en) * 2000-03-03 2007-01-16 Duke University Microstamping activated polymer surfaces
US6406777B1 (en) * 2000-06-14 2002-06-18 The United States Of America As Represented By The Secretary Of The Navy Metal and glass structure for use in surface enhanced Raman spectroscopy and method for fabricating same
DE10036907B4 (en) * 2000-07-28 2012-03-22 Xantec Bioanalytics Gmbh Process for the preparation of a coating on a gold vapor-deposited glass substrate, coating produced by this process and their use
US7678539B2 (en) * 2000-08-10 2010-03-16 Corning Incorporated Arrays of biological membranes and methods and use thereof
JP4887530B2 (en) * 2000-08-21 2012-02-29 独立行政法人産業技術総合研究所 Magnetic fine particles and method for producing the same
US7205161B2 (en) 2001-01-10 2007-04-17 Symyx Technologies, Inc. Polymer brushes for immobilizing molecules to a surface or substrate having improved stability
US6692700B2 (en) 2001-02-14 2004-02-17 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
ES2441412T3 (en) 2001-03-09 2014-02-04 Trovagene, Inc. Conjugated probes and optical analyte detection
US8895311B1 (en) 2001-03-28 2014-11-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US7829025B2 (en) 2001-03-28 2010-11-09 Venture Lending & Leasing Iv, Inc. Systems and methods for thermal actuation of microfluidic devices
US7323140B2 (en) 2001-03-28 2008-01-29 Handylab, Inc. Moving microdroplets in a microfluidic device
US7010391B2 (en) 2001-03-28 2006-03-07 Handylab, Inc. Methods and systems for control of microfluidic devices
US6852287B2 (en) 2001-09-12 2005-02-08 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
US6844028B2 (en) * 2001-06-26 2005-01-18 Accelr8 Technology Corporation Functional surface coating
US7501157B2 (en) * 2001-06-26 2009-03-10 Accelr8 Technology Corporation Hydroxyl functional surface coating
US6989267B2 (en) 2001-07-02 2006-01-24 Agilent Technologies, Inc. Methods of making microarrays with substrate surfaces having covalently bound polyelectrolyte films
JP2005512021A (en) * 2001-07-09 2005-04-28 アリゾナ ボード オブ リージェンツ Affinity biosensor for monitoring biological processes
WO2003021253A2 (en) * 2001-08-27 2003-03-13 Zeptosens Ag Bioanalytical recognition surface with optimised recognition element density
EP1421217A2 (en) * 2001-08-27 2004-05-26 Zeptosens AG Surface for the immobilisation of nucleic acids
US9671396B2 (en) * 2001-09-05 2017-06-06 Joon Won Park Solid substrate comprising array of dendrons and methods for using the same
EP1425583A1 (en) * 2001-09-12 2004-06-09 Eidgenössische Technische Hochschule Zürich Device with chemical surface patterns
CN1639568A (en) * 2001-09-21 2005-07-13 宝生物工程株式会社 Support for ligand immobilization
FI118061B (en) * 2001-09-24 2007-06-15 Beanor Oy Procedure and bio donor for analysis
US8043868B2 (en) 2001-12-21 2011-10-25 Imec Method and apparatus for detecting an analyte
EP1321761B1 (en) * 2001-12-21 2006-08-23 Interuniversitair Micro-Elektronica Centrum Method of detecting an analyte
AU2002366875B2 (en) * 2001-12-21 2008-08-21 Sense Proteomic Limited Probe for mass spectrometry
FI115166B (en) * 2001-12-31 2005-03-15 Biofons Oy Diagnostic procedures
US20050260259A1 (en) * 2004-04-23 2005-11-24 Bolotin Elijah M Compositions for treatment with glucagon-like peptide, and methods of making and using the same
DE60335608D1 (en) * 2002-02-27 2011-02-17 Pharmain Corp COMPOSITIONS FOR THE DELIVERY OF THERAPEUTICS AND OTHER MATERIALS AND METHOD FOR THE PRODUCTION AND USE THEREOF
US7635463B2 (en) * 2002-02-27 2009-12-22 Pharmain Corporation Compositions for delivery of therapeutics and other materials
AU2002233097A1 (en) * 2002-02-28 2003-09-09 Eidgenossische Technische Hochschule Zurich Oberflachentechnik Biomedical implant surfaces with osteogenic properties
EP1495326A1 (en) * 2002-04-12 2005-01-12 Micronas GmbH Method for immobilizing molecules on surfaces
CA2482842C (en) * 2002-04-16 2009-10-27 Eidgenossisch Technische Hochschule Zurich Environmentally compatible additives for aqueous lubricants
US20030198967A1 (en) * 2002-04-23 2003-10-23 Matson Robert S. Multi-functional microarrays and methods
US7010182B2 (en) * 2002-07-31 2006-03-07 Luna Innovations Incorporated Biosensors having enhanced environmental sensitivity
US7998699B2 (en) 2002-08-15 2011-08-16 University Of South Florida Early detection of pathogens in blood
US7384742B2 (en) * 2002-08-16 2008-06-10 Decision Biomarkers, Inc. Substrates for isolating reacting and microscopically analyzing materials
US20090118605A1 (en) * 2002-08-30 2009-05-07 Northwestern University Surface-enhanced raman nanobiosensor
US20040043508A1 (en) * 2002-09-03 2004-03-04 Frutos Anthony G. Polymer-coated substrates for binding biological molecules
US20040062882A1 (en) * 2002-09-30 2004-04-01 Andrea Liebmann-Vinson Cell adhesion resisting surfaces
ES2208121B1 (en) * 2002-11-29 2005-08-16 Consejo Sup. Investig. Cientificas ANTIBODIES AND IMMOBILIZED ANTIGENS ON SILICON MAGNETIC PARTICLES AS BIOSENSORS.
US7070922B2 (en) * 2002-12-04 2006-07-04 International Business Machines Corporation Surface treatment
US20040121339A1 (en) * 2002-12-19 2004-06-24 Jizhong Zhou Special film-coated substrate for bio-microarray fabrication and use thereof
CN100347545C (en) * 2003-04-08 2007-11-07 成都夸常科技有限公司 Method of preoceeding qualitative and/or quantitative analysis against target substance in sample and its detecting device
JP4006523B2 (en) * 2003-04-10 2007-11-14 独立行政法人産業技術総合研究所 Protein array and production method thereof
ATE412902T1 (en) * 2003-04-14 2008-11-15 Caliper Life Sciences Inc REDUCING MIGRATION SHIFT ASSAY INTERFERENCE
DE10320312A1 (en) * 2003-05-06 2004-12-02 Friz Biochem Gmbh Substrate as a carrier for ligates
US20040228766A1 (en) * 2003-05-14 2004-11-18 Witty Thomas R. Point of care diagnostic platform
US7160867B2 (en) * 2003-05-16 2007-01-09 Isotechnika, Inc. Rapamycin carbohydrate derivatives
EP2402089A1 (en) 2003-07-31 2012-01-04 Handylab, Inc. Processing particle-containing samples
US9206414B2 (en) * 2003-08-13 2015-12-08 Axcelon Biopolymers Corporation Anisotropic nanocomposite hydrogel
PT2103694E (en) 2003-08-15 2011-07-26 Univ South Florida Materials and methods for capture of pathogens and removal of aurintricarboxylic acid from a sample
US20050070027A1 (en) * 2003-09-30 2005-03-31 Jacques Gollier Double resonance interrogation of grating-coupled waveguides
JP2006514299A (en) * 2003-10-27 2006-04-27 センス プロテオミック リミテッド Enzyme arrays and assays
US20050244898A1 (en) * 2003-11-19 2005-11-03 Cohen Robert E Large-area two-dimensional non-adhesive cell arrays for sensing and cell-sorting applications
US7510942B2 (en) * 2003-11-25 2009-03-31 Arizona Board Of Regents, Acting For And On Behalf Of Arizona State University Molecular modifications of metal/dielectric interfaces
US20050131513A1 (en) * 2003-12-16 2005-06-16 Cook Incorporated Stent catheter with a permanently affixed conductor
US20050180957A1 (en) * 2004-01-16 2005-08-18 Scharp David W. Method of using fibrin-bound angiogenic factors to stimulate vascularization of transplant site of encapsulated cells
US7723126B2 (en) * 2004-03-24 2010-05-25 Wisconsin Alumni Research Foundation Plasma-enhanced functionalization of inorganic oxide surfaces
JP4535490B2 (en) * 2004-04-09 2010-09-01 株式会社資生堂 Protein adsorption prevention method
WO2005108620A2 (en) 2004-05-03 2005-11-17 Handylab, Inc. Processing polynucleotide-containing samples
US8852862B2 (en) 2004-05-03 2014-10-07 Handylab, Inc. Method for processing polynucleotide-containing samples
JP2007538233A (en) * 2004-05-21 2007-12-27 エムディーエス インコーポレイテッド ドゥーイング ビジネス スルー イッツ エムディーエス ファーマ サービシーズ ディビジョン Method for quantifying cell binding properties of medical devices
US20060051396A1 (en) * 2004-06-16 2006-03-09 Affinergy, Inc. Biofunctional coatings
US10150945B2 (en) * 2004-07-23 2018-12-11 Jnc Corporation Cell culture device and manufacturing method therof
KR20070061808A (en) * 2004-08-04 2007-06-14 악셀라 바이오센서스 인코포레이티드 Patterned surfaces with chemical crosslinkers for use in diffraction-based sensing
US7595157B2 (en) * 2004-08-19 2009-09-29 Biocept, Inc. Microarrays utilizing hydrogels
US20110177955A1 (en) * 2004-10-12 2011-07-21 Luis Alberto Burzio Multiplexed protein adsorption assay
US20060110594A1 (en) * 2004-11-24 2006-05-25 Frutos Anthony G Polymer-coated substrates for binding biomolecules and methods of making and using thereof
WO2006070841A1 (en) 2004-12-28 2006-07-06 Japan Science And Technology Agency Method for immobilizing self-organizing material or fine particle on substrate, and substrate manufactured by using such method
US7604984B2 (en) * 2004-12-29 2009-10-20 Corning Incorporated Spatially scanned optical reader system and method for using same
US20060141527A1 (en) * 2004-12-29 2006-06-29 Caracci Stephen J Method for creating a reference region and a sample region on a biosensor and the resulting biosensor
US7629173B2 (en) * 2004-12-29 2009-12-08 Corning Incorporated Optical reader system and method for monitoring and correcting lateral and angular misalignments of label independent biosensors
JP2006335912A (en) * 2005-06-03 2006-12-14 Fujifilm Holdings Corp Immobilizing agent for physiologically active substance
DE602006009980D1 (en) * 2005-02-23 2009-12-10 Fujifilm Corp biosensor
JP2006271860A (en) * 2005-03-30 2006-10-12 Japan Lifeline Co Ltd Medical device and its manufacturing method
US8465771B2 (en) 2005-03-30 2013-06-18 The University Of Western Ontario Anisotropic hydrogels
WO2006113785A2 (en) * 2005-04-18 2006-10-26 Brigham Young University Laser modification and functionalization of substrates
CN101208599B (en) 2005-04-26 2014-12-10 拜耳知识产权有限责任公司 Novel equipment and method for coating substrates for analyte detection by way of an affinity assay method
US7723438B2 (en) * 2005-04-28 2010-05-25 International Business Machines Corporation Surface-decorated polymeric amphiphile porogens for the templation of nanoporous materials
US20070009568A1 (en) * 2005-06-03 2007-01-11 Bhatnagar Rajendra S Methods for immobilizing molecules on surfaces
US8475886B2 (en) 2005-08-05 2013-07-02 Corning Incorporated Methods for producing surfaces that resist non-specific protein binding and cell attachment
WO2007035527A2 (en) * 2005-09-15 2007-03-29 Duke University Non-fouling polymeric surface modification and signal amplification method for biomolecular detection
US7763423B2 (en) * 2005-09-30 2010-07-27 Pacific Biosciences Of California, Inc. Substrates having low density reactive groups for monitoring enzyme activity
US20090148346A1 (en) * 2005-10-07 2009-06-11 Canon Kabushiki Kaisha Substrate with binding functional group
JP4818056B2 (en) * 2005-10-07 2011-11-16 キヤノン株式会社 Structure having a binding functional group on a substrate for binding a capture molecule for capturing a target substance
JP4959710B2 (en) * 2005-11-01 2012-06-27 レンセレアー ポリテクニック インスティテュート 3D cell array chip and platform for toxicology assays
US20070117152A1 (en) * 2005-11-08 2007-05-24 Fujifilm Corporation Biosensor
AU2006313586A1 (en) * 2005-11-08 2007-05-18 Smart Holograms Limited Novel boronate complex and its use in a glucose sensor
US20070224164A1 (en) * 2005-11-14 2007-09-27 Maciej Radosz Degradable nanogel for drug delivery
US20070110786A1 (en) * 2005-11-15 2007-05-17 Boston Scientific Scimed, Inc. Medical articles having enhanced therapeutic agent binding
US20090298197A1 (en) * 2005-11-15 2009-12-03 Oxonica Materials Inc. Sers-based methods for detection of bioagents
KR100738083B1 (en) * 2005-12-20 2007-07-12 삼성전자주식회사 A substrate used in a microarray and a method for preparing the same
US8455088B2 (en) * 2005-12-23 2013-06-04 Boston Scientific Scimed, Inc. Spun nanofiber, medical devices, and methods
US7723422B2 (en) * 2005-12-23 2010-05-25 Boston Scientific Scimed, Inc. Functionalized block copolymers
US7674864B2 (en) * 2005-12-23 2010-03-09 Boston Scientific Scimed, Inc. Polymeric hybrid precursors, polymeric hybrid precursor composite matrices, medical devices, and methods
US7781203B2 (en) * 2005-12-29 2010-08-24 Corning Incorporated Supports for assaying analytes and methods of making and using thereof
US8834912B2 (en) * 2005-12-30 2014-09-16 Boston Scientific Scimed, Inc. Medical devices having multiple charged layers
EP1808187A1 (en) * 2006-01-11 2007-07-18 Straumann Holding AG Cell selective implant surface with controlled release of bioactive agents
US20090155811A1 (en) * 2006-01-27 2009-06-18 Oxonica, Inc. Lateral Flow Immunoassay With Encapsulated Detection Modality
WO2007100882A2 (en) * 2006-02-28 2007-09-07 Tyco Healthcare Group Lp Tissue adhesives and sealants and methods for their use
US8349349B2 (en) * 2006-02-28 2013-01-08 Covidien Lp Tissue adhesives and sealants and method for their use
US8835513B2 (en) 2006-02-28 2014-09-16 Covidien Lp Drug delivery devices
EP1997877B1 (en) * 2006-03-17 2014-03-05 Sanyo Chemical Industries, Ltd. Cell culture substrate
EP2001529A4 (en) * 2006-03-21 2011-11-09 Agency Science Tech & Res Cell-adhesive polyelectrolyte material for use as membrane and coating
EP2001990B1 (en) 2006-03-24 2016-06-29 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using same
US7998708B2 (en) 2006-03-24 2011-08-16 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US10900066B2 (en) 2006-03-24 2021-01-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US8545865B2 (en) * 2006-03-24 2013-10-01 Boston Scientific Scimed, Inc. Medical devices having polymer brushes
US8088616B2 (en) 2006-03-24 2012-01-03 Handylab, Inc. Heater unit for microfluidic diagnostic system
US11806718B2 (en) 2006-03-24 2023-11-07 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US8975216B2 (en) 2006-03-30 2015-03-10 Pacific Biosciences Of California Articles having localized molecules disposed thereon and methods of producing same
US20080076189A1 (en) * 2006-03-30 2008-03-27 Visigen Biotechnologies, Inc. Modified surfaces for the detection of biomolecules at the single molecule level
US20080050747A1 (en) * 2006-03-30 2008-02-28 Pacific Biosciences Of California, Inc. Articles having localized molecules disposed thereon and methods of producing and using same
WO2007118714A1 (en) * 2006-04-19 2007-10-25 Universiteit Gent Integrated surface mode biosensor
US8330951B2 (en) * 2006-04-28 2012-12-11 Hewlett-Packard Development Company, L.P. Nano-enhanced Raman spectroscopy substrate packaging structure
US10741034B2 (en) 2006-05-19 2020-08-11 Apdn (B.V.I.) Inc. Security system and method of marking an inventory item and/or person in the vicinity
US9790538B2 (en) 2013-03-07 2017-10-17 Apdn (B.V.I.) Inc. Alkaline activation for immobilization of DNA taggants
WO2008014223A2 (en) * 2006-07-24 2008-01-31 Becton, Dickinson And Company Assay particle concentration and imaging apparatus and method
EP2069412B1 (en) * 2006-08-07 2019-01-02 University of Washington Mixed charge copolymers and hydrogels
JPWO2008018355A1 (en) * 2006-08-08 2009-12-24 シャープ株式会社 Biosensor, manufacturing method thereof, and detection method using the biosensor
JP5001015B2 (en) * 2006-09-28 2012-08-15 富士フイルム株式会社 Biochemical instrument having a surface with reduced non-specific adsorption
WO2008060604A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
WO2008061165A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic cartridge and method of making same
AU2007323594A1 (en) * 2006-11-21 2008-05-29 Applied Biosystems, Llc Intermediates and methods for forming passivated surfaces on oxide layers and articles produced thereby
US8029902B2 (en) * 2006-12-11 2011-10-04 Wisconsin Alumni Research Foundation Plasma-enhanced functionalization of substrate surfaces with quaternary ammonium and quaternary phosphonium groups
EP2104846A2 (en) * 2006-12-29 2009-09-30 The University of Washington Dual-functional nonfouling surfaces and materials
JP2008185494A (en) * 2007-01-31 2008-08-14 Fujifilm Corp Physiologically active substance-immobilized substrate
EP2076774A1 (en) * 2007-02-28 2009-07-08 Corning Incorporated Surfaces and methods for biosensor cellular assays
JP2008249502A (en) * 2007-03-30 2008-10-16 Fujifilm Corp Solid substrate immobilized with physiologically active substance
US8093039B2 (en) * 2007-04-10 2012-01-10 The Trustees Of The Stevens Institute Of Technology Surfaces differentially adhesive to eukaryotic cells and non-eukaryotic cells
US8084077B2 (en) 2007-05-25 2011-12-27 Abbott Laboratories One-step phosphorylcholine-linked polymer coating and drug loading of stent
US9151751B2 (en) * 2007-06-29 2015-10-06 Applied Biosystems, Llc Systems and methods for electronic detection with nanoFETS
US8105783B2 (en) 2007-07-13 2012-01-31 Handylab, Inc. Microfluidic cartridge
US8133671B2 (en) 2007-07-13 2012-03-13 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US9186677B2 (en) 2007-07-13 2015-11-17 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US20090136385A1 (en) 2007-07-13 2009-05-28 Handylab, Inc. Reagent Tube
US8182763B2 (en) 2007-07-13 2012-05-22 Handylab, Inc. Rack for sample tubes and reagent holders
US8287820B2 (en) 2007-07-13 2012-10-16 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US9618139B2 (en) 2007-07-13 2017-04-11 Handylab, Inc. Integrated heater and magnetic separator
EP3741869A1 (en) 2007-07-13 2020-11-25 Handylab, Inc. Polynucleotide capture materials and methods of using same
USD621060S1 (en) 2008-07-14 2010-08-03 Handylab, Inc. Microfluidic cartridge
US7960336B2 (en) * 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
JP2009057549A (en) * 2007-08-06 2009-03-19 Canon Inc Structure, and method of producing the same
US8563527B2 (en) * 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
KR101050690B1 (en) 2007-09-07 2011-07-20 광주과학기술원 Multifunctional polymer membrane and its use
WO2009031804A2 (en) * 2007-09-07 2009-03-12 Gwangju Institute Of Science And Technology Multi-functional polymeric layers and its uses
US8445217B2 (en) 2007-09-20 2013-05-21 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
DE102007049013A1 (en) * 2007-10-11 2009-04-16 Endress + Hauser Conducta Gesellschaft für Mess- und Regeltechnik mbH + Co. KG Sensor with long-term stability for organic processes
US20090130746A1 (en) * 2007-10-25 2009-05-21 Canon U.S. Life Sciences, Inc. Microchannel surface coating
WO2009057120A2 (en) * 2007-10-31 2009-05-07 Given Imaging Ltd. Device, system and method for in-vivo analysis
JP5345335B2 (en) * 2007-12-03 2013-11-20 国立大学法人九州大学 Novel zwitterionic multibranched resin and protein chip surface modifier
EP2584360A1 (en) 2007-12-05 2013-04-24 Zyomyx Inc. Cell assay kit and method
US8796184B2 (en) 2008-03-28 2014-08-05 Sentilus, Inc. Detection assay devices and methods of making and using the same
US8815611B2 (en) * 2008-04-10 2014-08-26 Corning Incorporated Surface for label independent detection and method thereof
US8877298B2 (en) * 2008-05-27 2014-11-04 The Hong Kong University Of Science And Technology Printing using a structure coated with ultraviolet radiation responsive material
JP2009286968A (en) * 2008-05-30 2009-12-10 Canon Inc Graft polymer-containing substrate, its production method and magnetic biosensor
US20090311540A1 (en) * 2008-06-11 2009-12-17 Yoram Cohen Highly Sensitive and Selective Nano-Structured Grafted Polymer Layers
EP2339951A1 (en) * 2008-07-10 2011-07-06 Given Imaging Ltd. Device, method and kit for in vivo detection of a biomarker
USD618820S1 (en) 2008-07-11 2010-06-29 Handylab, Inc. Reagent holder
WO2010005444A1 (en) * 2008-07-11 2010-01-14 Handylab, Inc. Polynucleotide capture materials, and methods of using same
USD787087S1 (en) 2008-07-14 2017-05-16 Handylab, Inc. Housing
US8173198B2 (en) * 2008-07-23 2012-05-08 Life Technologies Corporation Deposition of metal oxides onto surfaces as an immobilization vehicle for carboxylated or phophated particles or polymers
US20100034704A1 (en) * 2008-08-06 2010-02-11 Honeywell International Inc. Microfluidic cartridge channel with reduced bubble formation
US8226603B2 (en) 2008-09-25 2012-07-24 Abbott Cardiovascular Systems Inc. Expandable member having a covering formed of a fibrous matrix for intraluminal drug delivery
US8076529B2 (en) 2008-09-26 2011-12-13 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix for intraluminal drug delivery
US8500687B2 (en) 2008-09-25 2013-08-06 Abbott Cardiovascular Systems Inc. Stent delivery system having a fibrous matrix covering with improved stent retention
US8049061B2 (en) 2008-09-25 2011-11-01 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix having hydrogel polymer for intraluminal drug delivery
US20100099203A1 (en) * 2008-10-03 2010-04-22 Molecular Sensing, Inc. Substrates with surfaces modified with PEG
DE102008053270A1 (en) 2008-10-27 2010-05-12 Medizinische Hochschule Hannover Apparatus and method for analyzing cells
US8580240B1 (en) 2008-11-19 2013-11-12 University Of Kentucky Research Foundation Compounds and methods for reducing the occurrence of post-surgical adhesions
AU2009324879A1 (en) * 2008-11-25 2010-06-17 The Regents Of The University Of California Functionalized titanium implants and related regenerative materials
EP2192410B1 (en) * 2008-11-26 2012-07-11 Corning Incorporated Nanoparticulate affininty capture for label independent detections system
EP2204650B1 (en) * 2008-12-31 2012-06-27 DWI an der RWTH Aachen e.V. Novel biosensor system based on recognition induced birefringence (RIB)
US20100184056A1 (en) * 2009-01-12 2010-07-22 Molecular Sensing, Inc. Sample collection and measurement in a single container by back scattering interferometry
DE202010017793U1 (en) * 2009-03-20 2012-08-31 Attana Ab Analytical method and sensor
WO2010117460A1 (en) * 2009-04-08 2010-10-14 Michigan Molecular Institute Surface modification of reverse osmosis membranes by hydrophilic dendritic polymers
WO2010123448A1 (en) * 2009-04-21 2010-10-28 Gyros Patent Ab Method for reducing adsorption losses on metal oxide surfaces
US20120208291A1 (en) * 2009-05-01 2012-08-16 Univeristy Of Utah Research Foundation Methods and compositions for measuring high affinity interactions with kinetic imaging of single molecule interaction (kismi)
US8501406B1 (en) 2009-07-14 2013-08-06 Pacific Biosciences Of California, Inc. Selectively functionalized arrays
WO2011028358A2 (en) * 2009-09-02 2011-03-10 Mayo Foundation For Medical Education And Research System and method for detecting healing adjacent to implanted medical device
US20110225809A1 (en) * 2010-03-17 2011-09-22 Alan Francis Daher Apparatus for removably attaching an item to a surface
EP3305192B1 (en) 2010-05-17 2019-10-09 Sentilus Holdco, LLC Detection devices and related methods of use
US9638632B2 (en) 2010-06-11 2017-05-02 Vanderbilt University Multiplexed interferometric detection system and method
EP2409766A1 (en) * 2010-07-23 2012-01-25 F. Hoffmann-La Roche AG Method for hydrophilising surfaces of fluid components and objects containing such components
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
CA2833262C (en) 2011-04-15 2020-08-18 Becton, Dickinson And Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
EP2541250A1 (en) * 2011-06-30 2013-01-02 Koninklijke Philips Electronics N.V. Molecular architecture on magnetic particles for affinity assays with low non-specific binding
WO2013009308A1 (en) * 2011-07-13 2013-01-17 Empire Technology Development Llc Method and system for analyte monitoring using surface plasmons with a refreshable surface
US20140127517A1 (en) * 2011-07-15 2014-05-08 Jason J. Locklin Compounds, methods of making, and methods of use
US9599613B2 (en) 2011-07-20 2017-03-21 University Of Washington Through Its Center For Commercialization Photonic blood typing
USD692162S1 (en) 2011-09-30 2013-10-22 Becton, Dickinson And Company Single piece reagent holder
KR102121853B1 (en) 2011-09-30 2020-06-12 벡톤 디킨슨 앤드 컴퍼니 Unitized reagent strip
WO2013065016A1 (en) 2011-11-02 2013-05-10 University Of Cape Town A method of detecting and/or quantifying an analyte in a biological sample
CN104040238B (en) 2011-11-04 2017-06-27 汉迪拉布公司 Polynucleotides sample preparation apparatus
US10031138B2 (en) * 2012-01-20 2018-07-24 University Of Washington Through Its Center For Commercialization Hierarchical films having ultra low fouling and high recognition element loading properties
CA2863637C (en) 2012-02-03 2021-10-26 Becton, Dickinson And Company External files for distribution of molecular diagnostic tests and determination of compatibility between tests
US9161756B2 (en) 2012-03-16 2015-10-20 Covidien Lp Closure tape dispenser
US9572580B2 (en) 2012-03-16 2017-02-21 Covidien Lp Closure tape dispenser
JP5844195B2 (en) * 2012-03-29 2016-01-13 三洋化成工業株式会社 Magnetic silica particles, immunoassay using the same, and reagent for immunoassay
US20140102899A1 (en) * 2012-04-09 2014-04-17 Applied Dna Sciences, Inc. Plasma treatment for dna binding
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
EP2875078B1 (en) * 2012-07-18 2017-12-20 President and Fellows of Harvard College Modification of surfaces for simulataneous repellency and targeted binding of desired moieties
US9758606B2 (en) 2012-07-31 2017-09-12 The Trustees Of Columbia University In The City Of New York Cyclopropenium polymers and methods for making the same
WO2014022365A1 (en) * 2012-07-31 2014-02-06 The Trustees Of Columbia University In The City Of New York Cyclopropenium polymers and methods for making the same
US9297032B2 (en) 2012-10-10 2016-03-29 Apdn (B.V.I.) Inc. Use of perturbants to facilitate incorporation and recovery of taggants from polymerized coatings
US9266370B2 (en) 2012-10-10 2016-02-23 Apdn (B.V.I) Inc. DNA marking of previously undistinguished items for traceability
US8994077B2 (en) * 2012-12-21 2015-03-31 International Business Machines Corporation Field effect transistor-based bio sensor
TWI535750B (en) 2013-01-14 2016-06-01 國立臺灣大學 Polymer composition on substrate and surface modification method
US10060851B2 (en) * 2013-03-05 2018-08-28 Plexense, Inc. Surface plasmon detection apparatuses and methods
US9963740B2 (en) 2013-03-07 2018-05-08 APDN (B.V.I.), Inc. Method and device for marking articles
US10359362B2 (en) 2013-04-15 2019-07-23 Plexense, Inc. Method for manufacturing nanoparticle array, surface plasmon resonance-based sensor and method for analyzing using same
US10544449B2 (en) 2013-06-14 2020-01-28 Pacific Biosciences Of California, Inc. Bis-biotinylation tags
DE102013211850A1 (en) * 2013-06-21 2014-12-24 Gilupi Gmbh Rapid test for detection of pathogen material, in particular to support the diagnosis of sepsis, and kit and device for performing a sepsis test
DE102013211837A1 (en) * 2013-06-21 2014-12-24 Gilupi Gmbh Catheter with detection device for the real-time detection of a sample material
US9366651B2 (en) 2013-07-03 2016-06-14 Matrix Sensors, Inc. Array of sensors with surface modifications
EP3058339B1 (en) 2013-10-07 2019-05-22 APDN (B.V.I.) Inc. Multimode image and spectral reader
US10745825B2 (en) 2014-03-18 2020-08-18 Apdn (B.V.I.) Inc. Encrypted optical markers for security applications
CN106103121B (en) 2014-03-18 2019-12-06 亚普蒂恩(B.V.I.)公司 Encrypted optical marker for security applications
CN106687805B (en) 2014-07-02 2020-05-29 生命技术公司 Surface treatment of semiconductor sensors
US11754563B2 (en) 2014-11-20 2023-09-12 Global Life Sciences Solutions Operations UK Ltd Porous membranes with a polymer grafting, methods and uses thereof
US10760182B2 (en) 2014-12-16 2020-09-01 Apdn (B.V.I.) Inc. Method and device for marking fibrous materials
JP2018506715A (en) 2015-01-23 2018-03-08 ヴァンダービルト ユニバーシティー Robust interferometer and method of use
TWI585401B (en) * 2015-12-17 2017-06-01 長庚大學 Light addressable potentiometric sensing unit
US10627396B2 (en) 2016-01-29 2020-04-21 Vanderbilt University Free-solution response function interferometry
EP3442719B1 (en) 2016-04-11 2021-09-01 APDN (B.V.I.) Inc. Method of marking cellulosic products
US10995371B2 (en) 2016-10-13 2021-05-04 Apdn (B.V.I.) Inc. Composition and method of DNA marking elastomeric material
US9999899B2 (en) 2016-11-01 2018-06-19 International Business Machines Corporation Controlled exposure of in-vivo sensors
US10920274B2 (en) 2017-02-21 2021-02-16 Apdn (B.V.I.) Inc. Nucleic acid coated submicron particles for authentication
WO2018222034A1 (en) * 2017-05-30 2018-12-06 Jurriaan Huskens Surface with poly-cationic polymers and functionalized using coupling chemistry, biosensor comprising the same and use thereof in bio-sensing
WO2018234564A1 (en) * 2017-06-22 2018-12-27 Paris Sciences Et Lettres - Quartier Latin Redox cleavable comb-like copolymer for controlled adhesion between cells and substrates
JP7358334B2 (en) 2018-03-28 2023-10-10 持田製薬株式会社 Novel reactive alginate derivative
US11357542B2 (en) 2019-06-21 2022-06-14 Covidien Lp Valve assembly and retainer for surgical access assembly
WO2021060336A1 (en) 2019-09-25 2021-04-01 持田製薬株式会社 Novel crosslinked alginic acid structure
AU2020364058A1 (en) 2019-10-11 2022-05-26 Quantum-Si Incorporated Surface modification in the vapor phase
WO2021090787A1 (en) * 2019-11-05 2021-05-14 国立大学法人東海国立大学機構 Polypeptide
WO2023205353A1 (en) * 2022-04-21 2023-10-26 Ultima Genomics, Inc. Self assembly of beads on substrates
EP4299596A1 (en) * 2022-06-30 2024-01-03 betaSENSE GmbH Molecular probe functionalized element

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4822867A (en) * 1987-02-02 1989-04-18 Semih Erhan Protein polymer grafts
US4886836A (en) * 1987-06-03 1989-12-12 Pall Corporation Activated medium with low non-specific protein adsorption
US5866322A (en) * 1988-01-29 1999-02-02 Abbott Laboratories Method for performing Rubella assay
EP0335308B1 (en) * 1988-03-31 1993-12-08 W.R. Grace & Co.-Conn. Protein non-adsorptive polyurea-urethane polymer coated devices
JP2570408B2 (en) * 1988-11-29 1997-01-08 東ソー株式会社 Carrier for immobilizing antigen, antibody or biologically active substance
US5330911A (en) * 1989-09-28 1994-07-19 Board Of Regents, The University Of Texas System Surfaces having desirable cell adhesive effects
US5459054A (en) * 1989-12-05 1995-10-17 Neocrin Company Cells encapsulated in alginate containing a high content of a- l- guluronic acid
US5626863A (en) 1992-02-28 1997-05-06 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5380536A (en) * 1990-10-15 1995-01-10 The Board Of Regents, The University Of Texas System Biocompatible microcapsules
US5529914A (en) * 1990-10-15 1996-06-25 The Board Of Regents The Univeristy Of Texas System Gels for encapsulation of biological materials
US5462990A (en) 1990-10-15 1995-10-31 Board Of Regents, The University Of Texas System Multifunctional organic polymers
SE467308B (en) * 1990-10-22 1992-06-29 Berol Nobel Ab SOLID SURFACE COATED WITH A HYDROPHILIC SURFACE WITH COVALENTLY BONDED BIOPOLYMERS, SET TO MAKE SUCH A SURFACE AND CONJUGATED THEREOF
US6093558A (en) * 1991-07-25 2000-07-25 Edge Biosystems, Inc. Binding protein of biologically active compositions to an adhesive formulation on a substrate
US5573934A (en) 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
DE4208645A1 (en) 1992-03-18 1993-09-23 Bayer Ag OPTICAL SOLID PHASE BIOSENSOR BASED ON FLUORESCENT COLOR-MARGINED POLYIONIC LAYERS
US5578442A (en) * 1992-03-23 1996-11-26 Vivorx, Inc. Graft copolymers of polycationic species and water-soluble polymers, and use therefor
US6113946A (en) * 1992-04-03 2000-09-05 The Regents Of The University Of California Self-assembling polynucleotide delivery system comprising dendrimer polycations
DE4310964A1 (en) * 1992-04-16 1993-10-21 Merck Patent Gmbh Activated carrier materials, their preparation and use
ATE207727T1 (en) * 1992-05-29 2001-11-15 Univ California COATED TRANSPLANT AND PRODUCTION PROCESS THEREOF
US5344455A (en) * 1992-10-30 1994-09-06 Medtronic, Inc. Graft polymer articles having bioactive surfaces
US5414075A (en) * 1992-11-06 1995-05-09 Bsi Corporation Restrained multifunctional reagent for surface modification
SE509662C2 (en) * 1993-04-29 1999-02-22 Tetra Laval Holdings & Finance Packaging laminate coated with a water-insoluble chitosan compound and methods of making the packaging laminate
ATE209782T1 (en) 1993-05-18 2001-12-15 Univ Utah Res Found DEVICE AND METHOD FOR HOMOGENEOUS MULTIANALYTE IMMUNO ASSAY
US5512492A (en) * 1993-05-18 1996-04-30 University Of Utah Research Foundation Waveguide immunosensor with coating chemistry providing enhanced sensitivity
US6284503B1 (en) * 1993-08-20 2001-09-04 University Of Utah Research Foundation Composition and method for regulating the adhesion of cells and biomolecules to hydrophobic surfaces
US5543326A (en) * 1994-03-04 1996-08-06 Heller; Adam Biosensor including chemically modified enzymes
CA2193968A1 (en) * 1994-06-28 1996-01-11 Jeffrey G. Clark Ph-modified biocompatible monomer and polymer compositions
US5853717A (en) * 1994-07-20 1998-12-29 Cytotherapeutics, Inc. Methods and compositions of growth control for cells encapsulated within bioartificial organs
US5507804A (en) * 1994-11-16 1996-04-16 Alcon Laboratories, Inc. Cross-linked polyethylene oxide coatings to improve the biocompatibility of implantable medical devices
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
US6017577A (en) * 1995-02-01 2000-01-25 Schneider (Usa) Inc. Slippery, tenaciously adhering hydrophilic polyurethane hydrogel coatings, coated polymer substrate materials, and coated medical devices
US5629213A (en) * 1995-03-03 1997-05-13 Kornguth; Steven E. Analytical biosensor
DE19530078A1 (en) * 1995-08-16 1997-02-20 Bayer Ag Optical solid phase biosensor based on streptavidin and biotin
US5916585A (en) * 1996-06-03 1999-06-29 Gore Enterprise Holdings, Inc. Materials and method for the immobilization of bioactive species onto biodegradable polymers
CN1173776C (en) * 1996-06-28 2004-11-03 卡钳技术有限公司 High-throughput screening assay systems in microscale fluidic devices
CA2202424A1 (en) * 1997-04-11 1998-10-11 Donald E. Brooks A tethered polymer macromolecule-excluding surface, its mode of synthesis and use
EP0975691B1 (en) 1997-04-21 2010-10-13 California Institute Of Technology Multifunctional polymeric coatings
US6287558B1 (en) * 1997-08-01 2001-09-11 Biohybrio Technologies Llc Devices containing cells or tissue and an agent that inhibits damage by a host cell molecule
US6121027A (en) * 1997-08-15 2000-09-19 Surmodics, Inc. Polybifunctional reagent having a polymeric backbone and photoreactive moieties and bioactive groups
US6348322B1 (en) * 1997-10-17 2002-02-19 Duke University Method of screening for specific binding interactions
US6235340B1 (en) * 1998-04-10 2001-05-22 Massachusetts Institute Of Technology Biopolymer-resistant coatings
US6150459A (en) * 1998-04-13 2000-11-21 Massachusetts Institute Of Technology Comb polymers for regulating cell surface interactions
US6303136B1 (en) * 1998-04-13 2001-10-16 Neurotech S.A. Cells or tissue attached to a non-degradable filamentous matrix encapsulated by a semi-permeable membrane
US6248127B1 (en) * 1998-08-21 2001-06-19 Medtronic Ave, Inc. Thromboresistant coated medical device
US6303179B1 (en) * 1999-02-08 2001-10-16 Medtronic, Inc Method for attachment of biomolecules to surfaces through amine-functional groups

Also Published As

Publication number Publication date
ATE419528T1 (en) 2009-01-15
EP1190252A1 (en) 2002-03-27
US20020128234A1 (en) 2002-09-12
JP2003516519A (en) 2003-05-13
WO2000065352A1 (en) 2000-11-02
AU4684600A (en) 2000-11-10
EP1190252B1 (en) 2008-12-31
US6884628B2 (en) 2005-04-26
DE60041255D1 (en) 2009-02-12
JP4647792B2 (en) 2011-03-09
AU769571B2 (en) 2004-01-29
CA2371011A1 (en) 2000-11-02

Similar Documents

Publication Publication Date Title
CA2371011C (en) Polyionic coatings in analytic and sensor devices
Wu et al. DNA and protein microarray printing on silicon nitride waveguide surfaces
Ostuni et al. The interaction of proteins and cells with self-assembled monolayers of alkanethiolates on gold and silver
Kenausis et al. Poly (L-lysine)-g-poly (ethylene glycol) layers on metal oxide surfaces: Attachment mechanism and effects of polymer architecture on resistance to protein adsorption
Rodriguez Emmenegger et al. Interaction of blood plasma with antifouling surfaces
US20100151491A1 (en) Chemical surface nanopatterns to increase activity of surface-immobilized biomolecules
JP2006234758A (en) Biosensor
CN101646943A (en) Method for blocking non-specific protein binding on a functionalized surface
Huang et al. Competitive protein adsorption on biomaterial surface studied with reflectometric interference spectroscopy
Vuoriluoto et al. Control of protein affinity of bioactive nanocellulose and passivation using engineered block and random copolymers
US20020068157A1 (en) Products with biofunctional coating
US7563624B2 (en) Measurement method using biosensor
US20040253596A1 (en) Bioanalytical recognition surface with optimized recognition element density
US20100129830A1 (en) Label Independent Detection Biosensor Composition and Methods Thereof
JP2007057458A (en) Biosensor
Davila et al. Bioaffinity sensor based on nanoarchitectonic films: Control of the specific adsorption of proteins through the dual role of an ethylene oxide spacer
Pan et al. Three-dimensional biosensor surface based on novel thorns-like polyelectrolytes
JP2006327984A (en) Method for synthesizing hetero-bifunctional oligoethylene glycol and method for producing biosensor using the same
JP2006266742A (en) Biosensor
Brynda et al. Ordered multilayer assemblies: Albumin/heparin for biocompatible coatings and monoclonal antibodies for optical immunosensors
EP1783493A1 (en) Biosensor for detecting or measuring a substance that interacts with a physiologically active substance comprising a polysaccharide capable of chemically immobilizing physiologically active subtances
Bellassai Surface Plasmon Resonance Imaging Biosensors for Cancer Diagnosis: Detection of Circulating Tumor DNA
JP2005221297A (en) Optical fiber sensor
JP4369295B2 (en) Measuring method using biosensor
JP4568175B2 (en) Biosensor and method for immobilizing physiologically active substance

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20200428