US20050008674A1 - Protein arrays for high-throughput screening - Google Patents

Protein arrays for high-throughput screening Download PDF

Info

Publication number
US20050008674A1
US20050008674A1 US10/911,945 US91194504A US2005008674A1 US 20050008674 A1 US20050008674 A1 US 20050008674A1 US 91194504 A US91194504 A US 91194504A US 2005008674 A1 US2005008674 A1 US 2005008674A1
Authority
US
United States
Prior art keywords
array
protein
proteins
monolayer
substrate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/911,945
Inventor
Peter Wagner
Dana Ault-Riche
Steffen Nock
Christian Itin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/911,945 priority Critical patent/US20050008674A1/en
Publication of US20050008674A1 publication Critical patent/US20050008674A1/en
Assigned to BILL & MELINDA GATES FOUNDATION reassignment BILL & MELINDA GATES FOUNDATION SECURITY AGREEMENT Assignors: ZYOMYX, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/0061The surface being organic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00612Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports the surface being inorganic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00617Delimitation of the attachment areas by chemical means
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00617Delimitation of the attachment areas by chemical means
    • B01J2219/00619Delimitation of the attachment areas by chemical means using hydrophilic or hydrophobic regions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/00626Covalent
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/0063Other, e.g. van der Waals forces, hydrogen bonding
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00635Introduction of reactive groups to the surface by reactive plasma treatment
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00639Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium
    • B01J2219/00641Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium the porous medium being continuous, e.g. porous oxide substrates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/00702Processes involving means for analysing and characterising the products
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00725Peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/10Libraries containing peptides or polypeptides, or derivatives thereof

Definitions

  • Genomics provides information on the genetic composition and the activity of an organism's genes.
  • Bioinformatics uses computer algorithms to recognize and predict structural patters in DNA and proteins, defining families of related genes and proteins. The information gained from the combination of these approaches is expected to boost the number of drug targets, usually proteins, from the current 500 to over 10,000 in the coming decade.
  • Automated multiwell formats are the best developed high-throughput screening systems. Automated 96-well plate-based screening systems are the most widely used. The current trend in plate based screening systems is to reduce the volume of the reaction wells flier, thereby increasing the density of the wells per plate (96-well to 384-, and 1536-well per plate). The reduction in reaction volumes results in increased throughput, dramatically decreased bioreagent costs, and a decrease in the number of plates which need to be managed by automation.
  • miniaturized cell-based assays have the potential to generate screening data of superior quality and accuracy, due to their in vivo nature.
  • the interaction of drug compounds with proteins other than the desired targets is a serious problem related to this approach which leads to a high rate of false positive results.
  • Microfluidics-based screening systems that measure in vitro reactions in solution make use of ten to several-hundred micrometer wide channels.
  • Micropumps, electroosmotic flow, integrated valves and mixing devices control liquid movement through the channel network.
  • Microfluidic networks prevent evaporation but, due to the large surface to volume ratio, result in significant protein inactivation. The successful use of microfluidic networks in biomolecule screening remains to be shown.
  • DNA microarray technology is not immediately transferable to protein screening microdevices.
  • microarrays are exclusively available for nucleic acid hybridiaztion assays (‘DNA-chips’). Their underlying chemistry and materials are not readily transferable to protein assays.
  • Nucleic acids withstand temperatures up to 100° C., can be dried and re-hydrated without loss of activity and bound directly to organic adhesion layers absorbed on surfaces such as glass.
  • proteins must remain hydrated, kept at ambient-temperatures, and are very sensitive to the physical and chemical properties of the support materials. Therefore, maintaining protein activity at the liquid-solid interface requires entirely different immobilization strategies than those used for nucleic acids. Additionally, the proper orientation of the protein at the interface is desirable to ensure accessibility of their active sites with interacting molecules.
  • cross-reactivity of a drug with related proteins can be the cause of low efficacy or even side effects in patients.
  • AZT a major treatment for AIDS
  • Block-reactivity with closely related proteins is also a problem with nonsteroidal anti-inflammatory drugs (NSAIDs) and aspirin.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • cyclooxygenase-2 an enzyme which promotes pain and inflammation.
  • cyclooxygenase-1 that is responsible for keeping the stomach lining and kidneys healthy, leading to common side-effects including stomach irritation.
  • the present invention is directed to protein arrays, protein-coated substrates, and methods of use thereof that satisfy the need for parallel, in vitro, high-throughput screening of functionally or structurally related protein targets against potential drug compounds in a manner that minimizes reagent volumes and protein inactivation problems.
  • the present invention provides for a proteinoated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where a protein with a different, known sequence is immobilized on each patch. Furthermore, each of the patches of the protein-coated substrate of the present invention is separated from neighboring patches by from about 50 nm to about 500 ⁇ m.
  • Biosensors, micromachined devices, and medical devices that comprise the protein-coated substrate of the present invention represent other aspects of the invention.
  • the present invention also provides an array of proteins comprising a plurality of patches arranged in discrete, known regions on a substrate, where a protein with a different, known sequence is immobilized on each patch. Furthermore, each of the patches of the protein-coated substrate of the present invention is separated from neighboring patches by from about 50 nm to about 500 ⁇ m.
  • the protein immobilized on one patch of the array is preferably different from the protein immobilized on a second patch.
  • the protein that is immobilized on one patch of the array is a member of the same protein family as or is otherwise functionally or structurally related to the proteins immobilized on the other patches of the array.
  • the patches of the array may also optionally further comprise monolayers (on which the proteins of the patches are immobilized).
  • At least one coating may be formed on the substrate or applied to the substrate of an array of the present invention such that the coating is positioned between the substrate and the monolayer of each patch
  • the coating, or the substrate itself if no coating is used, may optionally possess an ultraflat surface with a mean roughness of less than about 5 angstroms for areas of at least 25 ⁇ m 2 .
  • This ultraflat surface optionally may be produced by template stripping.
  • the monolayer of a patch on the array of the present invention may be a mixed monolayer composed of more than one type of molecule.
  • the patches of an array of the present invention may further comprise an affinity tag that enhances site-specific immobilization of the biological moiety onto the monolayer.
  • an adaptor molecule may also be present to link the affinity tag to the biological moiety on the patches of the array.
  • the affinity tag, biological moiety, and the adaptor preferably constitute a fusion protein.
  • the present-invention further provides for methods of using the array to screen a plurality of proteins in parallel for their ability to bind or otherwise interact with a component of a fluid sample. Most of these methods involve first delivering the fluid sample to the array. If binding is to be detected, the array may then be optionally washed to remove any unbound component from the area. The methods then involve detecting, either directly or indirectly, the presence or absence of the component retained at each patch or other evidence of an interaction of the protein of a given patch with the component.
  • Similar methods may be used diagnostically to screen a fluid sample with the array for the presence, absence, or amount of a plurality of analytes at the same time.
  • the present invention also provides for methods of determining in parallel whether or not a plurality of proteins belongs to a certain protein family. These methods involve delivering a fluid sample comprising a ligand of a known protein family to the patches of the array and then detecting, either directly or indirectly, for the interaction or binding of the known ligand to the patches that would be characteristic of the known protein family.
  • a protein-coated substrate that comprises a fusion protein immobilized on a monolayer on a portion of the surface of a substrate.
  • the fusion protein is immobilized with the aid of an affinity tag that enhances the site-specific immobilization of the fusion protein onto the monolayer.
  • the fusion protein comprises a polypeptide that serves as an adaptor molecule by linking another polypeptide to the affinity tag.
  • the monolayer of the proteincoated substrate comprises molecules of the formula X-R-Y where R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding the fusion protein onto the monolayer.
  • the protein-coated substrate may optionally also include a coating between the substrate and the monolayer and the affinity tag may optionally constitute a part of the fusion protein.
  • FIG. 1 shows the top view of an array of monolayer-covered patches.
  • FIG. 2 shows the cross section of an individual patch of the array of FIG. 1 .
  • FIG. 3 shows the cross section of a row of monolayer-covered patches of the array of FIG. 1 .
  • FIG. 4 shows aminoreactive monolayer molecules on a substrate.
  • FIG. 5 shows aminoreactive monolayer molecules on a coated substrate.
  • FIG. 6 shows the immobilization of a protein on a monolayer-coated substrate via an affinity tag.
  • FIG. 7 shows the immobilization of a protein on a monolayer-coated substrate via an affinity tag and an adaptor.
  • FIG. 8 shows four possible expression vectors useful for expressing fusion proteins of the desired protein, an affinity tag, and, optionally, an adaptor molecule.
  • FIG. 9 shows a schematic of a fluorescence detection unit which may be used to monitor interaction of the proteins of the array with an analyte.
  • a variety of protein arrays, methods, and protein-coated substrates useful for high-throughput drug screening, clinical diagnostics, and related processes are provided by the present invention.
  • substrate refers to the bulk underlying, and core material of the devices or arrays or other embodiments of the invention.
  • micromachining and “microfabricating” are both used herein to refer to any number of techniques which are useful in the generation of microstructures (structures of sub-millimeter scale). Such technologies include, but are not limited to, laser ablation, sputtering, electrodeposition, low-pressure vapor deposition, photolithography, and etching. Related technologies such as LIGA are also included Most of these techniques were originally developed for use in semiconductors, microelectronics, and microelectromechanical systems but are applicable to the present invention as well.
  • coating is used herein to refer to a layer that is either formed on or applied to the surface of the substrate.
  • a substrate such as silicon
  • a silicon oxide coating is formed on the surface upon exposure to air.
  • the coating is in no way derived from the substrate and may be placed upon the surface via mechanical, electrical or chemical means.
  • An example of this type of coating would be a metal coating that is applied to a polymer substrate.
  • a coating may be of any thickness, typically the coating has a thickness smaller than that of the substrate.
  • An “interlayer” is a second coating or layer that is positioned between the first coating and the substrate.
  • the primary purpose of a typical interlayer is to aid adhesion between the first coating and the substrate.
  • One such example is the use of a titanium interlayer to help attach a gold coating to a silicon chip.
  • other possible functions of an interlayer are also anticipated. For instance, some interlayers may perform a role in the detection system of the device.
  • affinity tag is used herein to refer to a functional moiety capable of immobilizing a protein onto the exposed functionality of a monolayer.
  • the affinity tag may be a simple chemical functional group. Other possibilities include amino acids, polypeptides, proteins, lipid bilayers, or a hydrogel.
  • the affinity tag may be either covalently or noncovalently attached to the protein (via chemical conjugation or as a fusion protein, for instance). Likewise, the affinity tag may bind to the monolayer either covalently or noncovalently.
  • an “adaptor molecule”, for purposes of this invention, is any entity that links an affinity tag to a protein.
  • the adaptor molecule need not necessarily be a discrete molecule that is noncovalently attached to both the affinity tag and the protein.
  • the adaptor molecule can be covalently attached to the affinity tag or the protein or both. (via chemical conjugation or as a fusion protein, for instance).
  • an affinity tag may also be an internal part of the protein, such as an amino acid.
  • Examples of adaptor molecules include polypeptides, proteins, membrane anchors, and biotin.
  • a “monolayer” may be disordered or ordered.
  • One face of the monolayer is composed of chemical functionalities on the termini of the organic molecules that are chemisorbed or physisorbed onto the surface material (headgroups).
  • the other face of the monolayer is exposed and may bear any number of chemical functionalities (end groups).
  • the molecules of the monolayer are highly ordered and tightly packed, largely due to hydrophobic and van der Waals interactions between the molecules.
  • An “array”, as used herein, refers to a two-dimensional pattern.
  • polypeptide and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. These terms also apply to amino acid polymers in which one or more amino acid residues is an artificial chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. An amino acid polymer in which one or more amino acid residues is an “nataral” amino acid, not corresponding to any naturally occurring amino acid, is also encompassed by the use of the terms “protein” and “polypeptide” herein.
  • Proteins are considered herein to be members of the same “protein family” or to be “related” if they show significant similarities in structure and/or function, as would be recognized by one of ordinary skill in the art, Related proteins can be identified by sequence homology searches of DNA and protein databases using standard bioinformatics resources and software packages (examples of public databases: NCBL NIH, EMBL, SwissProt, Brookhaven database, Washington University—Merck, private databases: Incyte, Hyseq, Human Genome Science; examples of software packages include EMOTIF, Blast, Fasta, Multalign, GCG Wisconsin University). Enzymatically related proteins of non-homologous sequence can be identified by one of ordinary skill in the art by screening the scientific literature (example: Medline database).
  • fusion protein refers to a protein composed of two or more polypeptides that, although typically unjoined in their native state, are joined by their respective amino and carboxyl termini through a peptide linkage to form a single continuous polypeptide. It is understood that the two or more polypeptide components can either be directly joined or indirectly joined through a peptide linker/spacer.
  • normal physiological condition is used herein to refer to conditions that are typical inside a living organism or a cell. While it is recognized that some organs or organisms provide extreme conditions, the intra-organismal and intra-cellular environment normally varies around pH 7 (i.e. from pH 6.5 to pH 7.5), contains water as the predominant solvent, and exists at a temperature above 0° C. and below 50° C. It will be recognized that the concentration of various salts depends on the organ, organism, cell, or cellular compartment used as a reference.
  • the present invention also provides a protein-coated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where each of the patches comprises an immobilized protein with a different, known sequence and where each of the patches is separated from the neighboring patches by from about 50 nm to about 500 ⁇ m.
  • the protein-coated substrate comprises 9 or more patches.
  • Biosensors, micromachined devices, and medical devices that contain the protein-coated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where each of the patches comprises an immobilized protein with a different, known sequence and where each of the patches is separated from neighboring patches by from about 50 nm to about 500 ⁇ m are also contemplated
  • the protein arrays comprise micrometer-scale, two-dimensional patterns of proteins immobilized on arrays of functionalized surface patches.
  • the array of proteins comprises a plurality of patches, preferably 9 or more, arranged in discrete known regions on a substrate, wherein each of the patches comprises an immobilized protein with a different, known sequence and wherein each of the patches is separated from neighboring patches by from about 50 nm to about 500 ⁇ m. In a preferred embodiment, the patches are separated from neighboring patches from about 200 nm to about 500 ⁇ m.
  • the diameter of each of the patches is proportional to the distance separating the patches. Therefore, the area of each patch may be from about 100 nm 2 to about 40,000 ⁇ m 2 . Each patch preferably has an area from about 1 ⁇ m 2 to about 10,000 ⁇ m 2 .
  • the array comprises 9 or more patches within a total area of 1 cm 2 . In preferred embodiments of the array, the array comprises 100 or more patches within a total area of 1 cm 2 . In another embodiment, the array comprises or more patches within a total area of 1 cm 2 .
  • the protein immobilized on one patch differs from the protein immobilized on a second patch of the same array.
  • the two biological moieties ate related.
  • the two different proteins are members of the same protein family.
  • the different proteins on the invention array may be either functionally related or just suspected of being functionally related.
  • the function of the immobilized proteins may be unknown.
  • the different proteins on the different patches of the array share a similarity in structure or sequence or are simply suspected of sharing a similarity in structure or sequence.
  • the immobilized proteins may be just fragments of different members of a protein family.
  • the proteins immobilized on the array of the invention may be members of a protein family such as a receptor family (examples: growth factor receptors, catecholamine receptors, amino acid derivative receptors, cytokine receptors, lectins), ligand family (examples: cytokines, serpins), enzyme family (examples: proteases, kinases, phosphatases, ras-like GTPases, hydrolases), and transcription factors (examples: steroid hormone receptors, heat-shock transcription factors, zinc-finger, leucine-zipper, homeodomain).
  • the different immobilized proteins are all HIV proteases or hepatitis C virus (HCV) proteases.
  • the protein immobilized on each patch is a different antibody or antibody fragment (Fab, for example).
  • the proteins on different patches are identical.
  • the substrate of the array may be either organic or inorganic, biological or non-biological or any combination of these materials.
  • the substrate is transparent or translucent.
  • the portion of the surface of the substrate on which the patches reside is preferably flat and firm or semi-firm.
  • Numerous materials are suitable for use as a substrate in the array embodiment of the invention.
  • the substrate of the invention array can comprise a material selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titanium dioxide, germanium, silicon nitride, zeolites, and gallium arsenide.
  • Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for substrates of the array.
  • polystyrene poly(tetra)fluorethylene; (poly)vinylidenedifluoride; polycarbonate; polymethylmethacrylate; polyvinylethylene; polyethyleneimine; poly(etherether)ketone; polyoxymethylene (POM); polyvinylphenol; polylactides; polymethacrylimide (PMI); polyalkenesulfone (PAS); polyhydroxyethylmethacrylate; polydimethylsiloxane; polyacrylamide; polyimide; co-block-polymers; and Eupergit®.
  • Photoresists, polymerized Langmuir-Blodgett films, and LIGA structures may also serve as substrates in the present invention.
  • the preferred substrates for the array comprise silicon, silica, glass, or a polymer.
  • the patches further comprise a monolayer on the surface of the substrate and the proteins of the patches are unmobilized on the monolayer.
  • the monolayer is preferably a self-assembling monolayer.
  • This monolayer may optionally comprise molecules of the formula X-R-Y, wherein R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding proteins onto the monolayer.
  • a variety of chemical moieties may function as monolayers in the array of the present invention.
  • three major classes of monolayer formation are preferably used to expose high densities of bioreactive omega-functionalities on the patches of the arrays (i) alkysiloxane monolayers (“silanes”) on hydroxylated surfaces (as taught in, for, example, U.S. Pat. No. 5,405,766, PCT Publication WO 96/38.726, U.S. Pat. No. 5,412,087, and U.S. Pat. No.
  • An array of the present invention may optionally further comprise a coating between the substrate and the monolayer of its patches.
  • This coating may either be formed on the substrate or applied to the substrate.
  • the substrate can be modified with a coating by using thin-film technology based on either physical vapor deposition (PVD) or plasma-enhanced chemical vapor deposition (PECVD).
  • PVD physical vapor deposition
  • PECVD plasma-enhanced chemical vapor deposition
  • plasma exposure can be used to directly activate the substrate.
  • plasma etch procedures can be used to oxidize a polymeric surface (i.e. polystyrene or polyethylene to expose polar functionlities such as hydroxyls, carboxylic acids, aldehydes and the like).
  • the coating may comprise a metal film.
  • Possible metal films include alumninum, chromium, titanium, nickel stainless steel zinc, lead, iron, magnesium, manganese, cadmium, tungsten, cobalt, and alloys or oxides thereof.
  • the metal film is a noble metal film.
  • Noble metals that may be used for a coating include, but are not limited to, gold, platinum, silver, copper, and palladium.
  • the coating comprises gold or a gold alloy. Electron-beam evaporation may be used to provide a thin coating of gold on the surface.
  • the metal film is from about 50 nm to about 500 nm in thickness.
  • the coating comprises a composition selected from the group consisting of silicon, silicon oxide, silicon nitride, silicon hydride, indium tin oxide, magnesium oxide, alumina, glass, hydroxylated surfaces, and a polymer.
  • the patches comprise a coating between the substrate and the monolayer, then it is understood that the coating must be composed of a material for which a suitable functional group X is available (see below). If no such coating is present, then it is understood that the substrate must be composed of a material for which a suitable functional group X is available.
  • the surface of the coating is ultraflat
  • the mean roughness of the surface of the coating is less than about 5 angstroms for areas of at least 25 ⁇ m 2 . In a preferred embodiment, the mean roughness of the surface of the coating is less than about 3 angstroms for areas of at least 25 m
  • the ultraflat coating can optionally be a template-stripped surface as described in Hegner et al., Surface Science, 1993, 291:3946 and Wagner et al., Langmuir, 1995, 11:3867-3875, both of which are incorporated herein by reference.
  • the coatings of many arrays will require the addition of at least one adhesion layer between said coating and said substrate.
  • the adhesion layer rill be at least 10 angstroms thick and may be much thicker.
  • a layer of titanium may be desirable between a silicon wafer and a goldcoating.
  • an epoxy glue such as Epo-tek 377®, Epo-tek 301-2®), (Epoxy Technology Inc., Billerica, Mass.) may be preferred to aid adherence of the coating to the substrate. Determinations as to what material should be used for the adhesion layer would be obvious to one skilled in the art once materials are chosen for both the substrate and coating.
  • additional adhesion mediators or interlayers may be necessary to improve the optical properties of the array, for instance, in waveguides for detection purposes.
  • Monolayer-compatible surface patches may optionally be fabricated by photolithography, micromolding (PCT Publication WO 96/29629), wet chemical etching, or any combination of these. Bio-reactive monolayers are then formed on the patches. Alternatively, arrays of bioreactive-monolayer functionalized surface patches can be created by microstamping (U.S. Pat. Nos. 5,512,131 and 5,731,152) or microcontact printing (LCP) (PCT Publication WO 96/29629). Subsequent immobilization of biomolecules results in two-dimensional protein arrays.
  • Inkjet chemical dispensers provide another option for patterning monolayer X-R-Y molecule or components thereof to nanometer or micrometer scale sites on the surface of the substrate or coating (Lemmo et al., Anal Chem., 1997, 69:543-'551).
  • Diffusion boundaries between the patches may be integrated as topographic patterns or surface functionalities with orthogonal wetting behavior.
  • walls of substrate material or photoresist may be used to separate some of the patches from some of the others or all of the patches from each other.
  • the patches are separated from each other by surfaces free of monolayers of the form X-R-Y.
  • non-bioreactive monolayers with different wettability may be used to separate patches from one another.
  • FIG. 1 shows the top view of one example of a monolayer-covered array. On the array, a number of patches 15 cover the surface of the substrate 3 .
  • FIG. 2 shows a detailed cross section of a patch of the array of FIG. 1 . This view illustrates the use of a coating 5 on the substrate 3 . An adhesion interlayer 6 is also included in the patch. On top of the patch resides a monolayer 7 .
  • FIG. 3 shows a cross section of one row of the monolayer-covered patches 15 of the array of FIG. 1 .
  • This figure also shows the use of a cover 2 over the array. Use of the cover 2 creates an inlet port 16 and an outlet port 17 for solutions to be passed over the array.
  • R may comprise a hydrocarbon chain from about 1 to about 200 carbons long.
  • the hydrocarbon chain may comprise an alkyl aryl, alkenyl alkynyl, cycloalkyl, alkaryl aralkyl group, or any combination thereof.
  • R is an alkyl chain from about 8 to about 22 carbons long and is optionally a straight alkane.
  • R may readily comprise a hydrocarbon chain from about 2 to about 200 carbons long and be interrupted by at least one hetero atom.
  • one or more of the hydrogen moieties of R can be substituted with deuterium.
  • X may be chosen as any group which affords chemisorption or physisorption of the monolayer onto the surface of the substrate (or the coating, if present).
  • X at least prior to incorporation into the monolayer, can in one embodiment be chosen to be an asymmetrical or symmetrical disulfide, sulfide, diselenide, selenide, thiol, isonitrile, selenol a trivalent phosphorus compound, isothiocyanate, isocyanate, xanthanate, thiocarbamate, a phosphine, an amine, thio acid or a dithio acid.
  • This embodiment is especially preferred when a coating or substrate is used that is a noble metal such as gold, silver, or platinum.
  • a preferred embodiment of the invention array comprises an X that, prior to incorporation into said monolayer, is a monohalosilane, dihalosilane, trihalosilane, trialkoxysilane, dialkoxysilane, or a monoalkoxysilane.
  • X is a trichlorosilane or trialkoxysilane.
  • the surface of the substrate (or coating thereon) is composed of a metal oxide such as titanium oxide, tantalum oxide, indium tin oxide, magnesium oxide, or alumina and X is a carboxylic acid.
  • X may optionally be a hydroxamic acid.
  • the substrate used in the invention is a polymer
  • a coating on the substrate such as a copper coating will be included in the device.
  • An appropriate functional group X for the coating would then be chosen for use in the device.
  • the surface of the polymer may be plasma-modified to expose desirable surface functionalities for monolayer formation.
  • EP 780423 describes the use of a monolayer molecule that has an alkene X functionality on a plasma exposed surface.
  • Still another possibility for the invention device comprised of a polymer is that the surface of the polymer on which the monolayer is formed is functionalized due to copolymerization of appropriately functionalized precursor molecules.
  • X prior to incorporation into the monolayer, can be a free-radical-producing moiety.
  • This functional group is especially appropriate when the surface on which the monolayer is formed is a hydrogenated silicon surface.
  • free-radical producing moieties include, but are not limited to, diacylperoxides, peroxides, and azo compounds.
  • unsaturated moieties such as unsubstituted alkenes, alkynes, cyano compounds and isonitrile compounds can be used for X, if the reaction with X is accompanied by ultraviolet, infrared, visible, or microwave radiation.
  • X prior to incorporation into the monolayer, may be a hydroxyl, carboxyl vinyl sulfonyl, phosphoryl silicon hydride, or an amino group.
  • the component, Y, of the monolayer is a functional group responsible for binding a protein onto the monolayer.
  • the Y group is either highly reactive (activated) towards the protein or is easily converted into such an activated form.
  • the coupling of Y with the protein occurs readily under normal physiological conditions not detrimental to the biological activity of the protein.
  • the functional group Y may either form a covalent linkage or a noncovalent linkage with the protein (or its affinity tag, if present).
  • the functional group Y forms a covalent linkage with the protein or its affinity tag. It is understood that following the attachment of the protein to Y, the chemical nature of Y may have changed.
  • Y is a functional group that is activated in situ. Possibilities for this type of functional group include, but are not limited to, such simple moieties such as a hydroxyl, carboxylamino, aldehyde, carbonyl, methyl, methylene, alkene, alkyne, carbonate, aryliodide, or a vinyl group. Appropriate modes of activation would be obvious to one skilled in the art Alternatively, Y can comprise a functional group that requires photoactivation prior to becoming activated enough to trap the biological moiety.
  • Y is a complex and highly reactive functional moiety that is compatible with monolayer formation and needs no in situ activation prior to reaction with the protein and/or affinity tag.
  • Such possibilities for Y include, but are not limited to, maleimide, N-hydroxysuccimmide (Wagner et al., Biophysical Journal, 1996, 70:2052-2066), nitrilotriacetic acid (U.S. Pat. No.
  • FIG. 4 shows one example of a monolayer on a substrate 3 .
  • substrate 3 comprises silicon (having a silicon oxide surface).
  • the monolayer is aminoreactive because it bears a functional group Y that is N-hydroxysuccimmide.
  • FIG. 5 shows another example of a monolayer on a substrate 3 .
  • a thin film coating 5 comprised of gold covers the surface of the substrate 3 .
  • an adhesion interlayer 6 is used to adhere the coating 5 to the substrate 3 and is comprised of titanium.
  • This monolayer is also aminoreactive because it bears a functional group Y that is N-hydroxysuccinimide.
  • the functional group Y of the array is selected from the group of simple functional moieties.
  • Possible Y functional groups include, but are not limited to, —OH, —NH 2 , —COOH, —COOR, —RSR, —PO 4 ⁇ 3 , OSO 3 ⁇ 2 , —SO 3 ⁇ , —COO ⁇ , —SOO ⁇ , —CONR 2 , —CN, —NR 2 , and the like.
  • Simple groups such as these are only preferred for Y when the affinity tag of the invention composes a layer of affinity tag molecules (such as poly-lysine) that coats the exposed portion of the monolayer prior to immobilization of the biological moiety (see below).
  • the monolayer molecules of the present invention can optionally be assembled on the surface in parts.
  • the monolayer need not necessarily be constructed by chemisorption or physisorption of molecules of the formula X-R-Y to the surface of the substrate (or coating).
  • X may be chemisorbed or physisorbed to the surface of the substrate (or coating) alone first Then, R or even just individual components of R can be attached to X through a suitable chemical reaction.
  • Y can be attached to the ends of the monolayer molecule through a suitable covalent linkage.
  • the monolayer of an individual patch may optionally comprise at least two different X-R-Y molecules. This second X-R-Y molecule may immobilize the same or a different protein from the first In addition, some of the monolayer molecules X-R-Y of a patch may have failed to attach any protein.
  • the monolayer of an individual patch on the array may comprise a second molecule that is of the formula, X-R-V where R is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins. V must also be biocompatible with proteins. Typically, V will consist of a hydroxyl, saccharide, or polyethylene glycol moiety (EP Publication 780423).
  • the array may further comprise at least one unreactive patch devoid of protein, wherein said patch comprises a monolayer of molecules of the formula X-R-V, where R is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins.
  • crosslinking between molecules of an individual patch's monolayer.
  • crosslinking confers additional stability to the monolayer.
  • Such methods are familiar to those skilled in the art (for instance, see Ulman, An Introduction to Ultrathin Organic Films: From Langmuir - Blodgett to Self - Assembly , Academic Press (1991)).
  • the protein may be attached to the monolayer via interaction with the Y-functional group.
  • Y-functional groups which fail to react with any protein molecules are preferably quenched prior to use of the array.
  • the array further comprises an affinity tag that enhances site-specific immobilization of the biological moiety onto the monolayer.
  • an affinity tag can confer enhanced binding or reaction of the protein with Y. This enhancement effect may be either kinetic or thermodynamic.
  • the affinity tag/Y-functional group pair used in the patches of the array preferably allows for immobilization of the biological molecules in a manner which does not require harsh reaction conditions that are adverse to protein stability or function. In most embodiments, immobilization in aqueous, biological buffers are ideal.
  • An affinity tag also preferably offers immobilization that is specific to a designated site or location on the biological moiety.
  • affinity tag For this to occur, attachment of the affinity tag to the protein must be site-specific. This site specific immobilization helps ensure that the reactive site of the protein remains accessible to ligands in solution. Another advantage of immobilization through affinity tags is that it allows for a common immobilization strategy to be used with multiple, different proteins.
  • the affinity tag comprises at least one amino acid.
  • the affinity tag may be a polypeptide comprising at least one monolayer-reactive amino acid.
  • the affinity tag may be a lone, monolayer-reactive amino acid. Examples of possible monolayer-reactive amino acids include cysteine; lysine, histidine, arginine, tyrosine, and glutamine.
  • a polypeptide or amino acid affinity tag is preferably expressed as a fusion protein with the biological moiety.
  • Amino acid tags provide either a single amino acid or a series of amino acids that can interact with the Y-functional group of the monolayer molecules. Amino acid affinity tags can be readily introduced into recombinant proteins to facilitate oriented immobilization by covalent binding to the bioreactive Y-functional group of the monolayer.
  • the affinity tag may comprise a poly(amino acid) tag.
  • a poly(amino acid) tag is a polypeptide that comprises from about 2 to about 100 residues of a single amino acid, optionally interrupted by residues of other amino acids.
  • the affinity tag may comprise a poly-cysteine, polylysine, poly-arginine, or poly-histidine.
  • Amino acid tags are preferably composed of two to twenty residues of a single amino acid, such as, for example, histidines, lysines, arginines, cysteines, glutanunes, tyrosines, or any combination of these.
  • an amino acid tag of one to twenty amino acids includes at least one to ten cysteines for thioether linkage; or one to ten lysines for amide linkage; or one to ten arginines for coupling to vicinal dicarbonyl groups.
  • cysteines for thioether linkage
  • ten lysines for amide linkage
  • arginines for coupling to vicinal dicarbonyl groups.
  • the position of the amino acid tag can be at the amino-, or carboxy-terminus of the protein or anywhere in-between.
  • affinity tags introduced for protein purification are preferentially located at the C-terminus of the recombinant protein to ensure that only full-length proteins are isolated during protein purification.
  • Affinity tags may also contain one or more unnatural amino acids.
  • Unnatural amino acids can be introduced using suppressor tRNAs that recognize stop codons (i.e. amber) (Noten et al., Science, 1989, 244:182-188; Ellman et al., Methods Enzym, 1991, 202:301-336; Cload et al., Chem Biol., 1996, 3:1033-1038).
  • the tRNAs are chemically amino-acylated to contain chemically altered (“unnatural”) amino acids for use with specific coupling chemistries (i.e. ketone modifications, photoreactive groups).
  • the affinity tag can comprise a whole protein, such as, but not limited to, glutathione S-transferase, an antibody, avidin, or streptavidin.
  • the affinity tag may be an organic bioconjugate which is chemically coupled to the protein of interest.
  • Biotin or antigens may be chemically cross linked to the protein.
  • a chemical cross linker may be used that attaches a simple functional moiety such as a thiol or an amine to the surface of a protein.
  • FIG. 6 shows a detailed cross section of a patch on one embodiment of the invention array.
  • a protein 10 is immobilized on a monolayer 7 on a substrate 3 .
  • An affinity tag 8 connects the biological moiety 10 to the monolayer 7 .
  • the monolayer 7 is formed on a coating 5 which is separated from the substrate 3 by an interlayer 6 .
  • the affinity tag is a component of a layer of affinity tag molecules immobilized on the monolayer of an individual patch.
  • a hydrogel composed of a material such as dextran can serve as a suitable layer of affinity tag molecules in the array.
  • Use of such hydrogels to immobilize biological moieties is described in U.S. Pat. No. 5,242,828.
  • Poly-lysine is another option for a material useful in forming an affinity-tag layer (for an example see U.S. Pat. No. 5,629,213).
  • the layer of affinity tag molecules could also constitute a phospholipid bilayer or a phospholipid monolayer as described in PCT Publication WO 96/38726. Use of a phospholipid monolayer or bilayer as an affinity tag would be suitable if the biological moiety to be immobilized is a membrane protein, such as an ion channel protein.
  • Protein solutions may be transferred to the appropriate patches via noncontact printing using a microdelivery device employing a ball-point pen type of mechanism.
  • microdelivery device employing a ball-point pen type of mechanism.
  • microcapirary-based dispensing systems may be used. These dispensing systems are preferably computer-aided. The use of other microprinting techniques for transferring protein solutions to the bioreactive patches is also possible.
  • Another major embodiment of the arrays of the present invention comprises an adaptor molecule that links the affinity tag to the immobilized biological moiety.
  • the additional spacing of the protein from the surface of the substrate (or coating) that is afforded by the use of an adaptor molecule is particularly advantageous since proteins are known to be prone to surface inactivation.
  • an adaptor molecule which is appropriate for a given affinity tag. For instance, if the affinity tag is streptavidin, then the adaptor could be a biotin molecule that is chemically conjugated to the protein which is to be immobilized. Alternatively, if the affinity tag is a phospholipid biolayer or monolayer then a membrane anchor could be chosen as a suitable adaptor molecule.
  • the adaptor molecule is a polypeptide, such as protein G or protein A
  • the affinity tag, adaptor molecule, and biological moiety together compose a fusion protein
  • a fusion protein may be readily expressed using standard recombinant DNA technology.
  • Adaptor proteins are especially useful to increase the solubility of the protein of interest and to increase the distance between the surface of the substrate or coating and the protein of interest. Use of an adaptor protein or polypeptide can also be very useful in facilitating the preparative steps of protein purification by affinity binding prior to immobilization on the array.
  • GFP glutathione-S-transferase
  • maltose-binding protein chitin-binding protein
  • thioredoxin green-fluorescent protein
  • GFP can also be used for quantification of surface binding.
  • FIG. 7 shows a cross section of a patch on one particular embodiment of the invention array.
  • the patch comprises a protein 10 immobilized on a monolayer 7 via both an affinity tag 8 and an adaptor molecule 9 .
  • the monolayer 7 rests on a coating 5 .
  • An interlayer 6 is used between the coating 5 and the substrate 3 .
  • proteins can be expressed from recombinant. DNA either in vivo or in vitro.
  • Amino acid affinity tags are introduced by polymerase chain reaction. Expression in vivo is in either bacteria ( Escherichia coli ), lower eukaryotes ( Saccharomyces cerevisiae, Saccharomyces pombe, Pichia pastoris ) or higher eukaryotes (bacculo-infected insect cells, insect cells mammalian cells), or in vitro ( Escherichia coli lysates, wheat germ extracts, reticulocyte lysates). Proteins are purified by affinity chromatography using commercially available resins.
  • DNA sequences encoding amino acid affinity tags and adaptor proteins are engineered into the expression vectors such that the genes of interest can be cloned in frame either 5′ or 3′ of the DNA sequence encoding the affinity tag and adaptor protein.
  • FIG. 8 shows four possible expression vectors for expressing the protein of interest, a polypeptide affinity tag, and a polypeptide adaptor molecule as a fusion protein.
  • the vector contains an origin of replication sequence 35 and a gene 36 capable of conferring antibiotic resistance to a host cell.
  • the insert of the vector contains a promoter sequence 30 and a termination signal sequence 34 . Between the sequences 30 and 34 , the insert also contains a gene 33 encoding the protein of interest and sequence 31 encoding the polypeptide affinity tag.
  • Sequence 32 which codes for a polypeptide adaptor molecule may also be included on the plasmid and is positioned between the protein and affinity-tag coding regions ( 33 and 31 , respectively).
  • cDNAs for the protein of interest will be amplified by PCR using cDNA libraries or EST (expressed sequence tag) clones as templates.
  • cDNAs can be cloned into commercial expression vectors (Qiagen, Novagen, Clontech) and introduced into the appropriate organism for expression (organisms include. Escherichia coli, Saccharomyces cerevisiae, Saccharomyces pombe, Pichiapastoris , bacculovirus/insect cells, insect cells, mammalian cells).
  • PCR-amplified DNA sequences are directly used in coupled in vitro transcription/translation systems ( Escherichia coil S30 lysates from T7 RNA polymerase expressing, preferably protease-deficient strains, wheat germ lysates, reticulocyte lysates with and without microsomes (Promega, Pharmacia, Panvera)).
  • the choice of organism for optimal expression depends on the extent of post-translational modifications (i.e. glycosylation, lipid-modifications).
  • Escherichia coli based protein expression will be the method of choice for soluble proteins that do not require extensive post-translational modifications for activity. Extracellular or intracellular domains of membrane proteins will be fused to protein adaptors for expression and purification.
  • PCR reactions are carried out under standard conditions. Oligonucleotide primers contain unique restriction sites for facile cloning into the expression vectors. Alternatively, the TA cloning system (Clontech) can be used. Expression vectors contain the sequences for affinity tags and the protein adaptors. PCR products are ligated into the expression vectors (under inducible promoters) and introduced into the appropriate competent Escherichia coli strain by calcium-dependent transformation (strains include: XL-1 blue, BL21, SG13009(Ion-)). Transformed Escherichia coli cells are plated and individual colonies transferred into 96-array blocks.
  • Cultures are grown to mid-log phase, induced for expression, and cells collected by centrifugation Cells are resuspended containing lysozyme and the membranes broken by rapid freeze/thaw cycles, or by sonication. Cell debris is removed by centrifugation and the supernatants transferred to 96-tube arrays.
  • the appropriate affinity matrix is added, protein of interest bound and nonspecifically bound proteins removed by repeated washing steps using 12-96 pin suction devices and centrifugation.
  • magnetic affinity beads and filtration devices can be used (Qiagen). The proteins are eluted and transferred to a new 96-well array.
  • Protein concentrations are determined and an aliquot of each protein is spotted onto a nitrocellulose filter and verified by Western analysis using an antibody directed against the affinity tag. The purity of each sample is assessed by SDS-PAGE and Coomassie staining or mass spectroscopy. Proteins are snap-frozen and stored at ⁇ 80° C.
  • Saccharomyces cerevisiae allows for core glycosylation and lipid modifications of proteins.
  • the approach described above for Escherichia coli can be used with slight modifications for transformation and cell lysis. Transformation of Saccharomyces cerevisiae is by lithium-acetate and cell lysis is either by lyticase digestion of the cell walls followed by freeze-thaw, sonication or glass-bead extraction. Variations of post-translational modifications can be obtained by different yeast strains (i.e. Saccharomyces pombe, Pichia pastoris ).
  • the advantage of the bacculovirus system or mammalian cells are the wealth of post-translational modifications that can be obtained.
  • the baeculo-system requires cloning of viruses, obtaining high titer stocks and infection of liquid insect cell suspensions (cells are SF9, SF21).
  • Mammalian cell-based expression requires transfection and cloning of cell lines. Soluble proteins are collected from the medium while intracellular or membrane bound proteins require cell lysis (either detergent solubilization, freeze-thaw). Proteins can then be purified analogous to the procedure described for Escherichia coli.
  • Escherichia coli lysates obtained from protease-deficient and 17 RNA polymerase overexpressing strains.
  • Escherichia coli lysates provide efficient protein expression (30-50 ⁇ g/ml lysate). The entire process is carried out in 96-well arrays.
  • Genes of interest are amplified by PCR using oligonucleotides that contain the gene-specific sequences containing a 17 RNA polymerase promoter and binding site and a sequence encoding the affinity tag.
  • an adaptor protein can be fused to the gene of interest by PCR.
  • Amplified DNAs can be directly transcribed and translated in the Escherichia coli lysates without prior cloning for fast analysis. The proteins are then isolated by binding to an affinity matrix and processed as described above.
  • the present invention also provides for methods of using the invention array.
  • the arrays of the present invention are particularly suited for the use in high-throughput drug screening. Other uses include medical diagnostics and biosensors.
  • Use of one of the protein arrays of the present invention may optionally involve placing the two-dimensional protein array in a flowchamber with approximately 1-10 microliters of fluid volume per 25 mm 2 overall surface.
  • the cover over the array in the flowchamber is preferably transparent or translucent.
  • the cover may comprise Pyrex or quartz glass.
  • the cover may be part of a detection system that monitors interaction between biological moieties immobilized on the array and an analyte.
  • the flowchambers should remain filled with appropriate aqueous solutions to preserve protein activity. Substrates and potential drug compounds may be flushed into the flow chamber as desired and their interaction with the immobilized proteins determined. Hence, no specialized microfluidic pumps, valves, or mixing techniques are required for fluid delivery to the array.
  • fluid can be delivered to each of the patches of the array individually.
  • the regions of the substrate surface may be microfabricated in such a way as to allow integration of the array with a number of fluid delivery channels oriented perpendicular to the array surface, each one of the delivery channels terminating at the site of an individual proteincoated patch.
  • Possible interactions towards which the present invention may be directed include, but are not limited to, antibody/antigen, antibody/hapten, enzyme/substrate, carrier protein/substrate, lectin/carbohydrate, receptor/hormone, receptor/effector, protein/DNA, protein/RNA, repressor/inducer, or the like.
  • a method for screening a plurality of proteins for their ability to interact with a component of a fluid sample comprises delivering the fluid sample to the invention array, and detecting the interaction of said component with the immobilized protein of each patch.
  • the invention device can be interfaced with optical detection methods such as absorption in the visible range, chemoluminescence, and fluorescence (including lifetime, polarization, fluorescence correlation spectroscopy (FCS), and fluorescence-resonance energy transfer (FRET)).
  • optical detection methods such as absorption in the visible range, chemoluminescence, and fluorescence (including lifetime, polarization, fluorescence correlation spectroscopy (FCS), and fluorescence-resonance energy transfer (FRET)
  • built-in detectors such as optical waveguides PCT Publication WO 96/26432 and U.S. Pat. No. 5,677,196, surface plasmons, and surface charge sensors are compatible with many embodiments of thee invention.
  • FIG. 9 shows a schematic diagram of one type of fluorescence detection unit which may be used to monitor interaction of immobilized proteins of an array with an analyte.
  • the protein array 21 is positioned on a base plate 20 .
  • Light from a 100 W mercury arc lamp 25 is directed through an excitation filter 24 and onto a beam splitter 23 .
  • the light is then directed through a lens 22 , such as a Micro Nikkor 55 mm 1:2:8 lens, and onto the array 21 .
  • Fluorescence emission from the array retuns through the lens 22 and the beam splitter 23 .
  • the emission is received by a cooled CCD camera 27 , such as the Slowscan TE/CCD-1024SF&SB (Princeton Instruments).
  • the camera is operably connected to a CPU 28 which is in turn operably connected to a VCR/monitor 29 .
  • Another aspect of the invention provides for a method for screening a plurality of proteins for their ability to bind a component of a fluid sample.
  • This method comprises, delivering said fluid sample to the invention array, washing the array with fluid which does not contain said component in order to elute unbound component from the surface of the array, and detecting, either directly or indirectly, the presence, absence, or amount of the component retained at each patch.
  • Another embodiment of the invention provides a method for detecting in parallel the ability of a drug candidate to interfere with the ability of a singular analyte or a plurality of analytes in a fluid sample to bind one or more immobilized proteins on the invention array.
  • This method comprises delivering the fluid sample to the invention array, washing said array with analyte free fluid to remove unbound analyte, and detecting, either directly or indirectly, the presence of analyte retained at each patch.
  • the array of the present invention may also be used in a diagnostic manner.
  • the plurality of proteins immobilized on the array are not preferably members of the same protein family.
  • One embodiment of the invention provides a method for detecting in parallel the presence of a plurality of analytes in a fluid sample which react with one or more of the immobilized proteins. This method comprises delivering the fluid sample to the invention array and detecting the interaction of the analyte with the immobilized protein at each patch.
  • a method for detecting in parallel the presence of a plurality of analytes in a fluid sample which bind said one or more of the immobilized proteins comprises delivering the fluid sample to the invention array, washing said array with an analyte-free fluid to remove unbound analyte, and detecting, either directly or indirectly, the presence of analyte retained at each patch.
  • Another embodiment of the present invention is a protein-coated substrate comprising the following a substrate; a monolayer on a portion of a surface of the substrate that comprises molecules of the formula X-R-Y where R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding a fusion protein onto the monolayer; an affinity tag that enhances site-specific immobilization of the fusion protein onto the monolayer; and a fusion protein of a first polypeptide linked to a second, adaptor polypeptide, immobilized on said monolayer by means of the affinity tag that is linked to the second, adaptor polypeptide.
  • the substrate may be organic or inorganic, biological or non-biological, or any combination of these materials.
  • the substrate of the invention can optionally comprise a material selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titanium dioxide, germanium, silicon nitride, zeolites, and gallium arsenide. Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for substrates. In addition, many ceramics and polymers may be used as substrates. Photoresists, polymerized Langmuir-Blodgett films, and LIGA structures may also serve as substrates in the present invention.
  • the preferred substrates of the present invention comprise sililcon, silica, glass, or a polymer.
  • the protein-coated substrate further comprises a coating between said substrate and said monolayer.
  • This coating may be formed on the substrate or applied to the substrate.
  • the coating on the substrate may comprise a metal film.
  • the coating is a noble metal film.
  • Many other different coatings, such as a polymer, glass, a hydroxylated surface, silicon nitride and silicon oxide are also compatible with the present invention.
  • the protein-coated substrate will further comprise at least one adhesion layer or mediator between the coating and the substrate.
  • a variety of chemical moieties may function as monolayers in the array of the present invention.
  • three major classes of monolayer formation are preferably used to expose high densities of bioreactive omega-functionalities on the patches of the array: (i) alkylsiloxane monolayers. (“silanes”) on hydroxylated surfaces (as taught in, for example, U.S. Pat. No. 5,405,766, PCT Publication WO 96/38726, U.S. Pat. No. 5,412,087, and U.S. Pat. No.
  • R of a monolayer molecule may comprise a hydrocarbon chain from about 1 to about 200 carbons long.
  • the hydrocarbon chain may comprise an alkyl, aryl, alkenyl alkynyl, cycloalkyl, alkaryl, aralkyl group, or any combination thereof.
  • R is an alkane from about 8 to about 22 carbons long.
  • R may readily comprise a hydrocarbon chain from about 2 to about 200 carbons long and interrupted by at least one hetero atom.
  • X may be chosen as any group which affords chemisorption or physisorption of the monolayer onto the surface of the substrate (or the coating, if present).
  • X at least prior to incorporation into the monolayer, may be chosen to be an asymmetrical or symmetrical disulfide, sulfide, diselenide, selenide, thiol, isonitrile, selenol trivalent phosphorus compounds, isothiocyanate, isocyanate, xanthanate, thiocarbamate, phosphines, amines, thio acid and dithio acid.
  • This embodiment is especially preferred when a coating or substrate is used that is a noble metal such as gold, silver, or platinum.
  • X prior to incorporation into the monolayer may be a hydroxyl, carboxyl vinyl sulfonyl phosphoryl silicon hydride, or an amino group. Another possibility is that prior to incorporation into the monolayer, X can be a free-radical-producing moiety.
  • the substrate of the protein-coated substrate is a material such as silicon, silicon oxide, indium tin oxide, magnesium oxide, alumina, quartz, glass, or silica
  • a preferred embodiment of the invention comprises an X that, prior to incorporation into said monolayer, is a monohalosilane, dihalosilane, trihalosilane, trialkoxysilane, dialkoxysilane, or a monoalkoxysilane.
  • the silane is a trichlorosilane or trialkoxysilane.
  • the component, Y, of the monolayer is responsible for binding a biological moiety onto the monolayer.
  • the Y group is either highly reactive (activated) towards the biological moiety or is easily converted into such an activated form.
  • the coupling of Y with the biological moiety occurs readily under physiological conditions.
  • Y is a functional group that is activated in situ. Possibilities for this type of functional group include, but are not limited to; such simple moieties such as a hydroxyl, carboxyl, amino, aldehyde, carbonyl methyl methylene, alkene, alkyne, carbonate, aryliodide, or a vinyl group.
  • Y can comprise a functional group that requires photoactivation prior to becoming activated enough to trap the biological moiety.
  • Y is a highly reactive functional moiety.
  • Y include, but are not limited to, maleimide, N-hydroxysuccinimide, nitrilotriacetic acid, activated hydroxyl haloacetyl bromoacetyl, iodoacetyl activated carboxyl, hydrazide, epoxy, aziridine, trifluoromethyldiaziridine, pyridyldisulfide, N-acyl-imidazole, imidazolecarbamate, succinimidylcarbonate, arylazide, anhydride, diazoacetate, benzophenone, isothiocyanate, isocyanate, imidoester, fluorobenzene, and biotin
  • the monolayer of the protein-coated substrate can optionally comprise at least two different X-R-Y molecules.
  • the monolayer can further comprise a second molecule that is of the formula X-R-V wherein k is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of polypeptides and proteins.
  • the affinity tag is a polypeptide or an amino acid.
  • the affinity tag comprises a poly(amino acid).
  • Amino acid tags provide either a single amino acid or a series of amino acids that can interact with the Y-functionalities of the monolayer.
  • Amino acid affinity tags can also be introduced to a specific site on a recombinant protein to facilitate oriented immobilization by covalent binding to the bioreactive Y-functional group of the monolayer.
  • the affinity tag may comprise a poly(amino acid) tag.
  • a poly(amino acid) tag is a polypeptide at comprises from about 2 to about 100 residues of a single amino acid.
  • the affinity tag may comprise a poly-cysteine, poly-lysine, poly-arginine, or poly-histidine.
  • Amino acid tags are preferably composed of two to twenty residues of a single amino acid, such as, for example, histidines, lysines, arginines, cysteines, glutamines, tyrosines, or any combination of these.
  • an amino acid tag of one to twenty amino acids includes at least one to ten cysteines for thioether linkage; or one to ten lysines for amide linkage; or one to ten arginines for coupling to vicinal dicarbonyl groups.
  • cysteines for thioether linkage or one to ten lysines for amide linkage; or one to ten arginines for coupling to vicinal dicarbonyl groups.
  • the position of the amino acid tag can be at the amino-, or carboxy-terminus of the protein or anywhere in-between.
  • affinity tags introduced for protein purification are preferentially located at the C-terminus of the recombinant protein to ensure that only full-length proteins are isolated during protein purification.
  • Affinity tags may also contain one or more unnatural amino acids.
  • Unnatural amino acids can be introduced using suppressor tRNAs that recognize stop codons (i.e. amber).
  • the tRNAs are chemically amino-acylated to contain chemically altered (“unnatural”) amino acids for use with specific coupling chemistries (i.e. ketone modifications, photoreactive groups).
  • the affinity tag can comprise a whole protein, such as, but not limited to, glutathione S-transferase, an antibody, avidin, or streptavidin.
  • protein conjugation and immobilization techniques known in the art may be adapted for the purpose of immobilizing proteins on activated monolayers.
  • an organic bioconjugate such as biotin or an antigen, amy be chemically cross linked to the protein to be immobilized.
  • the fusion protein that comprises the first and second polypeptides comprises the affinity tag as well.
  • Methods for the expression of fusion proteins have been outlined above.
  • the adaptor polypeptide is protein G or protein A.
  • Adaptor proteins are especially useful to increase the solubility of the protein of interest and to increase the distance between the surface of the substrate or coating and the protein of interest
  • Use of an adaptor protein or polypeptide can also be very useful in facilitating the preparative steps' of protein purification by affinity binding.
  • Examples of possible adaptor proteins include glutathione-S-transferase (GST, maltose-binding protein, chitin-binding protein, thioredoxin, green-fluorescent protein (GFP). GFP can also be used for quantification of surface binding.
  • two-dimensional arrays are fabricated onto the device material via standard microstereolithography and/or thin film deposition.
  • Alternative techniques include microcontact printing.
  • a computer-aided design pattern (reflecting the final channel geometries) is transferred to a photomask using standard techniques, which is then used to transfer the pattern onto a silicon wafer coated with photoresist.
  • the device with lateral dimensions of 20 ⁇ 20 mm contains an array of squared patches of a bioreactive layer (here: gold on silicon) each 0.1 ⁇ 0.1 mm in size and separated by hydrophobic surface areas with a 0.2 mm spacing.
  • a bioreactive layer here: gold on silicon
  • 4′ diameter Si(100) wafers (Virginia Semiconductor) or 4′′ diameter Corning 7740 glass wafers are used as bulk materials.
  • Si(100) wafers are first cleaned in a 5:1:1 deionized (DI) water: NH 3 : H 2 O 2 bath (RCA1, 90° C., 10 min), followed by a 5:1:1 DI water: HCl H 2 O 2 bath (RCA2, 90° C., 10 min) and finally passivated in 1% aqueous HF and singed at 150° C. for 30 min to become hydrophobic.
  • DI deionized
  • HCl H 2 O 2 bath RCA2, 90° C., 10 min
  • the wafer is then spincoated with photoresist (Shipley 1813), prebaked for 25 minutes at 90° C., exposed using a Karl Suss contact printer and developed according to standard protocols.
  • the wafer is then dried and postbaked at 110° C.
  • the wafer is primed with a 20 nm thin titanium layer, followed by a 200 nm thin gold layer both layers deposited using electron-beam evaporation (5 ⁇ /s, Thermionics).
  • the gold patches can be further chemically modified to achieve the desired bioreactive and biocompatible properties (see Example 3, below).
  • the array of gold patches is fabricated by thin film deposition through a hole mask which is in direct contact with the substrate.
  • Si(100) wafers are first cleaned in a 5:1:1 deionized (DI) water: NH 3 H 2 O 2 bath (RCA1, 90° C., 10 min), followed by a 5:1:1 DI water: HCl: H 2 O 2 bath (RCA2, 90° C., 10 min) and finally passivated in 1% aqueous HF and singed at 150° C. for 30 min to become hydrophobic.
  • DI deionized
  • HCl H 2 O 2 bath
  • the wafer is then brought into contact with a hole mask exhibiting the positive pattern of the desired patch array.
  • the wafer is primed with a 20 nm thin titanium layer, followed by a 200 nm thin gold layer both layers deposited using electron-beam evaporation (5 ⁇ /s, Thermionics).
  • the gold patches' can be further chemically modified to achieve the desired bioreactive and biocompatible properties (see Example 3, below).
  • Monolayers based on 11,11′-dithiobis(succinimidylundecanoate) can be deposited on Au(111) surfaces of microdevices described under Examples 1 and 2 by immersing them into a 1 mM solution of DSU in chloroform at room temperature for 1 hour. After rinsing with 10 volumes of solvent, the N-hydroxysuccinimide-terminated monolayer is dried under a stream of nitrogen and immediately used for protein immobilization.
  • Caspases are cysteine proteases of the papain superfamily, with a different active site and catalytic mechanism than observed for papain, Wilson, K P. et al., Nature, 1994 370:270-275. Caspases are important enzymes in the promotion of the cell death pathways and inflammation, Villa, et al., TIBS, 1997, 22:288-392. Identification of selective caspase inhibitors is essential to prevent cross-inhibition of other caspase-dependent pathways. Caspases 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, Villa, et al, TIBS, 1997, 22:288-392 and new caspase homologs identified by the human genome project are PCR amplified and cloned into an E.
  • NTA nickel-nitrilotriacetic acid
  • Caspases are snap-frozen and stored in 20 mM PIPES, pH 7.2, 150 mM NaCl, 0.1% CHAPS, 10% sucrose at ⁇ 80° C.
  • Caspase-fusion proteins can be immobilized to the aminoreactive monolayer surface of the bioreactive patches of the two-dimensional array (see Examples 1, 2, and 4 above). Caspase fusion proteins can be diluted to concentrations of 1 ⁇ g/ml in 20 mM PIPES, pH 7.2, 150 mM NaC, 0.91% CHAPS, 10% sucrose and applied onto the bioreactive patches using a computer-aided, capillary-based dispensing system. After an immobiization period of 30 min, the 2D array was rinsed and subjected to analysis; Ultrapure water with a resistance of 18 M ⁇ cm is generally useable for all aqueous buffers (purified by passage through a Barnstead Nanopure® system).
  • Caspase activity can be determined by a binding assay using three fluorescently labeled peptide aldehyde inhibitors that form a reversible thiohemiacetal moiety with the active site cysteine, Thornberry, Methods in Enzymology, 1994, 244:615-631.
  • the peptides are adapted to caspase 1, 3, 4, 7: Dns (dansyl)-SS-DEVD-CHO, caspase 1: Dns-SS-VDVAD-CHO, caspase 6: Dns-SS-VQID-CHO, Talanian, J. Biol Chem, 1997, 272:9677-9682.
  • the affinity for Ac-DEVD-CHO to caspase 1 is determined to be in the low nanomolar range, Thornberry, Methods in Enzymology, 1994, 244:615-631.
  • the assay buffer is 20 mM PIPES, pH 7.2, 150 mM NaCl, 0.1% CHAPS, 10% sucrose, Stennicke, and Salvesen, J. Biol. Chem, 1997, 272:25719-25723. Fluorescently labeled peptides are mixed to a final concentration of 1 to 5 nM each, the potential drug compound added and flushed onto the 2D array. Peptides are allowed to bind for 10-60 min., unbound peptide removed by washing with buffer and the fluorescence intensity measured (excitation at 360 nm, emission at 470 nm).

Abstract

Protein arrays and protein-coated substrates for the parallel, in vitro screening of biomolecular activity are provided. Methods of using the protein-coated substrates and protein arrays are also disclosed. A plurality of different members of a single protein family may be immobilized on the protein-coated substrate or array. The protein-coated substrates and protein arrays are particularly useful in high-throughput drug screening and clinical diagnostics.

Description

  • This application is a continuation of co-pending application Ser. No. 09/115,455, filed Jul. 14, 1998, which is incorporated herein by reference in its entirety for all purposes and the specific purposes disclosed throughout this application.
  • BACKGROUND OF THE INVENTION
  • A vast number of new drug targets are now being identified using a combination of genomics, bioinformatics, genetics, and high-throughput (HTP) biochemistry. Genomics provides information on the genetic composition and the activity of an organism's genes. Bioinformatics uses computer algorithms to recognize and predict structural patters in DNA and proteins, defining families of related genes and proteins. The information gained from the combination of these approaches is expected to boost the number of drug targets, usually proteins, from the current 500 to over 10,000 in the coming decade.
  • The number of chemical compounds available for screening as potential drugs is also growing dramatically due to recent advances in combinatorial chemistry, the production of large numbers of organic compounds through rapid parallel and automated synthesis. The compounds produced in the combinatorial libraries being generated will far outnumber those compounds being prepared by traditional, manual means, natural product extracts, or those in the historical compound files of large pharmaceutical companies.
  • Both the rapid increase of new drug targets and the availability of vast libraries of chemical compounds creates an enormous demand for new technologies which improve the screening process. Current technological approaches which attempt to address this need include multiwell-plate based screening systems, cell-based screening systems, microfluidics-based screening systems, and screening of soluble targets against solid-phase synthesized drug components.
  • Automated multiwell formats are the best developed high-throughput screening systems. Automated 96-well plate-based screening systems are the most widely used. The current trend in plate based screening systems is to reduce the volume of the reaction wells flier, thereby increasing the density of the wells per plate (96-well to 384-, and 1536-well per plate). The reduction in reaction volumes results in increased throughput, dramatically decreased bioreagent costs, and a decrease in the number of plates which need to be managed by automation.
  • However, although increases in well numbers per plate are desirable for high throughput efficiency, the use of volumes smaller than 1 microliter in the well format generates significant problems with evaporation, dispensing times, protein inactivation, and assay adaptation. Proteins are very sensitive to the physical and chemical properties of the reaction chamber surfaces. Proteins are prone to denaturation at the liquid/solid and liquid/air interfaces. Miniaturization of assays to volumes smaller than 1 microliter increases the surface to volume ratio substantially. (Changing volumes from 1 microliter to 10 nanoliter increases the surface ratio by 460%, leading to increased protein inactivation.) Furthermore, solutions of submicroliter volumes evaporate rapidly, within seconds to a few minutes, when in contact with air. Maintaining microscopic volumes in open systems is therefore very difficult
  • Other types of high-throughput assays, such as miniaturized cell-based assays are also being developed. Miniaturized cell-based assays have the potential to generate screening data of superior quality and accuracy, due to their in vivo nature. However, the interaction of drug compounds with proteins other than the desired targets is a serious problem related to this approach which leads to a high rate of false positive results.
  • Microfluidics-based screening systems that measure in vitro reactions in solution make use of ten to several-hundred micrometer wide channels. Micropumps, electroosmotic flow, integrated valves and mixing devices control liquid movement through the channel network. Microfluidic networks prevent evaporation but, due to the large surface to volume ratio, result in significant protein inactivation. The successful use of microfluidic networks in biomolecule screening remains to be shown.
  • Drug Screening of soluble targets against solid-phase synthesized drug components is intrinsically limited. The surfaces required for solid state organic synthesis are chemically diverse and often cause the inactivation or non-specific binding of proteins, leading to a high rate of false-positive results. Furthermore, the chemical diversity of drug compounds is limited by the combinatorial synthesis approach that is used to generate the compounds at the interface. Another major disadvantage of this approach stems from the limited accessibility of the binding site of the soluble target protein to the immobilized drug candidates.
  • DNA microarray technology is not immediately transferable to protein screening microdevices. To date, microarrays are exclusively available for nucleic acid hybridiaztion assays (‘DNA-chips’). Their underlying chemistry and materials are not readily transferable to protein assays. Nucleic acids withstand temperatures up to 100° C., can be dried and re-hydrated without loss of activity and bound directly to organic adhesion layers absorbed on surfaces such as glass. In contrast, proteins must remain hydrated, kept at ambient-temperatures, and are very sensitive to the physical and chemical properties of the support materials. Therefore, maintaining protein activity at the liquid-solid interface requires entirely different immobilization strategies than those used for nucleic acids. Additionally, the proper orientation of the protein at the interface is desirable to ensure accessibility of their active sites with interacting molecules.
  • In addition to the goal of achieving high-throughput screening of compounds against targets to identify potential drug leads, researchers also need to be able to identify a highly specific lead compound early in the drug discovery process. Analyzing a multitude of members of a protein family or forms of a polymorphic protein in parallel enables quick identification of highly specific lead compounds. Proteins within a structural family share similar binding sites and catalytic mechanisms. Often, a compound that effectively interferes with the activity of one family member also interferes with other members of the same family. Using standard technology to discover such additional interactions requires a tremendous effort in time and costs and as a consequence is simply not done.
  • However, cross-reactivity of a drug with related proteins can be the cause of low efficacy or even side effects in patients. For instance, AZT, a major treatment for AIDS, blocks not only viral polymerases, but also human polymerases, causing deleterious side effects. Cross-reactivity with closely related proteins is also a problem with nonsteroidal anti-inflammatory drugs (NSAIDs) and aspirin. These drugs inhibit cyclooxygenase-2, an enzyme which promotes pain and inflammation. However, the same drugs also strongly inhibit a related enzyme, cyclooxygenase-1, that is responsible for keeping the stomach lining and kidneys healthy, leading to common side-effects including stomach irritation.
  • For the foregoing reasons, there is a need for iminiaturized protein arrays and for methods for the parallel, in vitro, high-throughput screening of functionally and/or structurally related protein targets against potential drug compounds in a manner that minimizes reagent volumes and protein inactivation problems.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to protein arrays, protein-coated substrates, and methods of use thereof that satisfy the need for parallel, in vitro, high-throughput screening of functionally or structurally related protein targets against potential drug compounds in a manner that minimizes reagent volumes and protein inactivation problems.
  • In one embodiment, the present invention provides for a proteinoated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where a protein with a different, known sequence is immobilized on each patch. Furthermore, each of the patches of the protein-coated substrate of the present invention is separated from neighboring patches by from about 50 nm to about 500 μm.
  • Biosensors, micromachined devices, and medical devices that comprise the protein-coated substrate of the present invention represent other aspects of the invention.
  • The present invention also provides an array of proteins comprising a plurality of patches arranged in discrete, known regions on a substrate, where a protein with a different, known sequence is immobilized on each patch. Furthermore, each of the patches of the protein-coated substrate of the present invention is separated from neighboring patches by from about 50 nm to about 500 μm.
  • The protein immobilized on one patch of the array is preferably different from the protein immobilized on a second patch. In an especially preferred embodiment, the protein that is immobilized on one patch of the array is a member of the same protein family as or is otherwise functionally or structurally related to the proteins immobilized on the other patches of the array.
  • The patches of the array may also optionally further comprise monolayers (on which the proteins of the patches are immobilized).
  • At least one coating may be formed on the substrate or applied to the substrate of an array of the present invention such that the coating is positioned between the substrate and the monolayer of each patch
  • The coating, or the substrate itself if no coating is used, may optionally possess an ultraflat surface with a mean roughness of less than about 5 angstroms for areas of at least 25 μm2. This ultraflat surface optionally may be produced by template stripping.
  • The monolayer of a patch on the array of the present invention may be a mixed monolayer composed of more than one type of molecule.
  • The patches of an array of the present invention may further comprise an affinity tag that enhances site-specific immobilization of the biological moiety onto the monolayer.
  • In one embodiment of the invention, an adaptor molecule may also be present to link the affinity tag to the biological moiety on the patches of the array.
  • In another version of the invention, the affinity tag, biological moiety, and the adaptor (if present) preferably constitute a fusion protein.
  • The present-invention further provides for methods of using the array to screen a plurality of proteins in parallel for their ability to bind or otherwise interact with a component of a fluid sample. Most of these methods involve first delivering the fluid sample to the array. If binding is to be detected, the array may then be optionally washed to remove any unbound component from the area. The methods then involve detecting, either directly or indirectly, the presence or absence of the component retained at each patch or other evidence of an interaction of the protein of a given patch with the component.
  • Similar methods may be used diagnostically to screen a fluid sample with the array for the presence, absence, or amount of a plurality of analytes at the same time.
  • The present invention also provides for methods of determining in parallel whether or not a plurality of proteins belongs to a certain protein family. These methods involve delivering a fluid sample comprising a ligand of a known protein family to the patches of the array and then detecting, either directly or indirectly, for the interaction or binding of the known ligand to the patches that would be characteristic of the known protein family.
  • Another aspect of the invention is a protein-coated substrate that comprises a fusion protein immobilized on a monolayer on a portion of the surface of a substrate. The fusion protein is immobilized with the aid of an affinity tag that enhances the site-specific immobilization of the fusion protein onto the monolayer. Here the fusion protein comprises a polypeptide that serves as an adaptor molecule by linking another polypeptide to the affinity tag. The monolayer of the proteincoated substrate comprises molecules of the formula X-R-Y where R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding the fusion protein onto the monolayer. The protein-coated substrate may optionally also include a coating between the substrate and the monolayer and the affinity tag may optionally constitute a part of the fusion protein.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the top view of an array of monolayer-covered patches.
  • FIG. 2 shows the cross section of an individual patch of the array of FIG. 1.
  • FIG. 3 shows the cross section of a row of monolayer-covered patches of the array of FIG. 1.
  • FIG. 4 shows aminoreactive monolayer molecules on a substrate.
  • FIG. 5 shows aminoreactive monolayer molecules on a coated substrate.
  • FIG. 6 shows the immobilization of a protein on a monolayer-coated substrate via an affinity tag.
  • FIG. 7 shows the immobilization of a protein on a monolayer-coated substrate via an affinity tag and an adaptor.
  • FIG. 8 shows four possible expression vectors useful for expressing fusion proteins of the desired protein, an affinity tag, and, optionally, an adaptor molecule.
  • FIG. 9 shows a schematic of a fluorescence detection unit which may be used to monitor interaction of the proteins of the array with an analyte.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A variety of protein arrays, methods, and protein-coated substrates useful for high-throughput drug screening, clinical diagnostics, and related processes are provided by the present invention.
  • (a) Definitions
  • The term “substrate” as used herein refers to the bulk underlying, and core material of the devices or arrays or other embodiments of the invention.
  • The terms “micromachining” and “microfabricating” are both used herein to refer to any number of techniques which are useful in the generation of microstructures (structures of sub-millimeter scale). Such technologies include, but are not limited to, laser ablation, sputtering, electrodeposition, low-pressure vapor deposition, photolithography, and etching. Related technologies such as LIGA are also included Most of these techniques were originally developed for use in semiconductors, microelectronics, and microelectromechanical systems but are applicable to the present invention as well.
  • The term “coating” is used herein to refer to a layer that is either formed on or applied to the surface of the substrate. For instance, exposure of a substrate, such as silicon, to air can result in oxidation of the exposed surface. In the case of a substrate made of silicon, a silicon oxide coating is formed on the surface upon exposure to air. In other instances, the coating is in no way derived from the substrate and may be placed upon the surface via mechanical, electrical or chemical means. An example of this type of coating would be a metal coating that is applied to a polymer substrate. Although a coating may be of any thickness, typically the coating has a thickness smaller than that of the substrate.
  • An “interlayer” is a second coating or layer that is positioned between the first coating and the substrate. The primary purpose of a typical interlayer is to aid adhesion between the first coating and the substrate. One such example is the use of a titanium interlayer to help attach a gold coating to a silicon chip. However, other possible functions of an interlayer are also anticipated. For instance, some interlayers may perform a role in the detection system of the device.
  • The term “affinity tag” is used herein to refer to a functional moiety capable of immobilizing a protein onto the exposed functionality of a monolayer. In some cases, the affinity tag may be a simple chemical functional group. Other possibilities include amino acids, polypeptides, proteins, lipid bilayers, or a hydrogel. The affinity tag may be either covalently or noncovalently attached to the protein (via chemical conjugation or as a fusion protein, for instance). Likewise, the affinity tag may bind to the monolayer either covalently or noncovalently.
  • An “adaptor molecule”, for purposes of this invention, is any entity that links an affinity tag to a protein. The adaptor molecule need not necessarily be a discrete molecule that is noncovalently attached to both the affinity tag and the protein. The adaptor molecule can be covalently attached to the affinity tag or the protein or both. (via chemical conjugation or as a fusion protein, for instance). In some cases, an affinity tag may also be an internal part of the protein, such as an amino acid. Examples of adaptor molecules include polypeptides, proteins, membrane anchors, and biotin.
  • A “monolayer” may be disordered or ordered. One face of the monolayer is composed of chemical functionalities on the termini of the organic molecules that are chemisorbed or physisorbed onto the surface material (headgroups). The other face of the monolayer is exposed and may bear any number of chemical functionalities (end groups). Preferably, the molecules of the monolayer are highly ordered and tightly packed, largely due to hydrophobic and van der Waals interactions between the molecules.
  • An “array”, as used herein, refers to a two-dimensional pattern.
  • The terms “polypeptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. These terms also apply to amino acid polymers in which one or more amino acid residues is an artificial chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. An amino acid polymer in which one or more amino acid residues is an “nataral” amino acid, not corresponding to any naturally occurring amino acid, is also encompassed by the use of the terms “protein” and “polypeptide” herein.
  • Proteins are considered herein to be members of the same “protein family” or to be “related” if they show significant similarities in structure and/or function, as would be recognized by one of ordinary skill in the art, Related proteins can be identified by sequence homology searches of DNA and protein databases using standard bioinformatics resources and software packages (examples of public databases: NCBL NIH, EMBL, SwissProt, Brookhaven database, Washington University—Merck, private databases: Incyte, Hyseq, Human Genome Science; examples of software packages include EMOTIF, Blast, Fasta, Multalign, GCG Wisconsin University). Enzymatically related proteins of non-homologous sequence can be identified by one of ordinary skill in the art by screening the scientific literature (example: Medline database).
  • The term “fusion protein” refers to a protein composed of two or more polypeptides that, although typically unjoined in their native state, are joined by their respective amino and carboxyl termini through a peptide linkage to form a single continuous polypeptide. It is understood that the two or more polypeptide components can either be directly joined or indirectly joined through a peptide linker/spacer.
  • The term “normal physiological condition” is used herein to refer to conditions that are typical inside a living organism or a cell. While it is recognized that some organs or organisms provide extreme conditions, the intra-organismal and intra-cellular environment normally varies around pH 7 (i.e. from pH 6.5 to pH 7.5), contains water as the predominant solvent, and exists at a temperature above 0° C. and below 50° C. It will be recognized that the concentration of various salts depends on the organ, organism, cell, or cellular compartment used as a reference.
  • (b) Protein-Coated Substrates and Arrays
  • In another embodiment, the present invention also provides a protein-coated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where each of the patches comprises an immobilized protein with a different, known sequence and where each of the patches is separated from the neighboring patches by from about 50 nm to about 500 μm. In a preferred embodiment, the protein-coated substrate comprises 9 or more patches.
  • Biosensors, micromachined devices, and medical devices that contain the protein-coated substrate comprising a plurality of patches arranged in discrete, known regions on a substrate, where each of the patches comprises an immobilized protein with a different, known sequence and where each of the patches is separated from neighboring patches by from about 50 nm to about 500 μm are also contemplated
  • Arrays of proteins are also provided by the present invention. In one embodiment, the protein arrays comprise micrometer-scale, two-dimensional patterns of proteins immobilized on arrays of functionalized surface patches.
  • In one embodiment, the array of proteins comprises a plurality of patches, preferably 9 or more, arranged in discrete known regions on a substrate, wherein each of the patches comprises an immobilized protein with a different, known sequence and wherein each of the patches is separated from neighboring patches by from about 50 nm to about 500 μm. In a preferred embodiment, the patches are separated from neighboring patches from about 200 nm to about 500 μm.
  • In some versions of the array, the diameter of each of the patches is proportional to the distance separating the patches. Therefore, the area of each patch may be from about 100 nm2 to about 40,000 μm2. Each patch preferably has an area from about 1 μm2 to about 10,000 μm2.
  • In one embodiment of the array, the array comprises 9 or more patches within a total area of 1 cm2. In preferred embodiments of the array, the array comprises 100 or more patches within a total area of 1 cm2. In another embodiment, the array comprises or more patches within a total area of 1 cm2.
  • In one embodiment of the array, the protein immobilized on one patch differs from the protein immobilized on a second patch of the same array.
  • In another embodiment of the present invention, although the biological moiety of one reactive site is different from that of another, the two biological moieties ate related. In a preferred embodiment, the two different proteins are members of the same protein family. The different proteins on the invention array may be either functionally related or just suspected of being functionally related. In another embodiment of the invention array, however, the function of the immobilized proteins may be unknown. In this case, the different proteins on the different patches of the array share a similarity in structure or sequence or are simply suspected of sharing a similarity in structure or sequence. Alternatively, the immobilized proteins may be just fragments of different members of a protein family.
  • The proteins immobilized on the array of the invention may be members of a protein family such as a receptor family (examples: growth factor receptors, catecholamine receptors, amino acid derivative receptors, cytokine receptors, lectins), ligand family (examples: cytokines, serpins), enzyme family (examples: proteases, kinases, phosphatases, ras-like GTPases, hydrolases), and transcription factors (examples: steroid hormone receptors, heat-shock transcription factors, zinc-finger, leucine-zipper, homeodomain). In one embodiment, the different immobilized proteins are all HIV proteases or hepatitis C virus (HCV) proteases. In an alternative embodiment, the protein immobilized on each patch is a different antibody or antibody fragment (Fab, for example).
  • In an alternative embodiment of the invention array, the proteins on different patches are identical.
  • The substrate of the array may be either organic or inorganic, biological or non-biological or any combination of these materials. In one embodiment, the substrate is transparent or translucent. The portion of the surface of the substrate on which the patches reside is preferably flat and firm or semi-firm. Numerous materials are suitable for use as a substrate in the array embodiment of the invention. For instance, the substrate of the invention array can comprise a material selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titanium dioxide, germanium, silicon nitride, zeolites, and gallium arsenide. Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for substrates of the array. In addition, many ceramics and polymers may also be used as substrates. Polymers which may be used as substrates include, but are not limited to, the following: polystyrene; poly(tetra)fluorethylene; (poly)vinylidenedifluoride; polycarbonate; polymethylmethacrylate; polyvinylethylene; polyethyleneimine; poly(etherether)ketone; polyoxymethylene (POM); polyvinylphenol; polylactides; polymethacrylimide (PMI); polyalkenesulfone (PAS); polyhydroxyethylmethacrylate; polydimethylsiloxane; polyacrylamide; polyimide; co-block-polymers; and Eupergit®. Photoresists, polymerized Langmuir-Blodgett films, and LIGA structures may also serve as substrates in the present invention. The preferred substrates for the array comprise silicon, silica, glass, or a polymer.
  • In a preferred embodiment of the invention array, the patches further comprise a monolayer on the surface of the substrate and the proteins of the patches are unmobilized on the monolayer. The monolayer is preferably a self-assembling monolayer. This monolayer may optionally comprise molecules of the formula X-R-Y, wherein R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding proteins onto the monolayer.
  • A variety of chemical moieties may function as monolayers in the array of the present invention. However, three major classes of monolayer formation are preferably used to expose high densities of bioreactive omega-functionalities on the patches of the arrays (i) alkysiloxane monolayers (“silanes”) on hydroxylated surfaces (as taught in, for, example, U.S. Pat. No. 5,405,766, PCT Publication WO 96/38.726, U.S. Pat. No. 5,412,087, and U.S. Pat. No. 5,688,642); (ii) allyl-thiol/dialkyldisulfide monolayers on noble metals (preferably Au(111)) (as, for example, described in Allara et al., U.S. Pat. No. 4,690,715; Bamdad et al., U.S. Pat. No. 5,620,850, Wagner et al., Biophysical Journal, 1996, 70:2052-2066); and (iii) alkyl monolayer formation on oxide-free passivated silicon (as taught in, for example, Linford et al., J. Am. Chem. Soc., 1995, 117:3145-3155, Wagner et al., Journal of structural Biology, 1997, 119:189-201, U.S. Pat. No. 5,429,708). One of ordinary skill in the art, however, will recognize that many possible moieties may be substituted for X, R, and/or Y, dependent primarily upon the choice of substrate, coating, and affinity tag. Many examples of monolayers are described in Ulman, An Introduction to Ultrathin Organic Films: From Langmuir-Blodgett to Self Assembly, Academic press (1991).
  • An array of the present invention may optionally further comprise a coating between the substrate and the monolayer of its patches. This coating may either be formed on the substrate or applied to the substrate. The substrate can be modified with a coating by using thin-film technology based on either physical vapor deposition (PVD) or plasma-enhanced chemical vapor deposition (PECVD). Alternatively, plasma exposure can be used to directly activate the substrate. For instance, plasma etch procedures can be used to oxidize a polymeric surface (i.e. polystyrene or polyethylene to expose polar functionlities such as hydroxyls, carboxylic acids, aldehydes and the like).
  • The coating may comprise a metal film. Possible metal films include alumninum, chromium, titanium, nickel stainless steel zinc, lead, iron, magnesium, manganese, cadmium, tungsten, cobalt, and alloys or oxides thereof. In a preferred embodiment, the metal film is a noble metal film. Noble metals that may be used for a coating include, but are not limited to, gold, platinum, silver, copper, and palladium. In an especially preferred embodiment, the coating comprises gold or a gold alloy. Electron-beam evaporation may be used to provide a thin coating of gold on the surface. In a preferred embodiment, the metal film is from about 50 nm to about 500 nm in thickness.
  • In alternative embodiments, the coating comprises a composition selected from the group consisting of silicon, silicon oxide, silicon nitride, silicon hydride, indium tin oxide, magnesium oxide, alumina, glass, hydroxylated surfaces, and a polymer.
  • If the patches comprise a coating between the substrate and the monolayer, then it is understood that the coating must be composed of a material for which a suitable functional group X is available (see below). If no such coating is present, then it is understood that the substrate must be composed of a material for which a suitable functional group X is available.
  • In one embodiment of the invention array, the surface of the coating is ultraflat In this embodiment, the mean roughness of the surface of the coating is less than about 5 angstroms for areas of at least 25 μm2. In a preferred embodiment, the mean roughness of the surface of the coating is less than about 3 angstroms for areas of at least 25 m The ultraflat coating can optionally be a template-stripped surface as described in Hegner et al., Surface Science, 1993, 291:3946 and Wagner et al., Langmuir, 1995, 11:3867-3875, both of which are incorporated herein by reference.
  • It is contemplated that the coatings of many arrays will require the addition of at least one adhesion layer between said coating and said substrate. Typically, the adhesion layer rill be at least 10 angstroms thick and may be much thicker. For instance, a layer of titanium may be desirable between a silicon wafer and a goldcoating. In an alternative embodiment, an epoxy glue such as Epo-tek 377®, Epo-tek 301-2®), (Epoxy Technology Inc., Billerica, Mass.) may be preferred to aid adherence of the coating to the substrate. Determinations as to what material should be used for the adhesion layer would be obvious to one skilled in the art once materials are chosen for both the substrate and coating. In other embodiments, additional adhesion mediators or interlayers may be necessary to improve the optical properties of the array, for instance, in waveguides for detection purposes.
  • Deposition or formation of the coating (if present) on the substrate is done prior to the formation of patches of bioreactive monolayers thereon. Monolayer-compatible surface patches may optionally be fabricated by photolithography, micromolding (PCT Publication WO 96/29629), wet chemical etching, or any combination of these. Bio-reactive monolayers are then formed on the patches. Alternatively, arrays of bioreactive-monolayer functionalized surface patches can be created by microstamping (U.S. Pat. Nos. 5,512,131 and 5,731,152) or microcontact printing (LCP) (PCT Publication WO 96/29629). Subsequent immobilization of biomolecules results in two-dimensional protein arrays. Inkjet chemical dispensers provide another option for patterning monolayer X-R-Y molecule or components thereof to nanometer or micrometer scale sites on the surface of the substrate or coating (Lemmo et al., Anal Chem., 1997, 69:543-'551).
  • Diffusion boundaries between the patches may be integrated as topographic patterns or surface functionalities with orthogonal wetting behavior. For instance, walls of substrate material or photoresist may be used to separate some of the patches from some of the others or all of the patches from each other. In a preferred embodiment, the patches are separated from each other by surfaces free of monolayers of the form X-R-Y. Alternatively, non-bioreactive monolayers with different wettability may be used to separate patches from one another.
  • FIG. 1 shows the top view of one example of a monolayer-covered array. On the array, a number of patches 15 cover the surface of the substrate 3.
  • FIG. 2 shows a detailed cross section of a patch of the array of FIG. 1. This view illustrates the use of a coating 5 on the substrate 3. An adhesion interlayer 6 is also included in the patch. On top of the patch resides a monolayer 7.
  • FIG. 3 shows a cross section of one row of the monolayer-covered patches 15 of the array of FIG. 1. This figure also shows the use of a cover 2 over the array. Use of the cover 2 creates an inlet port 16 and an outlet port 17 for solutions to be passed over the array.
  • If the patches of the invention array comprise a monolayer of molecules of the formula X-R-Y, then R may comprise a hydrocarbon chain from about 1 to about 200 carbons long. The hydrocarbon chain may comprise an alkyl aryl, alkenyl alkynyl, cycloalkyl, alkaryl aralkyl group, or any combination thereof. In a preferred embodiment, R is an alkyl chain from about 8 to about 22 carbons long and is optionally a straight alkane. However, it is also contemplated that in an alternative-embodiment, R may readily comprise a hydrocarbon chain from about 2 to about 200 carbons long and be interrupted by at least one hetero atom. The interrupting hetero groups can include —O—, —CONH—, —CONHCO—, —NH—, —CSNH—, —CO—, —CS—, —S—, —SO—, —(OCH2CH2)n— (where n=1-20), —(CF2)n— (where n=1-22), and the like. Alternatively, one or more of the hydrogen moieties of R can be substituted with deuterium.
  • X may be chosen as any group which affords chemisorption or physisorption of the monolayer onto the surface of the substrate (or the coating, if present). When the substrate or coating is a metal or metal alloy, X, at least prior to incorporation into the monolayer, can in one embodiment be chosen to be an asymmetrical or symmetrical disulfide, sulfide, diselenide, selenide, thiol, isonitrile, selenol a trivalent phosphorus compound, isothiocyanate, isocyanate, xanthanate, thiocarbamate, a phosphine, an amine, thio acid or a dithio acid. This embodiment is especially preferred when a coating or substrate is used that is a noble metal such as gold, silver, or platinum.
  • If the substrate of the array is a material such as silicon, silicon oxide, indium tin oxide, magnesium oxide, alumina, quartz, glass, or silica, then a preferred embodiment of the invention array comprises an X that, prior to incorporation into said monolayer, is a monohalosilane, dihalosilane, trihalosilane, trialkoxysilane, dialkoxysilane, or a monoalkoxysilane. In preferred embodiments, X is a trichlorosilane or trialkoxysilane.
  • In other embodiments, the surface of the substrate (or coating thereon) is composed of a metal oxide such as titanium oxide, tantalum oxide, indium tin oxide, magnesium oxide, or alumina and X is a carboxylic acid. Alternatively, if the surface of the substrate (or coating thereon) of the device is copper, then X may optionally be a hydroxamic acid.
  • If the substrate used in the invention is a polymer, then in many cases a coating on the substrate such as a copper coating will be included in the device. An appropriate functional group X for the coating would then be chosen for use in the device. In an alternative embodiment comprising a polymer substrate, the surface of the polymer may be plasma-modified to expose desirable surface functionalities for monolayer formation. For instance, EP 780423 describes the use of a monolayer molecule that has an alkene X functionality on a plasma exposed surface. Still another possibility for the invention device comprised of a polymer is that the surface of the polymer on which the monolayer is formed is functionalized due to copolymerization of appropriately functionalized precursor molecules.
  • Another possibility is that prior to incorporation into the monolayer, X can be a free-radical-producing moiety. This functional group is especially appropriate when the surface on which the monolayer is formed is a hydrogenated silicon surface. Possible free-radical producing moieties include, but are not limited to, diacylperoxides, peroxides, and azo compounds. Alternatively, unsaturated moieties such as unsubstituted alkenes, alkynes, cyano compounds and isonitrile compounds can be used for X, if the reaction with X is accompanied by ultraviolet, infrared, visible, or microwave radiation.
  • In alternative embodiments, X, prior to incorporation into the monolayer, may be a hydroxyl, carboxyl vinyl sulfonyl, phosphoryl silicon hydride, or an amino group.
  • The component, Y, of the monolayer is a functional group responsible for binding a protein onto the monolayer. In a preferred embodiment of the invention, the Y group is either highly reactive (activated) towards the protein or is easily converted into such an activated form. In a preferred embodiment, the coupling of Y with the protein occurs readily under normal physiological conditions not detrimental to the biological activity of the protein. The functional group Y may either form a covalent linkage or a noncovalent linkage with the protein (or its affinity tag, if present). In a preferred embodiment, the functional group Y forms a covalent linkage with the protein or its affinity tag. It is understood that following the attachment of the protein to Y, the chemical nature of Y may have changed.
  • In one embodiment of the array of the present invention, Y is a functional group that is activated in situ. Possibilities for this type of functional group include, but are not limited to, such simple moieties such as a hydroxyl, carboxylamino, aldehyde, carbonyl, methyl, methylene, alkene, alkyne, carbonate, aryliodide, or a vinyl group. Appropriate modes of activation would be obvious to one skilled in the art Alternatively, Y can comprise a functional group that requires photoactivation prior to becoming activated enough to trap the biological moiety.
  • In an especially preferred embodiment of the array of the present invention, Y is a complex and highly reactive functional moiety that is compatible with monolayer formation and needs no in situ activation prior to reaction with the protein and/or affinity tag. Such possibilities for Y include, but are not limited to, maleimide, N-hydroxysuccimmide (Wagner et al., Biophysical Journal, 1996, 70:2052-2066), nitrilotriacetic acid (U.S. Pat. No. 5,620,850), activated hydroxyl, haloacetyl, bromoacetyt, iodoacetyl activated carboxyl, hydrazide, epoxy, aziridine, trifluoromethyldiaziridine, pyridyidisulfide, N-acyl-imidazole, imidazolecarbamate, succmimidylcarbonate, arylazide, anhydride, diazoacetate, benzophenone, isothiocyanate, isocyanate, imidoester, fluorobenzene, and biotin
  • FIG. 4 shows one example of a monolayer on a substrate 3. In this example, substrate 3 comprises silicon (having a silicon oxide surface). The monolayer is aminoreactive because it bears a functional group Y that is N-hydroxysuccimmide.
  • FIG. 5 shows another example of a monolayer on a substrate 3. In this-case, however, a thin film coating 5 comprised of gold covers the surface of the substrate 3. Also, in this embodiment, an adhesion interlayer 6 is used to adhere the coating 5 to the substrate 3 and is comprised of titanium. This monolayer is also aminoreactive because it bears a functional group Y that is N-hydroxysuccinimide.
  • In an alternative embodiment, the functional group Y of the array is selected from the group of simple functional moieties. Possible Y functional groups include, but are not limited to, —OH, —NH2, —COOH, —COOR, —RSR, —PO4 −3, OSO3 −2, —SO3 , —COO, —SOO, —CONR2, —CN, —NR2, and the like. Simple groups such as these are only preferred for Y when the affinity tag of the invention composes a layer of affinity tag molecules (such as poly-lysine) that coats the exposed portion of the monolayer prior to immobilization of the biological moiety (see below).
  • The monolayer molecules of the present invention can optionally be assembled on the surface in parts. In other words, the monolayer need not necessarily be constructed by chemisorption or physisorption of molecules of the formula X-R-Y to the surface of the substrate (or coating). Instead, in one embodiment, X may be chemisorbed or physisorbed to the surface of the substrate (or coating) alone first Then, R or even just individual components of R can be attached to X through a suitable chemical reaction. Upon completion of addition of the spacer R to the X moiety already immobilized on the surface, Y can be attached to the ends of the monolayer molecule through a suitable covalent linkage.
  • Not all monolayer molecules on a given patch need be identical to one another. Some may consist of mixed monolayers. For instance, the monolayer of an individual patch may optionally comprise at least two different X-R-Y molecules. This second X-R-Y molecule may immobilize the same or a different protein from the first In addition, some of the monolayer molecules X-R-Y of a patch may have failed to attach any protein.
  • As another alternative of the invention, the monolayer of an individual patch on the array may comprise a second molecule that is of the formula, X-R-V where R is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins. V must also be biocompatible with proteins. Typically, V will consist of a hydroxyl, saccharide, or polyethylene glycol moiety (EP Publication 780423).
  • As a still further alternative of the invention array, the array may further comprise at least one unreactive patch devoid of protein, wherein said patch comprises a monolayer of molecules of the formula X-R-V, where R is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins.
  • Regardless of the nature of the monolayer molecules, in some arrays it may be desirable to provide crosslinking between molecules of an individual patch's monolayer. In general, crosslinking confers additional stability to the monolayer. Such methods are familiar to those skilled in the art (for instance, see Ulman, An Introduction to Ultrathin Organic Films: From Langmuir-Blodgett to Self-Assembly, Academic Press (1991)).
  • After completion of the monolayer, the protein may be attached to the monolayer via interaction with the Y-functional group. Y-functional groups which fail to react with any protein molecules are preferably quenched prior to use of the array.
  • In a preferred embodiment, the array further comprises an affinity tag that enhances site-specific immobilization of the biological moiety onto the monolayer. The use of an affinity tag on the proteins of the array allows for at least one of two advantages. An affinity tag can confer enhanced binding or reaction of the protein with Y. This enhancement effect may be either kinetic or thermodynamic. The affinity tag/Y-functional group pair used in the patches of the array preferably allows for immobilization of the biological molecules in a manner which does not require harsh reaction conditions that are adverse to protein stability or function. In most embodiments, immobilization in aqueous, biological buffers are ideal. An affinity tag also preferably offers immobilization that is specific to a designated site or location on the biological moiety. For this to occur, attachment of the affinity tag to the protein must be site-specific. This site specific immobilization helps ensure that the reactive site of the protein remains accessible to ligands in solution. Another advantage of immobilization through affinity tags is that it allows for a common immobilization strategy to be used with multiple, different proteins.
  • In a preferred embodiment, the affinity tag comprises at least one amino acid. The affinity tag may be a polypeptide comprising at least one monolayer-reactive amino acid. Alternatively, the affinity tag may be a lone, monolayer-reactive amino acid. Examples of possible monolayer-reactive amino acids include cysteine; lysine, histidine, arginine, tyrosine, and glutamine. A polypeptide or amino acid affinity tag is preferably expressed as a fusion protein with the biological moiety. Amino acid tags provide either a single amino acid or a series of amino acids that can interact with the Y-functional group of the monolayer molecules. Amino acid affinity tags can be readily introduced into recombinant proteins to facilitate oriented immobilization by covalent binding to the bioreactive Y-functional group of the monolayer.
  • The affinity tag may comprise a poly(amino acid) tag. A poly(amino acid) tag is a polypeptide that comprises from about 2 to about 100 residues of a single amino acid, optionally interrupted by residues of other amino acids. For instance, the affinity tag may comprise a poly-cysteine, polylysine, poly-arginine, or poly-histidine. Amino acid tags are preferably composed of two to twenty residues of a single amino acid, such as, for example, histidines, lysines, arginines, cysteines, glutanunes, tyrosines, or any combination of these. According to a preferred embodiment, an amino acid tag of one to twenty amino acids includes at least one to ten cysteines for thioether linkage; or one to ten lysines for amide linkage; or one to ten arginines for coupling to vicinal dicarbonyl groups. One of ordinary skill in the art can readily pair suitable affinity tags with a given Y-functionality.
  • The position of the amino acid tag can be at the amino-, or carboxy-terminus of the protein or anywhere in-between. Where compatible with protein function, affinity tags introduced for protein purification are preferentially located at the C-terminus of the recombinant protein to ensure that only full-length proteins are isolated during protein purification.
  • Affinity tags may also contain one or more unnatural amino acids. Unnatural amino acids can be introduced using suppressor tRNAs that recognize stop codons (i.e. amber) (Noten et al., Science, 1989, 244:182-188; Ellman et al., Methods Enzym, 1991, 202:301-336; Cload et al., Chem Biol., 1996, 3:1033-1038). The tRNAs are chemically amino-acylated to contain chemically altered (“unnatural”) amino acids for use with specific coupling chemistries (i.e. ketone modifications, photoreactive groups).
  • In an alternative embodiment the affinity tag can comprise a whole protein, such as, but not limited to, glutathione S-transferase, an antibody, avidin, or streptavidin.
  • Other protein conjugation and immobilization techniques known in the art may be adapted for the purpose of immobilizing proteins on activated monolayers. For instance, in an alternative embodiment of the array, the affinity tag may be an organic bioconjugate which is chemically coupled to the protein of interest. Biotin or antigens may be chemically cross linked to the protein. Alternatively, a chemical cross linker may be used that attaches a simple functional moiety such as a thiol or an amine to the surface of a protein.
  • FIG. 6 shows a detailed cross section of a patch on one embodiment of the invention array. In this embodiment, a protein 10 is immobilized on a monolayer 7 on a substrate 3. An affinity tag 8 connects the biological moiety 10 to the monolayer 7. The monolayer 7 is formed on a coating 5 which is separated from the substrate 3 by an interlayer 6.
  • In an alternative embodiment of the array invention, the affinity tag is a component of a layer of affinity tag molecules immobilized on the monolayer of an individual patch. For instance, a hydrogel composed of a material such as dextran can serve as a suitable layer of affinity tag molecules in the array. Use of such hydrogels to immobilize biological moieties is described in U.S. Pat. No. 5,242,828. Poly-lysine is another option for a material useful in forming an affinity-tag layer (for an example see U.S. Pat. No. 5,629,213). The layer of affinity tag molecules could also constitute a phospholipid bilayer or a phospholipid monolayer as described in PCT Publication WO 96/38726. Use of a phospholipid monolayer or bilayer as an affinity tag would be suitable if the biological moiety to be immobilized is a membrane protein, such as an ion channel protein.
  • If the proteins of different patches on the array are different, then different solutions, each containing an affinity-tagged protein, must be delivered to their individual patches. Protein solutions may be transferred to the appropriate patches via noncontact printing using a microdelivery device employing a ball-point pen type of mechanism. Alternatively, microcapirary-based dispensing systems may be used. These dispensing systems are preferably computer-aided. The use of other microprinting techniques for transferring protein solutions to the bioreactive patches is also possible.
  • Another major embodiment of the arrays of the present invention comprises an adaptor molecule that links the affinity tag to the immobilized biological moiety. The additional spacing of the protein from the surface of the substrate (or coating) that is afforded by the use of an adaptor molecule is particularly advantageous since proteins are known to be prone to surface inactivation. One of ordinary skill in the art will be able to choose an adaptor molecule which is appropriate for a given affinity tag. For instance, if the affinity tag is streptavidin, then the adaptor could be a biotin molecule that is chemically conjugated to the protein which is to be immobilized. Alternatively, if the affinity tag is a phospholipid biolayer or monolayer then a membrane anchor could be chosen as a suitable adaptor molecule.
  • In a preferred embodiment, the adaptor molecule is a polypeptide, such as protein G or protein A In a preferred embodiment, the affinity tag, adaptor molecule, and biological moiety together compose a fusion protein Such a fusion protein may be readily expressed using standard recombinant DNA technology. Adaptor proteins are especially useful to increase the solubility of the protein of interest and to increase the distance between the surface of the substrate or coating and the protein of interest. Use of an adaptor protein or polypeptide can also be very useful in facilitating the preparative steps of protein purification by affinity binding prior to immobilization on the array. Examples of possible adaptor proteins include glutathione-S-transferase (GST), maltose-binding protein, chitin-binding protein, thioredoxin, green-fluorescent protein (GFP). GFP can also be used for quantification of surface binding.
  • FIG. 7 shows a cross section of a patch on one particular embodiment of the invention array. The patch comprises a protein 10 immobilized on a monolayer 7 via both an affinity tag 8 and an adaptor molecule 9. The monolayer 7 rests on a coating 5. An interlayer 6 is used between the coating 5 and the substrate 3.
  • In preparation for immobilization to the devices and arrays of the present invention, proteins can be expressed from recombinant. DNA either in vivo or in vitro. Amino acid affinity tags are introduced by polymerase chain reaction. Expression in vivo is in either bacteria (Escherichia coli), lower eukaryotes (Saccharomyces cerevisiae, Saccharomyces pombe, Pichia pastoris) or higher eukaryotes (bacculo-infected insect cells, insect cells mammalian cells), or in vitro (Escherichia coli lysates, wheat germ extracts, reticulocyte lysates). Proteins are purified by affinity chromatography using commercially available resins.
  • DNA sequences encoding amino acid affinity tags and adaptor proteins are engineered into the expression vectors such that the genes of interest can be cloned in frame either 5′ or 3′ of the DNA sequence encoding the affinity tag and adaptor protein.
  • FIG. 8 shows four possible expression vectors for expressing the protein of interest, a polypeptide affinity tag, and a polypeptide adaptor molecule as a fusion protein. The vector contains an origin of replication sequence 35 and a gene 36 capable of conferring antibiotic resistance to a host cell. The insert of the vector contains a promoter sequence 30 and a termination signal sequence 34. Between the sequences 30 and 34, the insert also contains a gene 33 encoding the protein of interest and sequence 31 encoding the polypeptide affinity tag. Sequence 32 which codes for a polypeptide adaptor molecule may also be included on the plasmid and is positioned between the protein and affinity-tag coding regions (33 and 31, respectively).
  • Preferably, production of families of related proteins involves parallel processing from cloning to protein expression and protein purification cDNAs for the protein of interest will be amplified by PCR using cDNA libraries or EST (expressed sequence tag) clones as templates. For in vivo expression of the proteins, cDNAs can be cloned into commercial expression vectors (Qiagen, Novagen, Clontech) and introduced into the appropriate organism for expression (organisms include. Escherichia coli, Saccharomyces cerevisiae, Saccharomyces pombe, Pichiapastoris, bacculovirus/insect cells, insect cells, mammalian cells). For in vitro expression PCR-amplified DNA sequences are directly used in coupled in vitro transcription/translation systems (Escherichia coil S30 lysates from T7 RNA polymerase expressing, preferably protease-deficient strains, wheat germ lysates, reticulocyte lysates with and without microsomes (Promega, Pharmacia, Panvera)). The choice of organism for optimal expression depends on the extent of post-translational modifications (i.e. glycosylation, lipid-modifications).
  • Escherichia coli based protein expression will be the method of choice for soluble proteins that do not require extensive post-translational modifications for activity. Extracellular or intracellular domains of membrane proteins will be fused to protein adaptors for expression and purification.
  • The entire approach can be performed using 96-well assay plates. PCR reactions are carried out under standard conditions. Oligonucleotide primers contain unique restriction sites for facile cloning into the expression vectors. Alternatively, the TA cloning system (Clontech) can be used. Expression vectors contain the sequences for affinity tags and the protein adaptors. PCR products are ligated into the expression vectors (under inducible promoters) and introduced into the appropriate competent Escherichia coli strain by calcium-dependent transformation (strains include: XL-1 blue, BL21, SG13009(Ion-)). Transformed Escherichia coli cells are plated and individual colonies transferred into 96-array blocks. Cultures are grown to mid-log phase, induced for expression, and cells collected by centrifugation Cells are resuspended containing lysozyme and the membranes broken by rapid freeze/thaw cycles, or by sonication. Cell debris is removed by centrifugation and the supernatants transferred to 96-tube arrays. The appropriate affinity matrix is added, protein of interest bound and nonspecifically bound proteins removed by repeated washing steps using 12-96 pin suction devices and centrifugation. Alternatively, magnetic affinity beads and filtration devices can be used (Qiagen). The proteins are eluted and transferred to a new 96-well array. Protein concentrations are determined and an aliquot of each protein is spotted onto a nitrocellulose filter and verified by Western analysis using an antibody directed against the affinity tag. The purity of each sample is assessed by SDS-PAGE and Coomassie staining or mass spectroscopy. Proteins are snap-frozen and stored at −80° C.
  • Saccharomyces cerevisiae allows for core glycosylation and lipid modifications of proteins. The approach described above for Escherichia coli can be used with slight modifications for transformation and cell lysis. Transformation of Saccharomyces cerevisiae is by lithium-acetate and cell lysis is either by lyticase digestion of the cell walls followed by freeze-thaw, sonication or glass-bead extraction. Variations of post-translational modifications can be obtained by different yeast strains (i.e. Saccharomyces pombe, Pichia pastoris).
  • The advantage of the bacculovirus system or mammalian cells are the wealth of post-translational modifications that can be obtained. The baeculo-system requires cloning of viruses, obtaining high titer stocks and infection of liquid insect cell suspensions (cells are SF9, SF21). Mammalian cell-based expression requires transfection and cloning of cell lines. Soluble proteins are collected from the medium while intracellular or membrane bound proteins require cell lysis (either detergent solubilization, freeze-thaw). Proteins can then be purified analogous to the procedure described for Escherichia coli.
  • For in vitro translation the system of choice is Escherichia coli lysates obtained from protease-deficient and 17 RNA polymerase overexpressing strains. Escherichia coli lysates provide efficient protein expression (30-50 μg/ml lysate). The entire process is carried out in 96-well arrays. Genes of interest are amplified by PCR using oligonucleotides that contain the gene-specific sequences containing a 17 RNA polymerase promoter and binding site and a sequence encoding the affinity tag. Alternatively, an adaptor protein can be fused to the gene of interest by PCR. Amplified DNAs can be directly transcribed and translated in the Escherichia coli lysates without prior cloning for fast analysis. The proteins are then isolated by binding to an affinity matrix and processed as described above.
  • Alternative systems which maybe used include wheat germ extracts and reticulocyte extracts. In vitro synthesis of membrane proteins and or post-translationally modified proteins will require reticulocyte lysates in combination with microsomes.
  • The present invention also provides for methods of using the invention array. The arrays of the present invention are particularly suited for the use in high-throughput drug screening. Other uses include medical diagnostics and biosensors.
  • Use of one of the protein arrays of the present invention may optionally involve placing the two-dimensional protein array in a flowchamber with approximately 1-10 microliters of fluid volume per 25 mm2 overall surface. The cover over the array in the flowchamber is preferably transparent or translucent. In one embodiment, the cover may comprise Pyrex or quartz glass. In other embodiments, the cover may be part of a detection system that monitors interaction between biological moieties immobilized on the array and an analyte. The flowchambers should remain filled with appropriate aqueous solutions to preserve protein activity. Substrates and potential drug compounds may be flushed into the flow chamber as desired and their interaction with the immobilized proteins determined. Hence, no specialized microfluidic pumps, valves, or mixing techniques are required for fluid delivery to the array.
  • In an alternative embodiment, fluid can be delivered to each of the patches of the array individually. For instance, in one embodiment, the regions of the substrate surface may be microfabricated in such a way as to allow integration of the array with a number of fluid delivery channels oriented perpendicular to the array surface, each one of the delivery channels terminating at the site of an individual proteincoated patch.
  • Possible interactions towards which the present invention may be directed include, but are not limited to, antibody/antigen, antibody/hapten, enzyme/substrate, carrier protein/substrate, lectin/carbohydrate, receptor/hormone, receptor/effector, protein/DNA, protein/RNA, repressor/inducer, or the like.
  • A method for screening a plurality of proteins for their ability to interact with a component of a fluid sample comprises delivering the fluid sample to the invention array, and detecting the interaction of said component with the immobilized protein of each patch.
  • A wide range of detection methods is applicable to this and other methods of the invention. The invention device can be interfaced with optical detection methods such as absorption in the visible range, chemoluminescence, and fluorescence (including lifetime, polarization, fluorescence correlation spectroscopy (FCS), and fluorescence-resonance energy transfer (FRET)). Furthermore, built-in detectors such as optical waveguides PCT Publication WO 96/26432 and U.S. Pat. No. 5,677,196, surface plasmons, and surface charge sensors are compatible with many embodiments of thee invention.
  • FIG. 9 shows a schematic diagram of one type of fluorescence detection unit which may be used to monitor interaction of immobilized proteins of an array with an analyte. In the illustrated detection unit, the protein array 21 is positioned on a base plate 20. Light from a 100 W mercury arc lamp 25 is directed through an excitation filter 24 and onto a beam splitter 23. The light is then directed through a lens 22, such as a Micro Nikkor 55 mm 1:2:8 lens, and onto the array 21. Fluorescence emission from the array retuns through the lens 22 and the beam splitter 23. After next passing through an emission filter 26, the emission is received by a cooled CCD camera 27, such as the Slowscan TE/CCD-1024SF&SB (Princeton Instruments). The camera is operably connected to a CPU 28 which is in turn operably connected to a VCR/monitor 29.
  • Another aspect of the invention provides for a method for screening a plurality of proteins for their ability to bind a component of a fluid sample. This method comprises, delivering said fluid sample to the invention array, washing the array with fluid which does not contain said component in order to elute unbound component from the surface of the array, and detecting, either directly or indirectly, the presence, absence, or amount of the component retained at each patch.
  • Another embodiment of the invention provides a method for detecting in parallel the ability of a drug candidate to interfere with the ability of a singular analyte or a plurality of analytes in a fluid sample to bind one or more immobilized proteins on the invention array. This method comprises delivering the fluid sample to the invention array, washing said array with analyte free fluid to remove unbound analyte, and detecting, either directly or indirectly, the presence of analyte retained at each patch.
  • The array of the present invention may also be used in a diagnostic manner. In the diagnostic embodiments of the invention, the plurality of proteins immobilized on the array are not preferably members of the same protein family. One embodiment of the invention provides a method for detecting in parallel the presence of a plurality of analytes in a fluid sample which react with one or more of the immobilized proteins. This method comprises delivering the fluid sample to the invention array and detecting the interaction of the analyte with the immobilized protein at each patch.
  • A method for detecting in parallel the presence of a plurality of analytes in a fluid sample which bind said one or more of the immobilized proteins, comprises delivering the fluid sample to the invention array, washing said array with an analyte-free fluid to remove unbound analyte, and detecting, either directly or indirectly, the presence of analyte retained at each patch.
  • (c) Alternative Embodiments of Protein-Coated Substrates.
  • Another embodiment of the present invention is a protein-coated substrate comprising the following a substrate; a monolayer on a portion of a surface of the substrate that comprises molecules of the formula X-R-Y where R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding a fusion protein onto the monolayer; an affinity tag that enhances site-specific immobilization of the fusion protein onto the monolayer; and a fusion protein of a first polypeptide linked to a second, adaptor polypeptide, immobilized on said monolayer by means of the affinity tag that is linked to the second, adaptor polypeptide.
  • One of ordinary skill in the art will appreciate that numerous materials and compounds may be selected for the elements of the substrate, R, Y, X the affinity tag, and the adaptor polypeptide of this embodiment
  • The substrate may be organic or inorganic, biological or non-biological, or any combination of these materials. The substrate of the invention can optionally comprise a material selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titanium dioxide, germanium, silicon nitride, zeolites, and gallium arsenide. Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for substrates. In addition, many ceramics and polymers may be used as substrates. Photoresists, polymerized Langmuir-Blodgett films, and LIGA structures may also serve as substrates in the present invention. The preferred substrates of the present invention comprise sililcon, silica, glass, or a polymer.
  • In an alternative embodiment, the protein-coated substrate further comprises a coating between said substrate and said monolayer. This coating may be formed on the substrate or applied to the substrate. The coating on the substrate may comprise a metal film. In a preferred embodiment, the coating is a noble metal film. Many other different coatings, such as a polymer, glass, a hydroxylated surface, silicon nitride and silicon oxide are also compatible with the present invention.
  • In some embodiments of the invention, the protein-coated substrate will further comprise at least one adhesion layer or mediator between the coating and the substrate.
  • A variety of chemical moieties may function as monolayers in the array of the present invention. However, three major classes of monolayer formation are preferably used to expose high densities of bioreactive omega-functionalities on the patches of the array: (i) alkylsiloxane monolayers. (“silanes”) on hydroxylated surfaces (as taught in, for example, U.S. Pat. No. 5,405,766, PCT Publication WO 96/38726, U.S. Pat. No. 5,412,087, and U.S. Pat. No. 5,688,642); (ii) alkyl-thiol/dialkyldisulfide monolayers on noble metals (preferably Au(111)) (as, for example, described in Allara et al., U.S. Pat. No. 4,690,715; Bamdad et al., U.S. Pat. No. 5,620,850; Wagner et al., Biophysical Journal, 1996, 70:2052-2066); and (iii) ally monolayer formation on oxide-free passivated silicon (as taught in, for example, Linford et al., J. Am Chem Soc., 1995, 117:3145-3155, Wagner et al., Journal of Structural Biology, 1997, 119:189-201, U.S. Pat. No. 5,429,7083. One of ordinary skill in the art, however, will recognize that many possible moieties may be substituted for X, R, and/or Y, dependent primarily upon the choice of substrate, coating and affinity tag.
  • R of a monolayer molecule may comprise a hydrocarbon chain from about 1 to about 200 carbons long. The hydrocarbon chain may comprise an alkyl, aryl, alkenyl alkynyl, cycloalkyl, alkaryl, aralkyl group, or any combination thereof. In a preferred embodiment, R is an alkane from about 8 to about 22 carbons long. However, it is also contemplated that in an alternative embodiment, R may readily comprise a hydrocarbon chain from about 2 to about 200 carbons long and interrupted by at least one hetero atom.
  • X may be chosen as any group which affords chemisorption or physisorption of the monolayer onto the surface of the substrate (or the coating, if present). For instance, especially if the substrate or coating is a metal or metal alloy, X, at least prior to incorporation into the monolayer, may be chosen to be an asymmetrical or symmetrical disulfide, sulfide, diselenide, selenide, thiol, isonitrile, selenol trivalent phosphorus compounds, isothiocyanate, isocyanate, xanthanate, thiocarbamate, phosphines, amines, thio acid and dithio acid. This embodiment is especially preferred when a coating or substrate is used that is a noble metal such as gold, silver, or platinum.
  • In alternative embodiments, X prior to incorporation into the monolayer, may be a hydroxyl, carboxyl vinyl sulfonyl phosphoryl silicon hydride, or an amino group. Another possibility is that prior to incorporation into the monolayer, X can be a free-radical-producing moiety.
  • If the substrate of the protein-coated substrate is a material such as silicon, silicon oxide, indium tin oxide, magnesium oxide, alumina, quartz, glass, or silica, then a preferred embodiment of the invention comprises an X that, prior to incorporation into said monolayer, is a monohalosilane, dihalosilane, trihalosilane, trialkoxysilane, dialkoxysilane, or a monoalkoxysilane. In preferred embodiments, the silane is a trichlorosilane or trialkoxysilane.
  • The component, Y, of the monolayer is responsible for binding a biological moiety onto the monolayer. In a preferred embodiment of the invention, the Y group is either highly reactive (activated) towards the biological moiety or is easily converted into such an activated form. In a preferred embodiment, the coupling of Y with the biological moiety occurs readily under physiological conditions.
  • In one embodiment of the present invention, Y is a functional group that is activated in situ. Possibilities for this type of functional group include, but are not limited to; such simple moieties such as a hydroxyl, carboxyl, amino, aldehyde, carbonyl methyl methylene, alkene, alkyne, carbonate, aryliodide, or a vinyl group. Alternatively, Y can comprise a functional group that requires photoactivation prior to becoming activated enough to trap the biological moiety.
  • In an especially preferred embodiment of the protein-coated substrate of the present invention, Y is a highly reactive functional moiety. Such possibilities for Y include, but are not limited to, maleimide, N-hydroxysuccinimide, nitrilotriacetic acid, activated hydroxyl haloacetyl bromoacetyl, iodoacetyl activated carboxyl, hydrazide, epoxy, aziridine, trifluoromethyldiaziridine, pyridyldisulfide, N-acyl-imidazole, imidazolecarbamate, succinimidylcarbonate, arylazide, anhydride, diazoacetate, benzophenone, isothiocyanate, isocyanate, imidoester, fluorobenzene, and biotin
  • The monolayer of the protein-coated substrate can optionally comprise at least two different X-R-Y molecules. Alternatively, the monolayer can further comprise a second molecule that is of the formula X-R-V wherein k is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of polypeptides and proteins.
  • In one embodiment of the protein-coated substrate, the affinity tag is a polypeptide or an amino acid. In a preferred embodiment of the protein-coated substrate, the affinity tag comprises a poly(amino acid). Amino acid tags provide either a single amino acid or a series of amino acids that can interact with the Y-functionalities of the monolayer. Amino acid affinity tags can also be introduced to a specific site on a recombinant protein to facilitate oriented immobilization by covalent binding to the bioreactive Y-functional group of the monolayer.
  • The affinity tag may comprise a poly(amino acid) tag. A poly(amino acid) tag is a polypeptide at comprises from about 2 to about 100 residues of a single amino acid. For instance, the affinity tag may comprise a poly-cysteine, poly-lysine, poly-arginine, or poly-histidine. Amino acid tags are preferably composed of two to twenty residues of a single amino acid, such as, for example, histidines, lysines, arginines, cysteines, glutamines, tyrosines, or any combination of these. According to a preferred embodiment, an amino acid tag of one to twenty amino acids includes at least one to ten cysteines for thioether linkage; or one to ten lysines for amide linkage; or one to ten arginines for coupling to vicinal dicarbonyl groups. One of ordinary skill in the art can readily pair suitable affinity tags with a given Y-functional group.
  • The position of the amino acid tag can be at the amino-, or carboxy-terminus of the protein or anywhere in-between. Where compatible with protein function, affinity tags introduced for protein purification are preferentially located at the C-terminus of the recombinant protein to ensure that only full-length proteins are isolated during protein purification.
  • Affinity tags may also contain one or more unnatural amino acids. Unnatural amino acids can be introduced using suppressor tRNAs that recognize stop codons (i.e. amber). The tRNAs are chemically amino-acylated to contain chemically altered (“unnatural”) amino acids for use with specific coupling chemistries (i.e. ketone modifications, photoreactive groups).
  • In an alternative embodiment the affinity tag can comprise a whole protein, such as, but not limited to, glutathione S-transferase, an antibody, avidin, or streptavidin.
  • Other protein conjugation and immobilization techniques known in the art may be adapted for the purpose of immobilizing proteins on activated monolayers. For instance, an organic bioconjugate, such as biotin or an antigen, amy be chemically cross linked to the protein to be immobilized.
  • In a preferred embodiment, the fusion protein that comprises the first and second polypeptides comprises the affinity tag as well. Methods for the expression of fusion proteins have been outlined above.
  • In one embodiment of the protein-coated substrate, the adaptor polypeptide is protein G or protein A. Adaptor proteins are especially useful to increase the solubility of the protein of interest and to increase the distance between the surface of the substrate or coating and the protein of interest Use of an adaptor protein or polypeptide can also be very useful in facilitating the preparative steps' of protein purification by affinity binding. Examples of possible adaptor proteins include glutathione-S-transferase (GST, maltose-binding protein, chitin-binding protein, thioredoxin, green-fluorescent protein (GFP). GFP can also be used for quantification of surface binding.
  • (d) EXAMPLES
  • The following specific examples are intended to illustrate the invention and should not be construed as limiting the scope of the claims:
  • Example 1 Fabrication of a Two-Dimensional Array by Photolithography
  • In a preferred embodiment of the invention, two-dimensional arrays are fabricated onto the device material via standard microstereolithography and/or thin film deposition. Alternative techniques include microcontact printing. Usually, a computer-aided design pattern (reflecting the final channel geometries) is transferred to a photomask using standard techniques, which is then used to transfer the pattern onto a silicon wafer coated with photoresist.
  • In a typical example, the device (“chip”) with lateral dimensions of 20×20 mm contains an array of squared patches of a bioreactive layer (here: gold on silicon) each 0.1×0.1 mm in size and separated by hydrophobic surface areas with a 0.2 mm spacing. 4′ diameter Si(100) wafers (Virginia Semiconductor) or 4″ diameter Corning 7740 glass wafers are used as bulk materials. Si(100) wafers are first cleaned in a 5:1:1 deionized (DI) water: NH3: H2O2 bath (RCA1, 90° C., 10 min), followed by a 5:1:1 DI water: HCl H2O2 bath (RCA2, 90° C., 10 min) and finally passivated in 1% aqueous HF and singed at 150° C. for 30 min to become hydrophobic., The wafer is then spincoated with photoresist (Shipley 1813), prebaked for 25 minutes at 90° C., exposed using a Karl Suss contact printer and developed according to standard protocols. The wafer is then dried and postbaked at 110° C. for 25 min In the next step, the wafer is primed with a 20 nm thin titanium layer, followed by a 200 nm thin gold layer both layers deposited using electron-beam evaporation (5 Å/s, Thermionics). After resist stripping and a short plasma treatment, the gold patches can be further chemically modified to achieve the desired bioreactive and biocompatible properties (see Example 3, below).
  • Example 2 Fabrication of a Two-Dimensional Array by Deposition Through a Hole Mask
  • In another preferred embodiment the array of gold patches is fabricated by thin film deposition through a hole mask which is in direct contact with the substrate. In a typical example, Si(100) wafers are first cleaned in a 5:1:1 deionized (DI) water: NH3H2O2 bath (RCA1, 90° C., 10 min), followed by a 5:1:1 DI water: HCl: H2O2 bath (RCA2, 90° C., 10 min) and finally passivated in 1% aqueous HF and singed at 150° C. for 30 min to become hydrophobic. The wafer is then brought into contact with a hole mask exhibiting the positive pattern of the desired patch array. In the next step, the wafer is primed with a 20 nm thin titanium layer, followed by a 200 nm thin gold layer both layers deposited using electron-beam evaporation (5 Å/s, Thermionics). After removal of the mask, the gold patches' can be further chemically modified to achieve the desired bioreactive and biocompatible properties (see Example 3, below).
  • Example 3 Synthesis of an Aminoreactive Monolayer Molecule (Following the Procedure Outined in Wagner et al., Biophys. J., 1996, 70:2052-2066).
  • General 1H- and 13C-NMR spectra are recorded on Bruker instruments (100 to 400 MHz). Chemical shifts (δ) are reported in ppm relative to internal standard ((CH3)4Si δ=0.00 (1H- and 13C-NMR)). FAB-mass spectra are recorded on a VG-SABSEQ instrument (Cs+, 20 keV). Transmission infrared spectra are obtained as dispersions in KBr on an FTIR Perkin-Elmer 1600 Series instrument. Thin-layer chromatography (TLC) is performed on precoated silica gel. 60 F254 plates (MERCK, Darmstadt, FRG), and detection was done using Cl2/toluidine, PdCl2 and UV-detection under NH3-vapor. Medium pressure liquid chromatography (MPLC) is performed on a Labomatic MD80 (LABOMATIC INSTR AG, Allschwil, Switzerland) using a Buechi column (460×36 mm; BUECHI Flawil, Switzerland), filled with silica gel 60 (particle size 15-40 μm) from Merck
  • Synthesis of 1, 11′-dithiobis(succinmidylundecanoate) (DSU). Sodium thiosulfate (55.3 g, 350 mmol) is added to a suspension of 11-bromo-undecanoic acid (92.8 g, 350 mmol) in 50% aqueous 1,4-dioxane (1000 ml). The mixture is heated at reflux (90° C.) for 2 h until the reaction to the intermediate Bunte salt was complete (clear solution). The oxidation to the corresponding disulfide is carried out in situ by adding iodine in portions until the solution retained with a yellow to brown colour. The surplus of iodine is retitrated with 15% sodium pyrosulfite in water. After removal of 1,4 dioxane by rotary evaporation the creamy suspension is filtered to yield product 11,11′-dithiobis(undecanoic acid). Recrystalliaation from ethyl acetate/THF provides a white solid (73.4 g, 96.5/o): mp 94° C., 1H NMR (400 MHz, CDCl3/CD3OD 95:5): δ 2.69 (t, 2H, J=7.3 Hz), 2.29 (t, 2H, J=7.5 Hz), 1.76-1.57 (m, 4H), and 1.40-1.29 (m, 12H); FAB-MS (Cs+, 20 keV): m/z (relative intensity) 434 (100, M+). Anal. Calcd. for C22H42O4S2:C, 60.79; H, 9.74; S, 14.75. Found: C, 60.95; H. 9.82; S, 14.74. To a solution of 11,11′-dithiobis(undecanoic acid). (1.0 g, 2.3 mmol) in THF (50 ml) is added N-hydroxysuccinimide (0.575 g, 5 mmol) followed by DCC (1.03 g, 5 mmol) at 0° C. After the reaction mixture is allowed to warm to 23° C. and is stirred for 36 h at room temperature, the dicyclohexylurea (DCU) is filtered. Removal of the solvent under reduced pressure and recrystaliztion from acetone/hexane provides 11,11′-dithobis(succinimidylundecanoate) as a white solid. Final purification is achieved by medium pressure liquid chromatography (9 bar) using silica gel and a 2:1 mixture of ethyl acetate and hexane. The organic phase is concentrated and dried in vacuum to afford 11,11′-dithiobis(succinimidylundecanoate) (1.12 g, 78%): mp 95 Ic; 1H NMR (400 MHz, CDCl3): δ 2.83 (s, 4H), 2.68 (t, 2HK J=7.3 Hz), 2.6(t, 2H, J=7.5 Hz), 1.78-1.63 (m, 4H), and 1.43-1.29 (m, 12H); FAB-MS (Cs+, 20 keV): m/z (relative intensity) 514 (100), 628 (86, M+). Anal. Calcd. for C30H48N2O8S2: C, 57.30; H, 7.69; N, 4.45; S, 10.20. Found: C, 57.32; H, 7.60; N, 4.39; S. 10.25.
  • Example 4 Formation of an Aminoreactive Monolayer on Gold (Following the Procedure of Wagner et al., Biophys. J, 1996, 70:2052-2066).
  • Monolayers based on 11,11′-dithiobis(succinimidylundecanoate) (SU) can be deposited on Au(111) surfaces of microdevices described under Examples 1 and 2 by immersing them into a 1 mM solution of DSU in chloroform at room temperature for 1 hour. After rinsing with 10 volumes of solvent, the N-hydroxysuccinimide-terminated monolayer is dried under a stream of nitrogen and immediately used for protein immobilization.
  • Example 5 Expression and Purification of Human Caspase Fusion Proteins
  • Caspases are cysteine proteases of the papain superfamily, with a different active site and catalytic mechanism than observed for papain, Wilson, K P. et al., Nature, 1994 370:270-275. Caspases are important enzymes in the promotion of the cell death pathways and inflammation, Villa, et al., TIBS, 1997, 22:288-392. Identification of selective caspase inhibitors is essential to prevent cross-inhibition of other caspase-dependent pathways. Caspases 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, Villa, et al, TIBS, 1997, 22:288-392 and new caspase homologs identified by the human genome project are PCR amplified and cloned into an E. coli expression vector containing an N-terminal histidine tag, Hochuli, et al., Biotechnolbgy, 1988 6:1321, a factor Xa cleavage site, a lysine tag and a tri-glycine linker. Fusion proteins are expressed, purified by nickel-nitrilotriacetic acid (NTA) agarose chromatography, the histidine tag removed by factor Xa cleavage, followed by gel filtration. Caspases are snap-frozen and stored in 20 mM PIPES, pH 7.2, 150 mM NaCl, 0.1% CHAPS, 10% sucrose at −80° C.
  • Example 6 Immobilization of Fusion Proteins on a 2D-Protein Array
  • Caspase-fusion proteins can be immobilized to the aminoreactive monolayer surface of the bioreactive patches of the two-dimensional array (see Examples 1, 2, and 4 above). Caspase fusion proteins can be diluted to concentrations of 1 μg/ml in 20 mM PIPES, pH 7.2, 150 mM NaC, 0.91% CHAPS, 10% sucrose and applied onto the bioreactive patches using a computer-aided, capillary-based dispensing system. After an immobiization period of 30 min, the 2D array was rinsed and subjected to analysis; Ultrapure water with a resistance of 18 MΩcm is generally useable for all aqueous buffers (purified by passage through a Barnstead Nanopure® system).
  • Example 7 Assay of Caspase Activity on a Two Dimensional Array
  • Caspase activity can be determined by a binding assay using three fluorescently labeled peptide aldehyde inhibitors that form a reversible thiohemiacetal moiety with the active site cysteine, Thornberry, Methods in Enzymology, 1994, 244:615-631. The peptides are adapted to caspase 1, 3, 4, 7: Dns (dansyl)-SS-DEVD-CHO, caspase 1: Dns-SS-VDVAD-CHO, caspase 6: Dns-SS-VQID-CHO, Talanian, J. Biol Chem, 1997, 272:9677-9682. The affinity for Ac-DEVD-CHO to caspase 1 is determined to be in the low nanomolar range, Thornberry, Methods in Enzymology, 1994, 244:615-631. The assay buffer is 20 mM PIPES, pH 7.2, 150 mM NaCl, 0.1% CHAPS, 10% sucrose, Stennicke, and Salvesen, J. Biol. Chem, 1997, 272:25719-25723. Fluorescently labeled peptides are mixed to a final concentration of 1 to 5 nM each, the potential drug compound added and flushed onto the 2D array. Peptides are allowed to bind for 10-60 min., unbound peptide removed by washing with buffer and the fluorescence intensity measured (excitation at 360 nm, emission at 470 nm).
  • All documents cited in the above specification are herein incorporated by reference. Various modifications and variations of the present invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in the art are intended to be within the scope of the following claims.

Claims (41)

1.-41. (canceled)
42. An array of proteins, comprising: a substrate; and a plurality of patches arranged in discrete known regions on said substrate, wherein each of said patches comprises an immobilized protein, and wherein the array comprises 100 or more patches per 1 cm2.
43. The array of claim 42, wherein the array comprises 1000 or more patches per 1 cm2.
44. The array of claim 42, wherein at least some of said patches comprise different immobilized proteins.
45. The array of claim 44 which comprises at least about 9 different immobilized proteins.
46. The array of claim 45 which comprises at least about 100 different immobilized proteins.
47. The array of claim 46 which comprises at least about 1000 different immobilized proteins.
48. The array of claim 42, wherein the area of the substrate surface covered by each of the patches is about 100 nm2 or greater.
49. The array of claim 42, wherein the area of the substrate surface covered by each of the patches is between about 100 nm2 and about 40,000 m2.
50. The array of claim 42, wherein the area of the substrate surface covered by each of the patches is between about 1 μm2 to about 10,000 μm2.
51. The array of claim 42, wherein said plurality of patches further comprise functionalized surfaces formed on said substrate by microstamping or microcontact printing.
52. An array of claim 42, wherein said proteins are functionally related or are suspected of being functionally related.
53. An array of claim 42, wherein said proteins are structurally related or are suspected of being structurally related.
54. An array of claim 42, wherein said proteins are members of the same family.
55. An array of claim 54, wherein the proteins are selected from the group consisting of growth factor receptors, catecholamine receptors, amino acid derivative receptors, cytokine receptors, extracellular matrix receptors, immunoglobulins, lectins, cytokines, serpins, proteases, kinases, phosphatases, ras-like GTPases, hydrolases, steroid hormone receptors, heat-shock transcription factors, zinc-finger proteins, leucine-zipper proteins, homeodomain proteins, hepatitis C virus (HCV) proteases and HIV proteases.
56. An array of claim 42, wherein said patches further comprise a monolayer on the surface of the substrate and wherein the proteins of said patches are immobilized on said monolayer.
57. An array of claim 56, wherein the monolayer of an individual patch comprises molecules of the formula X-R-Y, wherein R is a spacer, X is a functional group that binds R to the surface, and Y is a functional group for binding proteins onto the monolayer.
58. An array of claim 57, wherein the monolayer of an individual patch comprises at least two different X-R-Y molecules.
59. An array of claim 56 or 57, wherein said substrate is selected from the group consisting of silicon, silicon oxide, indium tin oxide, magnesium oxide, alumina, quartz, glass, and silica, wherein X, prior to incorporation into said monolayer, is selected from the group consisting of a monohalosilane, dihalosilane, trihalosilane, trichlorosilane, trialkoxysilane, dialkoxysilane, and monoalkoxysilane, wherein R is an alkyl from about 8 to 22 carbons long, and wherein Y comprises a functional group selected from the group consisting of a maleimide, N-hydroxysuccinimide, nitrilotriacetic acid, activated hydroxyl, haloacetyl, bromoacetyl, iodoacetyl, activated carboxyl, hydrazide, epoxy, aziridine, trifluoromethyldiaziridine, pyridyldisulfide, N-acyl-imidazole, imidazolecarbamate, succinimidylcarbonate, arylazide, anhydride, diazoacetate, benzophenone, isothiocyanate, isocyanate, imidoester, fluorobenzene, and biotin.
60. An array of claim 56, further comprising a coating between said substrate and said monolayer, wherein said coating is formed on the substrate or applied to the substrate.
61. An array of claim 60, wherein the mean roughness of said coating is less than about 5 angstroms for areas of at least 25 μm2.
62. An array of claim 61, wherein the mean roughness of said coating is less than about 3 angstroms for areas of at least 25 μm2.
63. An array of claim 60, wherein said coating is an ultraflat, template-stripped surface.
64. An array of claim 62, wherein said coating comprises a metal film, wherein the monolayer of an individual patch comprises at least two different X-R-Y molecules, and wherein X prior to incorporation into said monolayer, is a functional group selected from the group consisting of an asymmetrical or symmetrical disulfide, sulfide, diselenide, selenide, thiol, isonitrile, selenol, trivalent phosphorus compounds, isothiocyanate, isocyanate, xanthanate, thiocarbamate, phosphines, amines, thio acid and dithio acid, wherein R is an alkyl chain from about 8 to about 22 carbons long, and wherein Y comprises a functional group selected from the group consisting of a maleimide, N-hydroxysuccinimide, nitrilotriacetic acid, activated hydroxyl, haloacetyl, bromoacetyl, iodoacetyl, activated carboxyl, hydrazide, epoxy, aziridine, trifluoromethyldiaziridine, pyridyldisulfide, N-acyl-imidazole, imidazolecarbaamate, succinimidylcarbonate, arylazide, anhydride, diazoacetate, benzophenonee, isothiocyanate, isocyanate, imidoester, fluorobenzene, and biotin.
65. An array of claim 56, wherein the monolayer of an individual patch further comprises a second molecule, wherein said second molecule is of the formula X-R-V, and wherein R is spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins.
66. An array of claim 56, wherein said array further comprises at least one patch comprising a monolayer of molecules of the formula X-R-V, wherein R is a spacer, X is a functional group that binds R to the surface, and V is a moiety resistant to the non-specific binding of proteins.
67. An array of claim 56, further comprising crosslinking between molecules of the monolayer of a patch.
68. An array of claim 56, further comprising an affinity tag, wherein said affinity tag enhances site specific immobilization of the biological moiety onto the monolayer.
69. An array of claim 68, wherein said affinity tag comprises an unnatural amino acid.
70. An array of claim 68, wherein said affinity tag comprises a poly(amino acid).
71. An array of claim 70, wherein said poly(amino acid) is selected from the group consisting of poly-cysteine, polylysine, poly-arginine, and poly-histidine.
72. An array of claim 68, wherein said affinity tag comprises a polypeptide or a protein.
73. An array of claim 66, wherein the affinity tag and the biological moiety together compose a fusion protein.
74. An array of claim 66, further comprising an adaptor molecule that links the affinity tag to the immobilized biological moiety.
75. An array of claim 74, wherein said adaptor molecule is a polypeptide or a protein.
76. An array of claim 75, wherein said adaptor molecule is selected from the group consisting of green fluorescent protein, glutathione S-transferase, maltose-binding protein chitin-binding protein, and thioredoxin.
77. An array of claim 74, wherein the affinity tag, adaptor molecule, and biological moiety together compose a fusion protein.
78. A method for screening a plurality of proteins for their ability to interact with a component of a fluid sample, comprising: delivering the fluid sample to the array of claim 42; and detecting, either directly or indirectly, the interaction of said component with the immobilized protein of each patch.
79. A method for screening a plurality of proteins for their ability to bind a component of a fluid sample, comprising: delivering said fluid sample to the array of claim 42; washing the array with fluid which does not contain said component in order to elute unbound component; and detecting, either directly or indirectly, the presence of said component retained at each patch.
80. A method for detecting in a fluid sample the presence of a plurality of analytes which react with said proteins, comprising: delivering the fluid sample to the array of claim 42; and detecting the interaction of the analyte with the immobilized protein at each patch.
81. A method for detecting in a fluid sample the presence of a plurality of analytes which bind said proteins, comprising: delivering the fluid sample to the array of claim 42; washing said array with an analyte-free fluid to remove unbound analyte; and detecting, either directly or indirectly, the presence of analyte retained at each patch.
US10/911,945 1998-07-14 2004-08-04 Protein arrays for high-throughput screening Abandoned US20050008674A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/911,945 US20050008674A1 (en) 1998-07-14 2004-08-04 Protein arrays for high-throughput screening

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/115,455 US6406921B1 (en) 1998-07-14 1998-07-14 Protein arrays for high-throughput screening
US10/112,840 US20020106702A1 (en) 1998-07-14 2002-03-29 Protein arrays for high-throughput screening
US10/911,945 US20050008674A1 (en) 1998-07-14 2004-08-04 Protein arrays for high-throughput screening

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/112,840 Continuation US20020106702A1 (en) 1998-07-14 2002-03-29 Protein arrays for high-throughput screening

Publications (1)

Publication Number Publication Date
US20050008674A1 true US20050008674A1 (en) 2005-01-13

Family

ID=22361520

Family Applications (14)

Application Number Title Priority Date Filing Date
US09/115,455 Expired - Lifetime US6406921B1 (en) 1998-07-14 1998-07-14 Protein arrays for high-throughput screening
US09/353,215 Expired - Fee Related US6475808B1 (en) 1998-07-14 1999-07-14 Arrays of proteins and methods of use thereof
US09/353,555 Expired - Lifetime US6329209B1 (en) 1998-07-14 1999-07-14 Arrays of protein-capture agents and methods of use thereof
US09/570,588 Expired - Fee Related US6475809B1 (en) 1998-07-14 2000-05-12 Protein arrays for high-throughput screening
US09/570,363 Expired - Fee Related US6630358B1 (en) 1998-07-14 2000-05-12 Arrays of proteins and methods of use thereof
US09/574,748 Expired - Lifetime US6365418B1 (en) 1998-07-14 2000-05-18 Arrays of protein-capture agents and methods of use thereof
US10/107,122 Abandoned US20030003599A1 (en) 1998-07-14 2002-03-26 Arrays of protein-capture agents and methods of use thereof
US10/112,840 Abandoned US20020106702A1 (en) 1998-07-14 2002-03-29 Protein arrays for high-throughput screening
US10/113,964 Abandoned US20020110933A1 (en) 1998-07-14 2002-03-29 Arrays of proteins and methods of use thereof
US10/911,877 Abandoned US20050014292A1 (en) 1998-07-14 2004-08-04 Protein arrays for high-throughput screening
US10/911,945 Abandoned US20050008674A1 (en) 1998-07-14 2004-08-04 Protein arrays for high-throughput screening
US11/392,262 Abandoned US20060228701A1 (en) 1998-07-14 2006-03-28 Arrays of protein-capture agents and methods of use thereof
US12/192,321 Abandoned US20090131278A1 (en) 1998-07-14 2008-08-15 Non-specific binding resistant protein arrays and methods for making the same
US12/950,698 Abandoned US20110086779A1 (en) 1998-07-14 2010-11-19 Arrays of protein capture agents and methods of use thereof

Family Applications Before (10)

Application Number Title Priority Date Filing Date
US09/115,455 Expired - Lifetime US6406921B1 (en) 1998-07-14 1998-07-14 Protein arrays for high-throughput screening
US09/353,215 Expired - Fee Related US6475808B1 (en) 1998-07-14 1999-07-14 Arrays of proteins and methods of use thereof
US09/353,555 Expired - Lifetime US6329209B1 (en) 1998-07-14 1999-07-14 Arrays of protein-capture agents and methods of use thereof
US09/570,588 Expired - Fee Related US6475809B1 (en) 1998-07-14 2000-05-12 Protein arrays for high-throughput screening
US09/570,363 Expired - Fee Related US6630358B1 (en) 1998-07-14 2000-05-12 Arrays of proteins and methods of use thereof
US09/574,748 Expired - Lifetime US6365418B1 (en) 1998-07-14 2000-05-18 Arrays of protein-capture agents and methods of use thereof
US10/107,122 Abandoned US20030003599A1 (en) 1998-07-14 2002-03-26 Arrays of protein-capture agents and methods of use thereof
US10/112,840 Abandoned US20020106702A1 (en) 1998-07-14 2002-03-29 Protein arrays for high-throughput screening
US10/113,964 Abandoned US20020110933A1 (en) 1998-07-14 2002-03-29 Arrays of proteins and methods of use thereof
US10/911,877 Abandoned US20050014292A1 (en) 1998-07-14 2004-08-04 Protein arrays for high-throughput screening

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/392,262 Abandoned US20060228701A1 (en) 1998-07-14 2006-03-28 Arrays of protein-capture agents and methods of use thereof
US12/192,321 Abandoned US20090131278A1 (en) 1998-07-14 2008-08-15 Non-specific binding resistant protein arrays and methods for making the same
US12/950,698 Abandoned US20110086779A1 (en) 1998-07-14 2010-11-19 Arrays of protein capture agents and methods of use thereof

Country Status (8)

Country Link
US (14) US6406921B1 (en)
EP (2) EP1097380A1 (en)
JP (2) JP2002520618A (en)
AT (1) ATE397752T1 (en)
AU (2) AU765508B2 (en)
CA (2) CA2337654A1 (en)
DE (1) DE69938867D1 (en)
WO (2) WO2000004382A1 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020106702A1 (en) * 1998-07-14 2002-08-08 Peter Wagner Protein arrays for high-throughput screening
US20020115225A1 (en) * 1998-07-14 2002-08-22 Peter Wagner Microdevices for high-throughput screening of biomolecules
US20030082588A1 (en) * 2001-07-13 2003-05-01 Viswanadham Garimella Method for immobilizing molecules onto surfaces
US20030138973A1 (en) * 1998-07-14 2003-07-24 Peter Wagner Microdevices for screening biomolecules
US20030207467A1 (en) * 2000-05-04 2003-11-06 Michael Snyder Protein chips for high throughput screening of protein activity
US20040241751A1 (en) * 1998-07-14 2004-12-02 Peter Wagner Arrays of protein-capture agents and methods of use thereof
US20050048580A1 (en) * 2001-01-23 2005-03-03 President And Fellows Of Harvard College Nucleic-acid programmable protein arrays
US20050100947A1 (en) * 1998-07-14 2005-05-12 Zyomyx, Inc. Array devices and methods of use thereof
US20050118665A1 (en) * 2003-06-09 2005-06-02 Zhou Fang X. Methods for conducting assays for enzyme activity on protein microarrays
US20050233473A1 (en) * 2002-08-16 2005-10-20 Zyomyx, Inc. Methods and reagents for surface functionalization
US20050260653A1 (en) * 2004-04-14 2005-11-24 Joshua Labaer Nucleic-acid programmable protein arrays
US20050287560A1 (en) * 2001-07-13 2005-12-29 Nanosphere, Inc. Method for preparing substrates having immobilized molecules and substrates
US20060073593A1 (en) * 2001-02-07 2006-04-06 Invitrogen Corporation Compositions and methods for molecular biology
US20070134714A1 (en) * 2002-05-28 2007-06-14 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
EP1882520A1 (en) * 2006-07-25 2008-01-30 Samsung Electronics Co., Ltd. Patterned spot microarray using photocatalyst and a method of manufacturing the same
US20080071071A1 (en) * 2006-06-29 2008-03-20 President And Fellows Of Harvard College Evaluating proteins
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
US9694518B2 (en) 2014-06-20 2017-07-04 The Regents Of The University Of Michigan Breath-activated images and anti-counterfeit authentication features formed of nanopillar arrays

Families Citing this family (750)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5776748A (en) 1993-10-04 1998-07-07 President And Fellows Of Harvard College Method of formation of microstamped patterns on plates for adhesion of cells and other biological materials, devices and uses therefor
ATE270529T1 (en) * 1996-02-16 2004-07-15 Smith & Nephew Inc TRANSPLANT ANCHORAGE
US6251691B1 (en) 1996-04-25 2001-06-26 Bioarray Solutions, Llc Light-controlled electrokinetic assembly of particles near surfaces
US7041510B2 (en) * 1996-04-25 2006-05-09 Bioarray Solutions Ltd. System and method for programmable illumination pattern generation
US7169556B2 (en) 1996-07-29 2007-01-30 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US6984491B2 (en) 1996-07-29 2006-01-10 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US7098320B1 (en) 1996-07-29 2006-08-29 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
AU4043497A (en) 1996-07-29 1998-02-20 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US6750016B2 (en) * 1996-07-29 2004-06-15 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US6506564B1 (en) * 1996-07-29 2003-01-14 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US6582921B2 (en) 1996-07-29 2003-06-24 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses thereof
US6368877B1 (en) * 1997-06-25 2002-04-09 Massachusetts Institute Of Technology Self-assembling peptide surfaces for cell patterning and interactions
US6974669B2 (en) * 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US20050037397A1 (en) * 2001-03-28 2005-02-17 Nanosphere, Inc. Bio-barcode based detection of target analytes
US20020144905A1 (en) * 1997-12-17 2002-10-10 Christian Schmidt Sample positioning and analysis system
CA2316966C (en) 1997-12-17 2008-04-08 Horst Vogel Positioning and electrophysiological characterization of individual cells and reconstituted membrane systems on microstructured carriers
US7244349B2 (en) 1997-12-17 2007-07-17 Molecular Devices Corporation Multiaperture sample positioning and analysis system
WO1999039210A1 (en) * 1998-01-29 1999-08-05 Miller, Samuel High density arrays for proteome analysis and methods and compositions therefor
GB9812783D0 (en) 1998-06-12 1998-08-12 Cenes Ltd High throuoghput screen
US6897073B2 (en) * 1998-07-14 2005-05-24 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US7132247B1 (en) * 1998-09-17 2006-11-07 Regents Of The University Of Minnesota Composite devices incorporating biological material and methods
WO2000022112A1 (en) * 1998-10-13 2000-04-20 The University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis and use
US20030190740A1 (en) 1998-10-13 2003-10-09 The University Of Georgia Research Foundation, Inc Stabilized bioactive peptides and methods of identification, synthesis, and use
IL143027A0 (en) * 1998-11-16 2002-04-21 Genway Biotech Inc Generation of antibodies using polynucleotide vaccination in avian species
US6827979B2 (en) 1999-01-07 2004-12-07 Northwestern University Methods utilizing scanning probe microscope tips and products therefor or produced thereby
US6635311B1 (en) 1999-01-07 2003-10-21 Northwestern University Methods utilizing scanning probe microscope tips and products therefor or products thereby
US7166475B2 (en) * 1999-02-26 2007-01-23 Cyclacel Ltd. Compositions and methods for monitoring the modification state of a pair of polypeptides
EP1159615A2 (en) * 1999-03-10 2001-12-05 National Institutes of Health, as represented by the Secretary, Department of Health and Human Services of the Government Universal protein array system
WO2000056774A1 (en) * 1999-03-19 2000-09-28 Duke University Methods of using bioelastomers
DE19916867A1 (en) * 1999-04-14 2000-10-19 Fraunhofer Ges Forschung Making a molecular array on which molecules are immobilized, using micro-compartments or microcapillary reactors on planar substrates with sensor elements employs microelectronic production techniques
US20040053290A1 (en) * 2000-01-11 2004-03-18 Terbrueggen Robert Henry Devices and methods for biochip multiplexing
US7638464B2 (en) * 1999-04-26 2009-12-29 Biocept, Inc. Three dimensional format biochips
CN1169188C (en) 1999-04-29 2004-09-29 赛弗根生物系统股份有限公司 Sample holder with hydrophobic coating for gas phase mass spectrometers
US6690399B1 (en) * 1999-05-07 2004-02-10 Tropix, Inc. Data display software for displaying assay results
JP4493125B2 (en) * 1999-05-07 2010-06-30 独立行政法人理化学研究所 Method for detecting interacting proteins
US7932213B2 (en) * 1999-05-11 2011-04-26 President And Fellows Of Harvard College Small molecule printing
US6824987B1 (en) * 1999-05-11 2004-11-30 President And Fellows Of Harvard College Small molecule printing
US20030073250A1 (en) * 1999-05-21 2003-04-17 Eric Henderson Method and apparatus for solid state molecular analysis
US6573369B2 (en) * 1999-05-21 2003-06-03 Bioforce Nanosciences, Inc. Method and apparatus for solid state molecular analysis
US20030186311A1 (en) * 1999-05-21 2003-10-02 Bioforce Nanosciences, Inc. Parallel analysis of molecular interactions
US20020042081A1 (en) * 2000-10-10 2002-04-11 Eric Henderson Evaluating binding affinities by force stratification and force panning
DE60044923D1 (en) * 1999-05-28 2010-10-21 Yokogawa Electric Corp Biochip reader
US7179638B2 (en) * 1999-07-30 2007-02-20 Large Scale Biology Corporation Microarrays and their manufacture by slicing
ES2367551T3 (en) * 1999-08-13 2011-11-04 Bayer Technology Services Gmbh DEVICE AND PROCEDURE FOR THE DETERMINATION OF MULTIPLE ANALYTICS.
US8111401B2 (en) * 1999-11-05 2012-02-07 Robert Magnusson Guided-mode resonance sensors employing angular, spectral, modal, and polarization diversity for high-precision sensing in compact formats
US7167615B1 (en) 1999-11-05 2007-01-23 Board Of Regents, The University Of Texas System Resonant waveguide-grating filters and sensors and methods for making and using same
DE60031441T2 (en) * 1999-11-29 2007-08-30 Unilever N.V. IMMOBILIZATION OF PROTEINS WITH THE HELP OF A POLYPEPTIDE EGMENTS
SG121902A1 (en) * 2000-01-11 2006-05-26 Maxygen Inc Integrated systems for diversity generation and screening
JP2004501340A (en) * 2000-01-13 2004-01-15 ナノスフェアー インコーポレイテッド Oligonucleotide-attached nanoparticles and methods of use
DE60142765D1 (en) * 2000-01-31 2010-09-23 Sense Proteomic Ltd METHOD FOR PRODUCING PROTEIN EXPRESSION ARRAYS AND APPLYING THE SAME IN FAST SCREENING
US7816098B2 (en) 2000-01-31 2010-10-19 Sense Proteomic Limited Methods of making and using a protein array
ATE477489T1 (en) * 2000-01-31 2010-08-15 Sense Proteomic Ltd METHOD FOR PRODUCING PROTEIN EXPRESSION ARRAYS AND APPLYING THE SAME IN RAPID SCREENING
US20080280771A1 (en) * 2000-02-08 2008-11-13 Regents Of The University Of Michigan Protein MicroarraySystem
US20050230315A1 (en) * 2003-01-13 2005-10-20 Regents Of The University Of Michigan Protein microarray system
CA2401118A1 (en) 2000-02-23 2001-08-30 Zyomyx, Inc. Microfluidic devices and methods
US6897015B2 (en) * 2000-03-07 2005-05-24 Bioforce Nanosciences, Inc. Device and method of use for detection and characterization of pathogens and biological materials
US6686461B1 (en) 2000-03-22 2004-02-03 Solulink Bioscience, Inc. Triphosphate oligonucleotide modification reagents and uses thereof
US7102024B1 (en) 2000-08-01 2006-09-05 Schwartz David A Functional biopolymer modification reagents and uses thereof
US6541071B1 (en) * 2000-03-23 2003-04-01 Corning Incorporated Method for fabricating supported bilayer-lipid membranes
EP1294513A4 (en) 2000-03-27 2005-11-23 Zyomyx Inc Site-specific, covalent bioconjugation of proteins
JP2003529772A (en) * 2000-03-30 2003-10-07 インフィネオン テクノロジーズ アクチエンゲゼルシャフト Biosensor, biosensor array, method for producing biosensor electrode, method for producing biosensor
CA2403296A1 (en) * 2000-04-10 2001-10-18 Benjamin F. Cravatt Proteomic analysis using activity-based probe libraries
US20010041349A1 (en) * 2000-04-17 2001-11-15 Andrew Patron Protein expression system arrays and use in biological screening
DE10020704B4 (en) * 2000-04-27 2006-09-28 Bioref Gmbh Biochip for archiving and laboratory medical analysis of biological sample material, process for its production and its use in diagnostic procedures
DE10027397A1 (en) * 2000-06-02 2001-12-13 Graffinity Pharm Design Gmbh Surface for the immobilization of ligands
US7153682B2 (en) * 2000-06-05 2006-12-26 Chiron Corporation Microarrays on mirrored substrates for performing proteomic analyses
ES2281424T3 (en) * 2000-06-05 2007-10-01 Novartis Vaccines And Diagnostics, Inc. MICROMATRICES TO CARRY OUT PROTEOMIC ANALYSIS.
US7148058B2 (en) * 2000-06-05 2006-12-12 Chiron Corporation Protein microarrays on mirrored surfaces for performing proteomic analyses
DE60140120D1 (en) 2000-06-14 2009-11-19 Vistagen Inc TOXICITY TYPING USING LIVESTOCK CELLS
AU2001269906A1 (en) * 2000-06-19 2002-01-02 Zyomyx, Inc. Methods for immobilizing polypeptides
US9709559B2 (en) 2000-06-21 2017-07-18 Bioarray Solutions, Ltd. Multianalyte molecular analysis using application-specific random particle arrays
EP1311839B1 (en) 2000-06-21 2006-03-01 Bioarray Solutions Ltd Multianalyte molecular analysis using application-specific random particle arrays
US20020115068A1 (en) * 2000-06-23 2002-08-22 Ian Tomlinson Matrix screening method
DE10031587A1 (en) * 2000-06-29 2002-01-10 Basf Ag Dosing of microscale suspensions for the production of material samples in combinatorial materials research and their testing
US6602669B2 (en) 2000-07-11 2003-08-05 Northwestern University Method of detection by enhancement of silver staining
US7518724B2 (en) * 2000-07-11 2009-04-14 Maven Technologies Image acquisition, processing, and display
US7023547B2 (en) * 2000-07-11 2006-04-04 Maven Technologies, Llc Apparatus including a biochip for imaging of biological samples and method
EP1301632A2 (en) * 2000-07-19 2003-04-16 Pointilliste, Inc. Nested sorting and high throughput screening
EP1354031A2 (en) * 2000-07-31 2003-10-22 Maxygen, Inc. Nucleotide incorporating enzymes
AU2001277245A1 (en) * 2000-08-03 2002-02-18 Massachusetts Institute Of Technology Microarrays of functional biomolecules, and uses therefor
US7270730B2 (en) 2000-08-04 2007-09-18 Essen Instruments, Inc. High-throughput electrophysiological measurement system
US7067046B2 (en) 2000-08-04 2006-06-27 Essen Instruments, Inc. System for rapid chemical activation in high-throughput electrophysiological measurements
US7678539B2 (en) 2000-08-10 2010-03-16 Corning Incorporated Arrays of biological membranes and methods and use thereof
US6977155B2 (en) 2000-08-10 2005-12-20 Corning Incorporated Arrays of biological membranes and methods and use thereof
AU2001284899A1 (en) * 2000-08-11 2002-02-25 Qianjin Hu Methods and universal monoclonal antibody array
CA2419544A1 (en) * 2000-08-14 2002-02-21 Surface Logix, Inc. Pathways arrays
JP4719403B2 (en) * 2000-08-15 2011-07-06 ディサーナ リミテッド Functional protein array
CN1261745C (en) * 2000-08-15 2006-06-28 拜澳富斯毫微科学有限公司 Nanoscale molecular matrix arrayer
KR20030043933A (en) * 2000-08-17 2003-06-02 센스 프로테오믹 리미티드 Method
US7094568B2 (en) * 2000-08-17 2006-08-22 Sense Proteomic Ltd. Method for producing proteins tagged at the N- or C-terminus
EP1184349A1 (en) * 2000-09-01 2002-03-06 A.S.B.L. Facultes Universitaires Notre-Dame De La Paix Method for obtaining a surface activation of a solid support for building biochips microarrays
AU2001286252A1 (en) * 2000-09-14 2002-03-26 Reverse Proteomics Research Institute Co., Ltd. Method, system, apparatus and device for discovering and preparing chemical compounds
US20040115726A1 (en) * 2001-09-14 2004-06-17 Renpei Nagashima Method, system, apparatus and device for discovering and preparing chemical compounds for medical and other uses.
EP2299256A3 (en) * 2000-09-15 2012-10-10 California Institute Of Technology Microfabricated crossflow devices and methods
GB0022978D0 (en) * 2000-09-19 2000-11-01 Oxford Glycosciences Uk Ltd Detection of peptides
AU2001292959A1 (en) 2000-09-22 2002-04-02 Clontech Laboratories, Inc. Highly sensitive proteomic analysis methods and kits and systems for practicing the same
EP1195606A1 (en) * 2000-10-03 2002-04-10 VBC-Genomics Forschungsges.m.b.H. Allergen-microarray assay
US20040209303A1 (en) 2000-10-03 2004-10-21 Martin Mark T. Methods and compositions for directed microwave chemistry
US7348182B2 (en) * 2000-10-03 2008-03-25 Mirari Biosciences, Inc. Directed microwave chemistry
ATE537264T1 (en) * 2000-10-03 2011-12-15 Mirari Biosciences Inc METHODS AND COMPOSITIONS FOR DIRECTED MICROWAVE CHEMISTRY
US7887885B2 (en) 2000-10-20 2011-02-15 Northwestern University Nanolithography methods and products therefor and produced thereby
AU2002241579A1 (en) 2000-10-24 2002-06-11 Fatemeh Mojtabai Ordered two-and three-dimensional structures of amphiphilic molecules
US20050100951A1 (en) * 2000-10-26 2005-05-12 Biocept, Inc. 3D format biochips and method of use
US7142296B2 (en) * 2000-10-30 2006-11-28 Sru Biosystems, Inc. Method and apparatus for detecting biomolecular interactions
US7153702B2 (en) * 2000-10-30 2006-12-26 Sru Biosystems, Inc. Label-free methods for performing assays using a colorimetric resonant reflectance optical biosensor
US7300803B2 (en) * 2000-10-30 2007-11-27 Sru Biosystems, Inc. Label-free methods for performing assays using a colorimetric resonant reflectance optical biosensor
US20030092075A1 (en) * 2000-10-30 2003-05-15 Sru Biosystems, Llc Aldehyde chemical surface activation processes and test methods for colorimetric resonant sensors
US7875434B2 (en) * 2000-10-30 2011-01-25 Sru Biosystems, Inc. Label-free methods for performing assays using a colorimetric resonant reflectance optical biosensor
US7023544B2 (en) * 2000-10-30 2006-04-04 Sru Biosystems, Inc. Method and instrument for detecting biomolecular interactions
US7615339B2 (en) * 2000-10-30 2009-11-10 Sru Biosystems, Inc. Method for producing a colorimetric resonant reflection biosensor on rigid surfaces
US7202076B2 (en) * 2000-10-30 2007-04-10 Sru Biosystems, Inc. Label-free high-throughput optical technique for detecting biomolecular interactions
US7575939B2 (en) * 2000-10-30 2009-08-18 Sru Biosystems, Inc. Optical detection of label-free biomolecular interactions using microreplicated plastic sensor elements
US7264973B2 (en) * 2000-10-30 2007-09-04 Sru Biosystems, Inc. Label-free methods for performing assays using a colorimetric resonant optical biosensor
US7306827B2 (en) * 2000-10-30 2007-12-11 Sru Biosystems, Inc. Method and machine for replicating holographic gratings on a substrate
US7217574B2 (en) 2000-10-30 2007-05-15 Sru Biosystems, Inc. Method and apparatus for biosensor spectral shift detection
US7094595B2 (en) * 2000-10-30 2006-08-22 Sru Biosystems, Inc. Label-free high-throughput optical technique for detecting biomolecular interactions
US7101660B2 (en) * 2000-10-30 2006-09-05 Sru Biosystems, Inc. Method for producing a colorimetric resonant reflection biosensor on rigid surfaces
US7371562B2 (en) * 2000-10-30 2008-05-13 Sru Biosystems, Inc. Guided mode resonant filter biosensor using a linear grating surface structure
US7175980B2 (en) * 2000-10-30 2007-02-13 Sru Biosystems, Inc. Method of making a plastic colorimetric resonant biosensor device with liquid handling capabilities
DE10054055A1 (en) * 2000-10-31 2002-05-23 Nmi Univ Tuebingen Methods for analyzing proteins
US20050084908A1 (en) * 2000-11-06 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Methods for detecting binding of low-molecular-weight compound and its binding partner molecule
US7232109B2 (en) * 2000-11-06 2007-06-19 California Institute Of Technology Electrostatic valves for microfluidic devices
US6699665B1 (en) 2000-11-08 2004-03-02 Surface Logix, Inc. Multiple array system for integrating bioarrays
US7374906B2 (en) 2000-11-08 2008-05-20 Surface Logix, Inc. Biological assays using gradients formed in microfluidic systems
US7123764B2 (en) 2000-11-08 2006-10-17 Surface Logix Inc. Image processing method for use in analyzing data of a chemotaxis or haptotaxis assay
WO2002040998A2 (en) * 2000-11-17 2002-05-23 Zeptosens Ag Kit and method for determining multiple analytes
JP2002153272A (en) * 2000-11-24 2002-05-28 Inst Of Physical & Chemical Res Biomolecule microarray
US6905816B2 (en) 2000-11-27 2005-06-14 Intelligent Medical Devices, Inc. Clinically intelligent diagnostic devices and methods
EP1397679B1 (en) 2000-11-27 2010-01-20 Intelligent Medical Devices LLC Clinically intelligent diagnostic devices and methods
ATE513854T1 (en) * 2000-12-13 2011-07-15 Bac Ip B V PROTEIN GRID OF VARIABLE DOMAIN OF THE CAMEL IMMUNOGLOBULIN HEAVY CHAIN
FR2818287B1 (en) * 2000-12-14 2003-01-17 Commissariat Energie Atomique SOLID SUPPORT FOR THE IMMOBILIZATION OF OLIGONUCLEOTIDES
WO2002048716A2 (en) 2000-12-14 2002-06-20 Paul Stroobant Differential phage capture proteomics
US6798521B2 (en) * 2000-12-29 2004-09-28 Texas Instruments Incorporated Robust integrated surface plasmon resonance sensor
US7205161B2 (en) * 2001-01-10 2007-04-17 Symyx Technologies, Inc. Polymer brushes for immobilizing molecules to a surface or substrate having improved stability
US20020110904A1 (en) * 2001-01-18 2002-08-15 Nelson Randall W. Integrated system for analysis of biomolecules
US20020169562A1 (en) * 2001-01-29 2002-11-14 Gregory Stephanopoulos Defining biological states and related genes, proteins and patterns
US7297501B2 (en) 2001-02-02 2007-11-20 University Of Pennsylvania Method and devices for running reactions on a target plate for MALDI mass spectrometry
US7332286B2 (en) 2001-02-02 2008-02-19 University Of Pennsylvania Peptide or protein microassay method and apparatus
US7138238B2 (en) * 2001-02-06 2006-11-21 Auburn University Ligand sensor devices and uses thereof
US6913697B2 (en) 2001-02-14 2005-07-05 Science & Technology Corporation @ Unm Nanostructured separation and analysis devices for biological membranes
JP4797196B2 (en) * 2001-02-14 2011-10-19 株式会社 フューエンス Microchip
US20020165675A1 (en) * 2001-03-03 2002-11-07 Golovlev Valeri V. Method and microelectronic device for multi-site molecule detection
WO2002071067A1 (en) * 2001-03-07 2002-09-12 Bio-Rad Laboratories, Inc. Assay system for simultaneous detection and measurement of multiple modified cellular proteins
US20020137106A1 (en) * 2001-03-09 2002-09-26 Ciphergen Biosystems, Inc. Detection of biological pathway components
CA2441086A1 (en) * 2001-03-19 2002-09-26 Wisconsin Alumni Research Foundation Identification of gene expression alterations underlying the aging process in mammals
AU2002306768A1 (en) * 2001-03-20 2002-10-03 Ortho-Clinical Diagnostics, Inc. Expression profiles and methods of use
US20060040286A1 (en) * 2001-03-28 2006-02-23 Nanosphere, Inc. Bio-barcode based detection of target analytes
WO2002079493A2 (en) * 2001-03-29 2002-10-10 Hybrigen, Inc. Improved hybrid gene libraries and uses thereof
US6960437B2 (en) 2001-04-06 2005-11-01 California Institute Of Technology Nucleic acid amplification utilizing microfluidic devices
AU2002258790A1 (en) * 2001-04-10 2002-10-28 The Trustees Of Columbia University In The City Of New York Novel microarrays and methods of use thereof
US6777239B2 (en) * 2001-04-17 2004-08-17 Xenoport, Inc. Epitope-captured antibody display
WO2002085926A2 (en) * 2001-04-19 2002-10-31 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Method for producing stable, regeneratable antibody arrays
KR20030031911A (en) * 2001-04-19 2003-04-23 싸이퍼젠 바이오시스템즈, 인코포레이티드 Biomolecule characterization using mass spectrometry and affinity tags
US20030054408A1 (en) * 2001-04-20 2003-03-20 Ramamoorthi Ravi Methods and systems for identifying proteins
WO2002088388A1 (en) * 2001-04-26 2002-11-07 Ruebben Albert A method and a device for quantification of mutation loads
AU2002340641A1 (en) * 2001-05-03 2002-11-18 Sigma-Genosys, Ltd. Methods for assembling protein microarrays
EP1392342B1 (en) * 2001-05-11 2015-09-23 Yale University Global analysis of protein activities using proteome chips
US9157875B2 (en) * 2001-05-16 2015-10-13 Benjamin P. Warner Drug development and manufacturing
US20080220441A1 (en) 2001-05-16 2008-09-11 Birnbaum Eva R Advanced drug development and manufacturing
US7147687B2 (en) * 2001-05-25 2006-12-12 Nanosphere, Inc. Non-alloying core shell nanoparticles
WO2002096262A2 (en) 2001-05-25 2002-12-05 Northwestern University Non-alloying core shell nanoparticles
US7262063B2 (en) 2001-06-21 2007-08-28 Bio Array Solutions, Ltd. Directed assembly of functional heterostructures
CA2450126A1 (en) * 2001-06-26 2003-01-09 Wisconsin Alumni Research Foundation Gene expression alterations underlying the retardation of aging by caloric restriction in mammals
US6844028B2 (en) * 2001-06-26 2005-01-18 Accelr8 Technology Corporation Functional surface coating
ATE514083T1 (en) * 2001-06-29 2011-07-15 Veri Q Inc METHOD AND COMPOSITIONS FOR DETERMINING THE PURITY AND PURIFICATION OF CHEMICALLY SYNTHESIZED NUCLEIC ACIDS
US20060019235A1 (en) * 2001-07-02 2006-01-26 The Board Of Trustees Of The Leland Stanford Junior University Molecular and functional profiling using a cellular microarray
JP2005514617A (en) * 2001-07-02 2005-05-19 ザ、ボード、オブ、トラスティーズ、オブ、ザ、リーランド、スタンフォード、ジュニア、ユニバーシティ Microarray for cell phenotyping and manipulation
US20030108949A1 (en) * 2001-07-03 2003-06-12 Gang Bao Filtration-based microarray chip
EP1421209A4 (en) * 2001-07-06 2006-04-19 Millipore Corp Satterned composite membrane and stenciling method for the manufacture thereof
US20030013208A1 (en) * 2001-07-13 2003-01-16 Milagen, Inc. Information enhanced antibody arrays
CN1549924A (en) * 2001-07-16 2004-11-24 �����ʸ���˹�ع�˾ Arrays of buffers for analysing biomolecules by their isoelectric point
US7858560B2 (en) * 2001-07-16 2010-12-28 Caprotec Bioanalytics Gmbh Capture compounds, collections thereof and methods for analyzing the proteome and complex compositions
US6444318B1 (en) * 2001-07-17 2002-09-03 Surmodics, Inc. Self assembling monolayer compositions
US7172804B2 (en) * 2001-07-17 2007-02-06 Northwestern University Film-immobilized capture particles
US20030134273A1 (en) * 2001-07-17 2003-07-17 Eric Henderson Combined molecular binding detection through force microscopy and mass spectrometry
JP4264002B2 (en) * 2001-07-17 2009-05-13 バイオ−ラド ラボラトリーズ インコーポレイテッド Latex-based adsorption chip
US7517496B2 (en) * 2001-07-17 2009-04-14 Bio-Rad Laboratories, Inc. Latex based adsorbent chip
US20030143612A1 (en) * 2001-07-18 2003-07-31 Pointilliste, Inc. Collections of binding proteins and tags and uses thereof for nested sorting and high throughput screening
US6977138B2 (en) 2001-07-24 2005-12-20 Massachusetts Institute Of Technology Reactive polymer coatings
EP1279963A1 (en) * 2001-07-27 2003-01-29 Université de Nantes Protein-target screening method using near-infrared fluorescent dyes
EP1281966A3 (en) * 2001-07-30 2003-06-18 Fuji Photo Film Co., Ltd. Method and apparatus for conducting a receptor-ligand reaction
DE60216115T2 (en) * 2001-08-01 2007-05-31 Merck & Co., Inc. BENZIMIDAZO 4,5-föISOCHINOLINONE DERIVATIVES
US7172905B2 (en) * 2001-08-07 2007-02-06 The University Of Chicago Polypeptide immobilization
US20100104631A1 (en) * 2001-08-13 2010-04-29 Lipella Pharmaceuticals Inc. Method of treatment for bladder dysfunction
US20030049626A1 (en) * 2001-08-14 2003-03-13 Milagen, Inc. Antibody-based analysis of matrix protein arrays
US20030040129A1 (en) * 2001-08-20 2003-02-27 Shah Haresh P. Binding assays using magnetically immobilized arrays
US6767731B2 (en) * 2001-08-27 2004-07-27 Intel Corporation Electron induced fluorescent method for nucleic acid sequencing
EP1560939A4 (en) * 2001-08-27 2007-04-25 Surface Logix Inc Immobilization of biological molecules onto surfaces coated with monolayers
US7075162B2 (en) * 2001-08-30 2006-07-11 Fluidigm Corporation Electrostatic/electrostrictive actuation of elastomer structures using compliant electrodes
EP2311934B1 (en) 2001-09-06 2013-06-05 Rapid Micro Biosystems, Inc. Rapid detection of replicating cells
US20030068655A1 (en) * 2001-09-12 2003-04-10 Protiveris, Inc. Microcantilever apparatus and methods for detection of enzymes
DE10145226A1 (en) * 2001-09-13 2003-04-10 Lifebits Ag Manufacture of carrier-bound molecules
DE10145700A1 (en) * 2001-09-17 2003-04-10 Infineon Technologies Ag Biochip arrangement, sensor arrangement and method for operating a biochip arrangement
JP2003099614A (en) * 2001-09-21 2003-04-04 Daiwa Securities Group Inc Holding number inside sale processor, holding number inside sale processing system and program
US7042488B2 (en) 2001-09-27 2006-05-09 Fujinon Corporation Electronic endoscope for highlighting blood vessel
JP4570363B2 (en) * 2001-10-02 2010-10-27 ノースウエスタン ユニヴァーシティ Protein and peptide nanoarrays
US20030073104A1 (en) * 2001-10-02 2003-04-17 Belcher Angela M. Nanoscaling ordering of hybrid materials using genetically engineered mesoscale virus
WO2003031066A1 (en) * 2001-10-11 2003-04-17 California Institute Of Technology Devices utilizing self-assembled gel and method of manufacture
US20050069962A1 (en) 2001-10-12 2005-03-31 Archer Robert M Antibody complexes and methods for immunolabeling
US8323903B2 (en) * 2001-10-12 2012-12-04 Life Technologies Corporation Antibody complexes and methods for immunolabeling
US20040002073A1 (en) 2001-10-15 2004-01-01 Li Alice Xiang Multiplexed analysis of polymorphic loci by concurrent interrogation and enzyme-mediated detection
WO2003104762A2 (en) * 2001-10-23 2003-12-18 Sloan Kettering Institute For Cancer Research Protein micro-arrays and multi-layered affinity interaction detection
US8440093B1 (en) 2001-10-26 2013-05-14 Fuidigm Corporation Methods and devices for electronic and magnetic sensing of the contents of microfluidic flow channels
AU2002367817B2 (en) 2001-11-09 2008-05-29 Nanosphere, Inc. Bioconjugate-nanoparticle probes
CA2467456A1 (en) * 2001-11-19 2003-05-30 Protometrix, Inc. Method of using a non-antibody protein to detect and measure an analyte
EP1527341B1 (en) * 2001-11-20 2009-06-17 Duke University Interfacial biomaterials
WO2003045975A2 (en) * 2001-11-27 2003-06-05 Compound Therapeutics, Inc. Solid-phase immobilization of proteins and peptides
US7691333B2 (en) 2001-11-30 2010-04-06 Fluidigm Corporation Microfluidic device and methods of using same
US6797393B2 (en) 2001-11-30 2004-09-28 Eastman Kodak Company Method for making biochip substrate
US7361310B1 (en) 2001-11-30 2008-04-22 Northwestern University Direct write nanolithographic deposition of nucleic acids from nanoscopic tips
US7118910B2 (en) 2001-11-30 2006-10-10 Fluidigm Corporation Microfluidic device and methods of using same
CA2469403C (en) 2001-12-05 2015-06-02 Jonathan Mark Boutell Protein arrays for allelic variants and uses thereof
US20050112616A1 (en) * 2001-12-10 2005-05-26 William Lee Functionalized materials and libraries thereof
EP1319954A1 (en) * 2001-12-12 2003-06-18 Centre National de Genotypage Methods for protein analysis using protein capture arrays
JP4382339B2 (en) * 2001-12-14 2009-12-09 富士フイルム株式会社 Measuring chip
GB0130318D0 (en) * 2001-12-19 2002-02-06 Univ Leeds Membrane
EP1456659B1 (en) * 2001-12-21 2008-03-12 GE Healthcare Bio-Sciences AB Immobilization of binding agents
KR20030057219A (en) * 2001-12-28 2003-07-04 삼성에스디아이 주식회사 Compound forming intermediated layer on matrix, composition for intermediated layer, and biochip using the same
KR100450202B1 (en) * 2002-01-07 2004-09-24 삼성에스디아이 주식회사 A method for forming a pattern of functional group for immobilizing physiological material
US20070178477A1 (en) * 2002-01-16 2007-08-02 Nanomix, Inc. Nanotube sensor devices for DNA detection
US20060228723A1 (en) * 2002-01-16 2006-10-12 Keith Bradley System and method for electronic sensing of biomolecules
US7056665B2 (en) * 2002-01-16 2006-06-06 Panomics, Inc. Screening methods involving the detection of short-lived proteins
AU2003210546A1 (en) * 2002-01-16 2003-09-02 Panomics, Inc. Methods for isolating and characterizing short-lived proteins and arrays derived therefrom
US20030134433A1 (en) * 2002-01-16 2003-07-17 Nanomix, Inc. Electronic sensing of chemical and biological agents using functionalized nanostructures
CA2472030A1 (en) * 2002-01-24 2003-07-31 Pointilliste, Inc. Use of collections of binding sites for sample profiling and other applications
SE0200269D0 (en) * 2002-01-31 2002-01-31 Ellem Bioteknik Ab Material for implantation
JP2003222589A (en) * 2002-01-31 2003-08-08 Communication Research Laboratory Dual-wavelength surface plasmon resonance spectroscopic device
WO2003066906A2 (en) * 2002-02-07 2003-08-14 Eastern Virginia Medical School Of The Medical College Of Hampton Roads Diagnostic microarray and method of use thereof
AU2003280177A1 (en) * 2002-02-21 2004-05-13 Sense Proteomic Limited Enzyme array and assay
EP1489167A4 (en) * 2002-03-01 2006-06-07 Nat Inst Of Advanced Ind Scien Immobilized cells and liposomes and method of immobilizing the same
US20050266455A1 (en) * 2002-03-02 2005-12-01 Sci Tec, Inc. Method and microelectronic device for multi-site molecule detection
US6815078B2 (en) 2002-03-06 2004-11-09 Eastman Kodak Company Substrate for protein microarray containing functionalized polymer
IL163880A0 (en) * 2002-03-07 2005-12-18 Zephyr Proteomix Ltd Microarrays of cellulose binding chimeric proteinsand methods of use thereof
AU2003224674A1 (en) * 2002-03-11 2003-09-29 Hk Pharmaceuticals, Inc. Compounds and methods for analyzing the proteome
US6703216B2 (en) 2002-03-14 2004-03-09 The Regents Of The University Of California Methods, compositions and apparatuses for detection of gamma-hydroxybutyric acid (GHB)
US20030228709A1 (en) * 2002-03-25 2003-12-11 Kozlowski Roland Zbignieiw Arrays
JP2005521425A (en) 2002-04-01 2005-07-21 フルイディグム コーポレイション Microfluidic particle analysis system
US20030215858A1 (en) * 2002-04-08 2003-11-20 Baylor College Of Medicine Enhanced gene expression system
US20030194709A1 (en) * 2002-04-10 2003-10-16 Xing Yang Hydrophobic zone device
US20040033546A1 (en) * 2002-04-10 2004-02-19 The Trustees Of Columbia University In The City Of New York Novel microarrays and methods of use thereof
US7687256B2 (en) * 2002-04-11 2010-03-30 Spire Corporation Surface activated biochip
AU2003267951A1 (en) * 2002-04-26 2003-12-19 Genencor International, Inc. Methods and articles for high throughput analysis of biomolecular interactions
US7445894B2 (en) * 2002-05-03 2008-11-04 Molecular Probes, Inc. Compositions and methods for detection and isolation of phosphorylated molecules
US20040171034A1 (en) 2002-05-03 2004-09-02 Brian Agnew Compositions and methods for detection and isolation of phosphorylated molecules
CA2483868A1 (en) * 2002-05-03 2004-05-21 Molecular Probes, Inc. Compositions and methods for detection and isolation of phosphorylated molecules
US20030211488A1 (en) 2002-05-07 2003-11-13 Northwestern University Nanoparticle probs with Raman spectrocopic fingerprints for analyte detection
US20030211478A1 (en) * 2002-05-08 2003-11-13 Gentel Corporation Transcription factor profiling on a solid surface
US20030208936A1 (en) * 2002-05-09 2003-11-13 Lee Charles Hee Method for manufacturing embroidery decorated cards and envelopes
US7618788B2 (en) * 2002-05-10 2009-11-17 Millipore Corporation Proteome epitope tags and methods of use thereof in protein modification analysis
US20060014212A1 (en) * 2002-05-10 2006-01-19 Epitome Biosystems, Inc. Proteome epitope tags and methods of use thereof in protein modification analysis
CA2485560A1 (en) * 2002-05-10 2004-06-03 Engeneos, Inc. Unique recognition sequences and methods of use thereof in protein analysis
US7460960B2 (en) * 2002-05-10 2008-12-02 Epitome Biosystems, Inc. Proteome epitope tags and methods of use thereof in protein modification analysis
EP1534755B1 (en) * 2002-05-10 2011-10-12 Bio-Layer Pty Limited Generation of surface coating diversity
EP1364710B1 (en) * 2002-05-13 2009-10-07 Becton, Dickinson and Company Self-aliquoting sample storage plate
US8785179B2 (en) * 2002-05-22 2014-07-22 Texas Instruments Incorporated Biosensor and method
US20050239193A1 (en) * 2002-05-30 2005-10-27 Bioforce Nanosciences, Inc. Device and method of use for detection and characterization of microorganisms and microparticles
US20040091498A1 (en) * 2002-05-31 2004-05-13 Aaron Diamond Aids Research Center Defensins: use as antiviral agents
EP1369692A3 (en) * 2002-06-04 2003-12-17 Interuniversitaire Microelectronica Centrum vzw ( IMEC) Sensor surface
EP1413886A1 (en) * 2002-10-25 2004-04-28 Interuniversitair Microelektronica Centrum ( Imec) Sensor surface
US7135343B2 (en) * 2002-06-17 2006-11-14 Agilent Technologies, Inc. Biomolecule resistant and their methods of use in assays
CA2490522A1 (en) * 2002-06-20 2003-12-31 Paul Stroobant Improved methods for performing differential capture proteomics
US7948041B2 (en) 2005-05-19 2011-05-24 Nanomix, Inc. Sensor having a thin-film inhibition layer
US7253277B2 (en) * 2002-07-02 2007-08-07 Nanosphere, Inc. Nanoparticle polyanion conjugates and methods of use thereof in detecting analytes
USH2223H1 (en) * 2002-07-11 2008-09-02 The United States Of America As Represented By The Secretary Of The Navy Patterned, micrometer-sized antibody features
US20040009528A1 (en) * 2002-07-11 2004-01-15 Shyh-Yu Shaw Protein chips
US20050008828A1 (en) * 2002-07-25 2005-01-13 Trustees Of Stevens Institute Of Technology Patterned polymer microgel and method of forming same
JP4099822B2 (en) * 2002-07-26 2008-06-11 セイコーエプソン株式会社 Dispensing device, dispensing method, and biological sample-containing solution ejection failure detection method
CA2833718C (en) * 2002-07-29 2017-01-03 Applied Biomimetic A/S Biomimetic membrane suitable for use in a solar cell
US20050074898A1 (en) * 2002-07-31 2005-04-07 Caliper Technologies Corp. High density reagent array preparation methods
US20040067597A1 (en) * 2002-07-31 2004-04-08 Caliper Technologies Corp. High density reagent array preparation methods
EP1540344A4 (en) * 2002-08-02 2007-02-21 Applera Corp Fluorescence polarization assay
GB2391867A (en) * 2002-08-13 2004-02-18 Smartbead Technologies Ltd Analysis system using coded supports
AU2003255253A1 (en) * 2002-08-13 2004-02-25 Discovery Partners International Spotting pattern for placement of compounds in an array
AU2003259041A1 (en) * 2002-08-15 2004-03-03 Proteoplex, Inc. Methods and apparatus for preparing and assaying biological samples to determine protein concentration
JP2005535898A (en) * 2002-08-16 2005-11-24 ザイオミックス インコーポレーテッド Methods and reagents for surface functionalization
EP1546723A4 (en) * 2002-08-16 2007-03-07 Decision Biomarkers Inc Reading of fluorescent arrays
US7384742B2 (en) * 2002-08-16 2008-06-10 Decision Biomarkers, Inc. Substrates for isolating reacting and microscopically analyzing materials
WO2004020065A2 (en) * 2002-08-28 2004-03-11 Mt Technologies, Inc. Microfluidic affinity system using polydimethylsiloxane and a surface modification process
AU2003299534A1 (en) * 2002-08-29 2004-05-13 Bioscale, Inc. Resonant sensor and sensing system
ATE520027T1 (en) * 2002-09-03 2011-08-15 Bayer Technology Services Gmbh ANALYTICAL PLATFORM AND DETECTION METHOD WITH ANALYTES TO BE DETECTED IN A SAMPLE AS IMMOBILIZED SPECIFIC BONDING PARTNERS, IF APPLICABLE AFTER FRACTIONATION
US20040043508A1 (en) * 2002-09-03 2004-03-04 Frutos Anthony G. Polymer-coated substrates for binding biological molecules
EP1561109A1 (en) * 2002-09-03 2005-08-10 Zeptosens AG Analytical platform and detection method with analytes which are to be detected in a sample in the form of immobilized specific binding partners
US7429492B2 (en) * 2002-09-09 2008-09-30 Sru Biosystems, Inc. Multiwell plates with integrated biosensors and membranes
US7927822B2 (en) * 2002-09-09 2011-04-19 Sru Biosystems, Inc. Methods for screening cells and antibodies
EP1548437A4 (en) * 2002-09-17 2006-06-07 Olympus Corp Method and apparatus for arranging liquid reaction components on substrate surface for detecting target substance by reaction among plural reaction components on substrate and article utilized in the method
US20050064508A1 (en) * 2003-09-22 2005-03-24 Semzyme Peptide mediated synthesis of metallic and magnetic materials
JP2004105070A (en) * 2002-09-18 2004-04-08 Inst Of Physical & Chemical Res Method for producing ligand-bonded protein using cell-free protein synthetic system and use thereof
US7960184B2 (en) * 2002-09-20 2011-06-14 George Mason Intellectual Properties, Inc. Methods and devices for active bioassay
US7143785B2 (en) 2002-09-25 2006-12-05 California Institute Of Technology Microfluidic large scale integration
EP1546412B1 (en) 2002-10-02 2014-05-21 California Institute Of Technology Microfluidic nucleic acid analysis
US20040137526A1 (en) * 2002-10-15 2004-07-15 The Regents Of The University Of Michigan Multidimensional protein separation system
CN1726394B (en) * 2002-10-15 2010-10-13 阿伯麦特里科斯公司 Sets of digital antibodies directed against short epitopes, and methods using same
DE10249608A1 (en) * 2002-10-18 2004-05-06 Gkss-Forschungszentrum Geesthacht Gmbh Device and method for structural analysis and detection of complex glycostructures
US20040081969A1 (en) * 2002-10-29 2004-04-29 Ilsley Diane D. Devices and methods for evaulating the quality of a sample for use in an array assay
AU2003291677A1 (en) * 2002-10-30 2004-05-25 Pointilliste, Inc. Methods for producing polypeptide-tagged collections and capture systems containing the tagged polypeptides
US20030153013A1 (en) * 2002-11-07 2003-08-14 Ruo-Pan Huang Antibody-based protein array system
EP1560917A1 (en) * 2002-11-08 2005-08-10 University of Copenhagen Method of immobilising a protein to a zeolite
US7526114B2 (en) 2002-11-15 2009-04-28 Bioarray Solutions Ltd. Analysis, secure access to, and transmission of array images
US20040096914A1 (en) * 2002-11-20 2004-05-20 Ye Fang Substrates with stable surface chemistry for biological membrane arrays and methods for fabricating thereof
AU2003300859A1 (en) * 2002-12-11 2004-06-30 New England Biolabs, Inc. Carrier-ligand fusions and uses thereof
JP2006517786A (en) * 2002-12-12 2006-08-03 ナノスフェアー インコーポレイテッド Direct SNP detection using unamplified DNA
US7785782B2 (en) * 2002-12-12 2010-08-31 Novartis Vaccines And Diagnostics, Inc. Device and method for in-line blood testing using biochips
EP1573044A4 (en) 2002-12-18 2006-07-05 Ciphergen Biosystems Inc Serum biomarkers in lung cancer
US20040121339A1 (en) * 2002-12-19 2004-06-24 Jizhong Zhou Special film-coated substrate for bio-microarray fabrication and use thereof
AU2003300389B2 (en) * 2002-12-22 2009-10-08 The Scripps Research Institute Protein arrays
US7785601B2 (en) 2002-12-31 2010-08-31 Sygnis Bioscience Gmbh & Co. Kg Methods of treating neurological conditions with hematopoietic growth factors
US7695723B2 (en) 2002-12-31 2010-04-13 Sygnis Bioscience Gmbh & Co. Kg Methods of treating neurological conditions with hematopoietic growth factors
JP2006512583A (en) * 2003-01-02 2006-04-13 バイオフォース ナノサイエンシズ インコーポレイテッド Method and apparatus for molecular analysis in small sample volumes
KR100523212B1 (en) * 2003-01-04 2005-10-24 한국과학기술원 A Protein Chip for Analyzing Interaction Between Protein and Substrate Peptide Therefor
US7736909B2 (en) * 2003-01-09 2010-06-15 Board Of Regents, The University Of Texas System Methods and compositions comprising capture agents
US7619060B2 (en) * 2003-01-10 2009-11-17 Protein Crystal Co., Ltd. Cytoplasmic polyhedrosis virus protein complex of a polyhedrin and a VP3 polypeptide
US7422865B2 (en) * 2003-01-13 2008-09-09 Agilent Technologies, Inc. Method of identifying peptides in a proteomic sample
US20040137158A1 (en) * 2003-01-15 2004-07-15 Kools Jacques Constant Stefan Method for preparing a noble metal surface
WO2004064972A2 (en) * 2003-01-16 2004-08-05 Hk Pharmaceuticals, Inc. Capture compounds, collections thereof and methods for analyzing the proteome and complex compositions
KR100994566B1 (en) * 2003-01-20 2010-11-15 삼성전자주식회사 An array device comprising a photoresist film having immobilization regions and a method using the same
AU2003290561A1 (en) * 2003-02-10 2004-09-06 Dana Ault-Riche Self-assembling arrays and uses thereof
US20050008851A1 (en) * 2003-02-18 2005-01-13 Fuji Photo Film Co., Ltd. Biosensor
WO2004076344A2 (en) * 2003-02-25 2004-09-10 Yeda Research And Development Company Ltd. Nanoscopic structure and devices using the same
WO2004075855A2 (en) 2003-02-26 2004-09-10 Biomed Solutions, Llc Process for in vivo treatment of specific biological targets in bodily fluid
US20050130174A1 (en) * 2003-02-27 2005-06-16 Nanosphere, Inc. Label-free gene expression profiling with universal nanoparticle probes in microarray assay format
CN1514243A (en) * 2003-04-30 2004-07-21 成都夸常科技有限公司 Method of preceeding qualitative and lor quantitative analysis against target substance its device and marker and detecting reagent box
EP1606419A1 (en) 2003-03-18 2005-12-21 Quantum Genetics Ireland Limited Systems and methods for improving protein and milk production of dairy herds
US20040185445A1 (en) * 2003-03-19 2004-09-23 Ye Fang Universal readout for target identification using biological microarrays
US8068990B2 (en) * 2003-03-25 2011-11-29 Hologic, Inc. Diagnosis of intra-uterine infection by proteomic analysis of cervical-vaginal fluids
US7191068B2 (en) * 2003-03-25 2007-03-13 Proteogenix, Inc. Proteomic analysis of biological fluids
WO2004087323A1 (en) * 2003-03-28 2004-10-14 Mergen Ltd. Multi-array systems and methods of use thereof
US20040197841A1 (en) * 2003-04-02 2004-10-07 Apffel James Alexander Methods and reagents for multiplexed analyses
US7476363B2 (en) 2003-04-03 2009-01-13 Fluidigm Corporation Microfluidic devices and methods of using same
US7604965B2 (en) 2003-04-03 2009-10-20 Fluidigm Corporation Thermal reaction device and method for using the same
US8828663B2 (en) 2005-03-18 2014-09-09 Fluidigm Corporation Thermal reaction device and method for using the same
US20050145496A1 (en) 2003-04-03 2005-07-07 Federico Goodsaid Thermal reaction device and method for using the same
CA2521171C (en) 2003-04-03 2013-05-28 Fluidigm Corp. Microfluidic devices and methods of using same
US20040248205A1 (en) * 2003-04-16 2004-12-09 Stern Lawrence J. Major histocompatibility complex (MHC)-peptide arrays
JP2006527190A (en) 2003-04-17 2006-11-30 サイファージェン バイオシステムズ インコーポレイテッド Polypeptides related to natriuretic peptides and their identification and use
WO2004094989A2 (en) * 2003-04-22 2004-11-04 Ciphergen Biosystems, Inc. Methods of host cell protein analysis
WO2004097368A2 (en) * 2003-04-28 2004-11-11 Ciphergen Biosystems, Inc. Improved immunoassays
US7425700B2 (en) 2003-05-22 2008-09-16 Stults John T Systems and methods for discovery and analysis of markers
KR100547015B1 (en) * 2003-05-23 2006-01-26 주식회사 올메디쿠스 Biosensor for analyzing quantitatively analyte with a predetermined size and larger than, and manufacturing method thereof
US20050250094A1 (en) * 2003-05-30 2005-11-10 Nanosphere, Inc. Method for detecting analytes based on evanescent illumination and scatter-based detection of nanoparticle probe complexes
US20060141559A1 (en) * 2003-06-10 2006-06-29 Takashi Suzuki Extract from cultured mammalian cell, process for preparation thereof and method of cell-free protein synthesis using the extract
WO2004113872A2 (en) * 2003-06-24 2004-12-29 The Trustees Of Columbia University In The City Of New York Covalent methods for immobilization of thiolated biomolecules on siliceous and metallic surfaces
US20040265921A1 (en) * 2003-06-30 2004-12-30 National University Of Singapore Intein-mediated attachment of ligands to proteins for immobilization onto a support
WO2005007677A2 (en) * 2003-07-10 2005-01-27 The Institute For Systems Biology Affinity capture of peptides by microarray and related methods
WO2005067425A2 (en) * 2003-07-10 2005-07-28 Polytechnic University Bacterial biosensors
EP1660858A4 (en) * 2003-07-21 2007-10-24 Amplified Proteomics Inc Multiplexed analyte detection
US20060014003A1 (en) * 2003-07-24 2006-01-19 Libera Matthew R Functional nano-scale gels
US20050059054A1 (en) 2003-07-25 2005-03-17 Richard Conrad Methods and compositions for preparing RNA from a fixed sample
US20050059024A1 (en) 2003-07-25 2005-03-17 Ambion, Inc. Methods and compositions for isolating small RNA molecules
WO2005014805A1 (en) * 2003-08-08 2005-02-17 Regents Of The University Of Minnesota A structured material for the production of hydrogen
AU2004263896A1 (en) 2003-08-08 2005-02-17 Genenews Inc. Osteoarthritis biomarkers and uses thereof
US7413712B2 (en) 2003-08-11 2008-08-19 California Institute Of Technology Microfluidic rotary flow reactor matrix
US7223609B2 (en) * 2003-08-14 2007-05-29 Agilent Technologies, Inc. Arrays for multiplexed surface plasmon resonance detection of biological molecules
AU2004267802B2 (en) * 2003-08-18 2011-03-17 Tethys Bioscience, Inc. Methods for reducing complexity of a sample using small epitope antibodies
US7754497B2 (en) * 2003-08-29 2010-07-13 Reverse Proteomics Research Institute Co., Ltd. Method for immobilizing proteins
US20070092964A1 (en) 2003-09-03 2007-04-26 Zymoyx, Inc. Ion detection using a pillar chip
US20050176026A1 (en) * 2003-09-05 2005-08-11 Franck Carl P. Liquid mixing reactor for biochemical assays
US20050053949A1 (en) * 2003-09-08 2005-03-10 Silin Vitalii Ivanovich Biochip for proteomics applications
US20050059083A1 (en) * 2003-09-15 2005-03-17 Becton Dickinson And Company High throughput method to identify ligands for cell attachment
US7927796B2 (en) 2003-09-18 2011-04-19 Bioarray Solutions, Ltd. Number coding for identification of subtypes of coded types of solid phase carriers
US7998435B2 (en) 2003-09-19 2011-08-16 Life Technologies Corporation High density plate filler
KR100518953B1 (en) * 2003-09-19 2005-10-12 주식회사 제노포커스 Method for Whole Surrounding Surface Display of Target Proteins Using Exosporium of Bacillus cereus Group
US9492820B2 (en) 2003-09-19 2016-11-15 Applied Biosystems, Llc High density plate filler
US7407630B2 (en) 2003-09-19 2008-08-05 Applera Corporation High density plate filler
US7695688B2 (en) * 2003-09-19 2010-04-13 Applied Biosystems, Llc High density plate filler
US8277760B2 (en) 2003-09-19 2012-10-02 Applied Biosystems, Llc High density plate filler
US8298780B2 (en) * 2003-09-22 2012-10-30 X-Body, Inc. Methods of detection of changes in cells
WO2005031305A2 (en) 2003-09-22 2005-04-07 Bioarray Solutions, Ltd. Surface immobilized polyelectrolyte with multiple functional groups capable of covalently bonding to biomolecules
US20070017870A1 (en) * 2003-09-30 2007-01-25 Belov Yuri P Multicapillary device for sample preparation
US7166212B2 (en) * 2003-09-30 2007-01-23 Chromba, Inc. Multicapillary column for chromatography and sample preparation
US20050069949A1 (en) * 2003-09-30 2005-03-31 International Business Machines Corporation Microfabricated Fluidic Structures
US20050069462A1 (en) * 2003-09-30 2005-03-31 International Business Machines Corporation Microfluidics Packaging
US20050079507A1 (en) * 2003-10-09 2005-04-14 Ye Fang Target evaluation using biological membrane arrays
WO2005047851A2 (en) * 2003-10-15 2005-05-26 The Trustees Of Columbia University In The City Of New York Device for measuring nanometer level pattern-dependent binding reactions
US20050084981A1 (en) * 2003-10-16 2005-04-21 Magdalena Ostrowski Method of depositing a bioactive material on a substrate
US20050112650A1 (en) * 2003-10-20 2005-05-26 Ciphergen Biosystems, Inc. Reactive polyurethane-based polymers
WO2005040800A1 (en) * 2003-10-23 2005-05-06 Consejo Superior De Investigaciones Científicas Method for producing and using a new protein array, said protein array and the applications thereof
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
US7563569B2 (en) 2003-10-28 2009-07-21 Michael Seul Optimization of gene expression analysis using immobilized capture probes
JP2007509629A (en) 2003-10-29 2007-04-19 バイオアレイ ソリューションズ リミテッド Complex nucleic acid analysis by cleavage of double-stranded DNA
US20050095648A1 (en) * 2003-10-30 2005-05-05 Mario Geysen Method for designing linear epitopes and algorithm therefor and polypeptide epitopes
CA2544836A1 (en) * 2003-11-06 2005-05-26 Guo Bin Wang High-density amine-functionalized surface
JP4796967B2 (en) 2003-11-07 2011-10-19 ヴァーミリオン インコーポレイテッド Biomarkers for Alzheimer's disease
US20060007515A1 (en) * 2003-11-13 2006-01-12 Dmitri Simonian Surface lubrication in microstructures
US7153896B2 (en) 2003-11-14 2006-12-26 Eastman Kodak Company Element for protein microarrays
US7332355B2 (en) 2003-11-18 2008-02-19 California Institute Of Technology Method and compositions for the detection of protein glycosylation
CA2545653C (en) 2003-11-21 2014-07-08 Anp Technologies, Inc. Asymmetrically branched polymer conjugates and microarray assays
US20090246889A1 (en) * 2003-11-22 2009-10-01 Samsung Electronics Co., Ltd. Substrate having oxide layer, method for detecting target material using the substrate, and optical sensor including the substrate
KR100580631B1 (en) * 2003-11-22 2006-05-16 삼성전자주식회사 A substrate having an oxide layer, method for detecting a target substance using the same and optical sensor containing the same
US20050109622A1 (en) * 2003-11-26 2005-05-26 Peter Peumans Method for controlling electrodeposition of an entity and devices incorporating the immobilized entity
CA2547861A1 (en) 2003-12-05 2005-06-23 Ciphergen Biosystems, Inc. Serum biomarkers for chagas disease
US7881871B2 (en) * 2003-12-12 2011-02-01 Bio-Layer Pty Limited Method for designing surfaces
US20050170445A1 (en) * 2004-01-07 2005-08-04 Duke University Methods of establishing profiles for use in evaluating wound healing and biocompatibility of implant materials and microarrays useful therefor
US20060018911A1 (en) * 2004-01-12 2006-01-26 Dana Ault-Riche Design of therapeutics and therapeutics
US20050208597A1 (en) * 2004-01-26 2005-09-22 The Board Of Trustees Of The Leland Stanford Junior University Microarray analysis of post-translational modifications
JP2005204609A (en) * 2004-01-26 2005-08-04 Canon Inc Kit for immobilizing organic material, structure in which organic material is immobilized and method for producing the same structure
EP1562046A1 (en) * 2004-02-03 2005-08-10 B.R.A.H.M.S Aktiengesellschaft Method of diagnosing sepsis by detecting selectively the concentration of superoxide dismutase 1 (SOD-1) in samples
WO2005112544A2 (en) 2004-02-19 2005-12-01 The Governors Of The University Of Alberta Leptin promoter polymorphisms and uses thereof
JP2005269902A (en) * 2004-03-22 2005-10-06 Seiko Epson Corp Method for immobilizing cell on solid-phase surface
US7276283B2 (en) * 2004-03-24 2007-10-02 Wisconsin Alumni Research Foundation Plasma-enhanced functionalization of carbon-containing substrates
US7723126B2 (en) * 2004-03-24 2010-05-25 Wisconsin Alumni Research Foundation Plasma-enhanced functionalization of inorganic oxide surfaces
CN100357738C (en) * 2004-03-26 2007-12-26 博奥生物有限公司 Method of detecting small molecule compound and its special biochip
JP2005312425A (en) * 2004-03-31 2005-11-10 Toyo Kohan Co Ltd Method for immobilizing polypeptide and solid support having polypeptide immobilized thereon, method for detection and purification of polypeptide using the same and solid support for immobilizing polypeptide
US7371331B2 (en) * 2004-04-01 2008-05-13 Valerie J Marty Method of creating a patterned monolayer on a surface
JP4451193B2 (en) * 2004-04-12 2010-04-14 大日本印刷株式会社 Method for producing pattern forming body
EP1745149A4 (en) 2004-04-15 2008-08-06 Univ Florida Neural proteins as biomarkers for nervous system injury and other neural disorders
BRPI0510266A (en) 2004-04-26 2007-10-30 Childrens Medical Center methods for detecting an angiogenic disease or disorder in an individual, for detecting cancer in an individual, for treating an individual affected with an angiogenic disease or disorder, for determining the likelihood of efficacy of an anti-angiogenic therapy , to determine the efficacy of a test therapy in modulating the levels of angiogenic platelet regulators, to create a platelet record or profile for an angiogenic disease or disorder, and to monitor the efficacy of a therapy in an individual with an angiogenic disease or disorder
WO2005106472A1 (en) * 2004-04-28 2005-11-10 Japan Science And Technology Agency Biochip producing method, biochip, biochip analyzing device, biochip analyzing method
WO2005118810A1 (en) 2004-06-03 2005-12-15 Athlomics Pty Ltd Agents and methods for diagnosing stress
US20060251795A1 (en) * 2005-05-05 2006-11-09 Boris Kobrin Controlled vapor deposition of biocompatible coatings for medical devices
DE102004031258A1 (en) * 2004-06-29 2006-02-09 Jennissen, Herbert P., Prof. Dr. Protein hybrids with polyhydroxyaromatic amino acid epitopes
AU2005259833B2 (en) 2004-07-02 2011-09-22 Anteo Technologies Pty Ltd Use of metal complexes
US20060009623A1 (en) * 2004-07-06 2006-01-12 National University Of Singapore C-terminal attachment of ligands to proteins for immobilization onto a support
US20060014155A1 (en) * 2004-07-16 2006-01-19 Wisconsin Alumni Research Foundation Methods for the production of sensor arrays using electrically addressable electrodes
WO2006007664A1 (en) * 2004-07-22 2006-01-26 Genomics Research Partners Pty Ltd Agents and methods for diagnosing osteoarthritis
EP1789437A4 (en) * 2004-07-30 2008-11-05 Sinai School Medicine Npc1l1 and npc1l1 inhibitors and methods of use thereof
US7848889B2 (en) 2004-08-02 2010-12-07 Bioarray Solutions, Ltd. Automated analysis of multiplexed probe-target interaction patterns: pattern matching and allele identification
KR20070061808A (en) * 2004-08-04 2007-06-14 악셀라 바이오센서스 인코포레이티드 Patterned surfaces with chemical crosslinkers for use in diffraction-based sensing
US20060040377A1 (en) * 2004-08-17 2006-02-23 Biocept, Inc. Protein microarrays
US20130040840A1 (en) * 2004-09-02 2013-02-14 Bioarray Solutions, Ltd. Nucleic acid amplification with integrated multiplex detection
US20060051348A1 (en) 2004-09-09 2006-03-09 Jorn Gorlach Method of producing a plurality of isolated antibodies to a plurality of cognate antigens
US20060052948A1 (en) * 2004-09-09 2006-03-09 Jorn Gorlach Method of identifying drugs, targeting moieties or diagnostics
WO2006034427A2 (en) * 2004-09-20 2006-03-30 Proteogenix, Inc. Diagnosis of fetal aneuploidy
ITVR20040149A1 (en) * 2004-09-22 2004-12-22 Sanitaria Scaligera Spa RAPID MONITORING SYSTEM OF THE BLOOD GROUP AND FOR THE DETECTION OF IMMUNOHEMATOLOGICAL REACTIONS
US7592139B2 (en) * 2004-09-24 2009-09-22 Sandia National Laboratories High temperature flow-through device for rapid solubilization and analysis
JP2006095452A (en) * 2004-09-30 2006-04-13 Fuji Photo Film Co Ltd Spin coat film forming method
WO2006041392A1 (en) * 2004-10-13 2006-04-20 Biacore Ab Preparation and use of a reactive solid support surface
SE0402476D0 (en) * 2004-10-13 2004-10-13 Biacore Ab Preparation and use of a reactive solid support surface
WO2006047417A2 (en) 2004-10-21 2006-05-04 University Of Florida Research Foundation, Inc. Detection of cannabinoid receptor biomarkers and uses thereof
US20110077164A1 (en) * 2004-10-23 2011-03-31 Andras Guttman Expression profiling platform technology
US20070293437A1 (en) * 2004-10-23 2007-12-20 Andras Guttman Expression Profiling Platform Technology
WO2006049498A1 (en) * 2004-11-05 2006-05-11 Modiquest B.V. Means and methods for isolating and/or identifying a target molecule
US20060246467A1 (en) * 2004-11-15 2006-11-02 California Institute Of Technology Biomarker sensors and method for multi-color imaging and processing of single-molecule life signatures
US20060223195A1 (en) * 2004-11-16 2006-10-05 Meyer Grant D Stress based removal of nonspecific binding from surfaces
US7745143B2 (en) * 2004-11-19 2010-06-29 Plexera, Llc Plasmon resonance biosensor and method
ATE390562T1 (en) * 2004-11-19 2008-04-15 Ebm Papst St Georgen Gmbh & Co ARRANGEMENT WITH A FAN AND A PUMP
WO2006062427A1 (en) * 2004-11-24 2006-06-15 Institut Molekulyarnoi Biologii Im. V.A.Engelgardta Rossiiskoi Akademii Nauk Method for quantitatively detecting biological toxins
ES2416207T3 (en) * 2004-11-29 2013-07-30 Centre National De La Recherche Scientifique Trichlorosilyl alkyl isocyanate molecule and surface modified mineral substrate
AU2005313995A1 (en) * 2004-12-08 2006-06-15 Lyotropic Therapeutics, Inc. Compositions for binding to assay substrata and methods of using
US20080003599A1 (en) * 2004-12-28 2008-01-03 Dary Ekaterina L Biological Microchip for Multiple Parallel Immunoassay of Compounds and Immunoassay Metods Using Said Microchip
US7811772B2 (en) * 2005-01-06 2010-10-12 Eastern Virginia Medical School Apolipoprotein A-II isoform as a biomarker for prostate cancer
JP4736439B2 (en) * 2005-01-25 2011-07-27 東レ株式会社 Nucleic acid immobilization carrier
DE602006018578D1 (en) 2005-01-28 2011-01-13 Childrens Medical Center DIAGNOSIS AND PROGNOSIS OF BUBBLE CANCER.
US7396689B2 (en) * 2005-02-04 2008-07-08 Decision Biomarkers Incorporated Method of adjusting the working range of a multi-analyte assay
CA2599589A1 (en) 2005-02-07 2006-08-17 Genenews,Inc. Mild osteoarthritis biomarkers and uses thereof
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
US20060234265A1 (en) * 2005-03-21 2006-10-19 Jim Richey Microarrays having multi-functional, compartmentalized analysis areas and methods of use
JP4435709B2 (en) * 2005-03-22 2010-03-24 富士フイルム株式会社 Biosensor
US20060257902A1 (en) * 2005-03-25 2006-11-16 Ambion, Inc. Methods and compositions for depleting abundant RNA transcripts
US8048638B2 (en) 2005-04-01 2011-11-01 University Of Florida Research Foundation, Inc. Biomarkers of liver injury
CA2635198C (en) 2005-04-01 2019-11-12 University Of Florida Research Foundation, Inc Biomarkers of liver injury
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
US7648844B2 (en) * 2005-05-02 2010-01-19 Bioscale, Inc. Method and apparatus for detection of analyte using an acoustic device
CA2606815C (en) 2005-05-02 2017-03-07 Anp Technologies, Inc. Polymer conjugate enhanced bioassays
US7300631B2 (en) * 2005-05-02 2007-11-27 Bioscale, Inc. Method and apparatus for detection of analyte using a flexural plate wave device and magnetic particles
US7749445B2 (en) * 2005-05-02 2010-07-06 Bioscale, Inc. Method and apparatus for analyzing bioprocess fluids
US7611908B2 (en) * 2005-05-02 2009-11-03 Bioscale, Inc. Method and apparatus for therapeutic drug monitoring using an acoustic device
US20070003954A1 (en) * 2005-05-12 2007-01-04 The Board Of Regents Of The University Of Texas System Protein and antibody profiling using small molecule microarrays
US8486629B2 (en) 2005-06-01 2013-07-16 Bioarray Solutions, Ltd. Creation of functionalized microparticle libraries by oligonucleotide ligation or elongation
US20070021433A1 (en) 2005-06-03 2007-01-25 Jian-Qiang Fan Pharmacological chaperones for treating obesity
NZ593388A (en) * 2005-06-08 2012-08-31 Dana Farber Cancer Inst Inc Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
WO2006138479A2 (en) * 2005-06-15 2006-12-28 Arizona Board Of Regents For And On Behalf Of Arizona State University Microstructure and microdomain microarrays, methods of making and using thereof
WO2007002567A2 (en) * 2005-06-23 2007-01-04 Nanosphere, Inc. Selective isolation and concentration of nucleic acids from complex samples
EP1904852A2 (en) 2005-06-24 2008-04-02 Ciphergen Biosystems, Inc. Biomarkers for ovarian cancer: beta-2 microglobulin
EP2476761A3 (en) 2005-07-07 2012-10-17 Athlomics Pty Ltd Polynucleotide marker genes and their expression, for diagnosis of endotoxemia
EP1917347B1 (en) 2005-08-22 2015-10-07 Cornell Research Foundation, Inc. Compositions and methods for analyzing protein interactions
US7464580B2 (en) * 2005-09-26 2008-12-16 Oakland University Ionic liquid high temperature gas sensors
CA2623408C (en) 2005-09-26 2014-04-01 Rapid Micro Biosystems, Inc. Cassette containing growth medium
FR2891279B1 (en) * 2005-09-27 2007-12-14 Centre Nat Rech Scient NEW CHIPS FOR SURFACE PLASMON DETECTION (SPR)
WO2007047408A2 (en) * 2005-10-12 2007-04-26 Pathologica, Llc. Promac signature application
US20070141627A1 (en) * 2005-10-19 2007-06-21 Behrens Timothy W Systemic Lupus Erythematosus
US8249814B2 (en) 2005-10-21 2012-08-21 Genenews Inc. Method, computer readable medium, and system for determining a probability of colorectal cancer in a test subject
AU2005337803B2 (en) 2005-10-29 2013-04-18 Bayer Intellectual Property Gmbh Process for determining one or more analytes in samples of biological origin having complex composition, and use thereof
FR2893130B1 (en) * 2005-11-08 2008-05-02 Thales Sa IMAGING DEVICE FOR BIOPUCE, AND BIOPUCE THEREFOR
WO2008054398A2 (en) 2005-11-09 2008-05-08 The Trustees Of Columbia University In The City Of New York Photochemical methods and photoactive compounds for modifying surfaces
JP5186689B2 (en) * 2005-11-10 2013-04-17 国立大学法人広島大学 Protein immobilization method and immobilizing agent via protein binding to silicon oxide-containing substance
ES2356080T3 (en) * 2005-11-10 2011-04-04 Bristol-Myers Squibb Pharma Company MOESINA, CAVEOLINA AND PROTEINA 1 ASSOCIATED WITH YES AS MARKERS OF RESPONSE TO DASATINIB IN CANCERES DE MAMA.
US8372596B2 (en) * 2005-11-10 2013-02-12 National University Of Corporation Hiroshima University Asbestos detection method, asbestos detection agent, asbestos detection kit, method for screening candidate for agent aiming at preventing or treating disease for which asbestos is causative or worsening factor
US7889347B2 (en) * 2005-11-21 2011-02-15 Plexera Llc Surface plasmon resonance spectrometer with an actuator driven angle scanning mechanism
US7463358B2 (en) * 2005-12-06 2008-12-09 Lumera Corporation Highly stable surface plasmon resonance plates, microarrays, and methods
WO2007070809A2 (en) * 2005-12-12 2007-06-21 Mcgill University Biomarkers for babesia
US8841255B2 (en) 2005-12-20 2014-09-23 Duke University Therapeutic agents comprising fusions of vasoactive intestinal peptide and elastic peptides
CN101384272B (en) 2005-12-20 2013-05-01 杜克大学 Methods and compositions for delivering active agents with enhanced pharmacological properties
US20130172274A1 (en) 2005-12-20 2013-07-04 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
JP2007171003A (en) * 2005-12-22 2007-07-05 Fujifilm Corp Substrate for mass spectrometry, analytical method, and apparatus
US7781203B2 (en) * 2005-12-29 2010-08-24 Corning Incorporated Supports for assaying analytes and methods of making and using thereof
AU2006332457A1 (en) * 2005-12-30 2007-07-12 Bio-Layer Pty Limited Binding of molecules
JP2009522576A (en) * 2006-01-03 2009-06-11 プレジデント アンド フェローズ オブ ハーバード カレッジ Small molecule printing
US7709227B2 (en) * 2006-01-04 2010-05-04 Phasebio Pharmaceuticals, Inc. Multimeric ELP fusion constructs
US7648834B2 (en) * 2006-01-17 2010-01-19 Moore Wayne E Plasmon fluorescence augmentation for chemical and biological testing apparatus
US20090011428A1 (en) * 2006-01-18 2009-01-08 The Regents Of The University Of California Fluid Membrane-Based Ligand Display System for Live Cell Assays and Disease Diagnosis Applications
CA2640614A1 (en) * 2006-01-27 2007-08-09 Eastern Virginia Medical School Proteomic fingerprinting of human ivf-derived embryos: identification of biomarkers of developmental potential
JP4833679B2 (en) * 2006-01-31 2011-12-07 富士通株式会社 Method and apparatus for producing molecular film with adjusted density
US8008067B2 (en) 2006-02-13 2011-08-30 University Of Maryland, Baltimore County Microwave trigger metal-enhanced chemiluminescence (MT MEC) and spatial and temporal control of same
EP2375254A1 (en) 2006-02-17 2011-10-12 The Children's Medical Center Corporation Free NGAL as a biomarker for cancer
US20070207504A1 (en) * 2006-03-06 2007-09-06 The Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Proteomic methods for the identification of differentiated adipose cells and adipose derived adult stem cells
CA2645159A1 (en) * 2006-03-10 2008-06-26 Michael S. Urdea Multiplex protein fractionation
EP1996949A4 (en) 2006-03-11 2010-01-20 Univ Leland Stanford Junior Beta-2 microglobulin as a biomarker for peripheral artery disease
US7855057B2 (en) * 2006-03-23 2010-12-21 Millipore Corporation Protein splice variant/isoform discrimination and quantitative measurements thereof
US7909928B2 (en) 2006-03-24 2011-03-22 The Regents Of The University Of Michigan Reactive coatings for regioselective surface modification
EP2007514A2 (en) * 2006-03-28 2008-12-31 Inanovate, Inc. Nano-particle biochip substrates
US7886577B2 (en) 2006-03-30 2011-02-15 Oakland University Devices with surface bound ionic liquids and method of use thereof
US8375768B2 (en) * 2006-03-30 2013-02-19 Oakland University Ionic liquid thin layer sensor for electrochemical and/or piezoelectric measurements
WO2007115207A2 (en) * 2006-03-31 2007-10-11 Regents Of The University Of Minnesota Irf-5 haplotypes in systemic lupus erythematosus
US20080003694A1 (en) * 2006-04-18 2008-01-03 Swanson Basil I Robust, self-assembled, biocompatible films
US7923054B2 (en) 2006-04-19 2011-04-12 Gore Enterprise Holdings, Inc. Functional porous substrates for attaching biomolecules
US20100216657A1 (en) * 2006-05-16 2010-08-26 Arcxis Biotechnologies, Inc. Pcr-free sample preparation and detection systems for high speed biologic analysis and identification
WO2007140388A2 (en) * 2006-05-31 2007-12-06 The Johns Hopkins University Ablation based laser machining of biomolecule patterns on substrates
US7947148B2 (en) 2006-06-01 2011-05-24 The Regents Of The University Of Michigan Dry adhesion bonding
KR100931027B1 (en) * 2006-06-27 2009-12-10 한국생명공학연구원 Cysteine-tagged Protein shock Variant at the N-terminus
EP2041556A1 (en) * 2006-06-28 2009-04-01 RGB Technologies AB A sensor kit and a system for detecting an analyte in a test environment
FR2903590B1 (en) * 2006-07-13 2013-05-10 Commissariat Energie Atomique CELL SENSING DEVICE BY CONTACT
AU2007273055B2 (en) 2006-07-14 2014-05-01 The Regents Of The University Of California Cancer biomarkers and methods of use thereof
US8343539B2 (en) * 2006-07-14 2013-01-01 Regents Of The University Of Minnesota Compounds that bind α5β1 integrin and methods of use
JP2009545739A (en) * 2006-08-02 2009-12-24 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Method for determining analyte concentration using analyte sensor molecules bound to a porous membrane
GB0617429D0 (en) * 2006-09-05 2006-10-18 Electrophoretics Ltd Markers of renal transplant rejection and renal damage
JP5994121B2 (en) 2006-09-07 2016-09-21 オタゴ イノベーション リミテッド Biomarker
US8658573B2 (en) * 2006-09-11 2014-02-25 The Trustees Of Columbia University In The City Of New York Photo-generated carbohydrate arrays and the rapid identification of pathogen-specific antigens and antibodies
WO2008039774A1 (en) 2006-09-25 2008-04-03 Mayo Foundation For Medical Education And Research Extracellular and membrane-associated prostate cancer markers
US20090087925A1 (en) * 2007-10-01 2009-04-02 Zyomyx, Inc. Devices and methods for analysis of samples with depletion of analyte content
US20080085512A1 (en) * 2006-10-05 2008-04-10 D Andrade Petula N Array assay devices and methods for making and using the same
DK2084519T3 (en) * 2006-10-10 2012-08-20 Los Alamos Nat Security Llc X-ray fluorescence analysis method
EP2074230B1 (en) * 2006-10-11 2012-11-28 Janssen Pharmaceutica NV Compositions and methods for treating and diagnosing irritable bowel syndrome
US20080167532A1 (en) * 2006-10-13 2008-07-10 Mayo Foundation For Medical Education And Research Assessing cancer treatment responsiveness
JP2008105973A (en) * 2006-10-24 2008-05-08 Toyo Kohan Co Ltd Method of preserving polypeptide immobilized support
JP2010508520A (en) * 2006-10-31 2010-03-18 エス アール ユー バイオシステムズ,インコーポレイテッド Method for blocking non-specific protein binding on functionalized surfaces
JP2008171800A (en) * 2006-10-31 2008-07-24 Fei Co Protective layer for charged particle beam processing
WO2008058018A2 (en) 2006-11-02 2008-05-15 Mayo Foundation For Medical Education And Research Predicting cancer outcome
WO2008070570A2 (en) * 2006-12-01 2008-06-12 Cedars-Sinai Medical Center Positive selection of serum proteins for proteomic analysis
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
NZ619576A (en) * 2006-12-27 2014-07-25 Harvard College Compositions and methods for the treatment of infections and tumors
KR100894419B1 (en) * 2006-12-29 2009-04-24 삼성전자주식회사 Biochip kit and method of testing biological sample
WO2008092041A2 (en) 2007-01-24 2008-07-31 Carnegie Mellon University Optical biosensors
EP2108042A4 (en) * 2007-02-02 2010-04-14 California Inst Of Techn Surface chemistry and deposition techniques
US7867783B2 (en) 2007-02-22 2011-01-11 Maven Technologies, Llc Apparatus and method for performing ligand binding assays on microarrays in multiwell plates
TW200837349A (en) * 2007-03-07 2008-09-16 Nat Univ Tsing Hua Biochip and manufacturing method thereof
KR100836206B1 (en) 2007-03-20 2008-06-09 연세대학교 산학협력단 Peg hydrogel for fabrication of micropattern and process thereof
US8399047B2 (en) 2007-03-22 2013-03-19 The Regents Of The Univeristy Of Michigan Multifunctional CVD coatings
JP4850855B2 (en) * 2007-03-22 2012-01-11 信越化学工業株式会社 Manufacturing method of substrate for producing microarray
AU2008236810A1 (en) 2007-03-27 2008-10-16 Board Of Regents Of The University Of Texas System Biomarkers for ovarian cancer
JP5656339B2 (en) * 2007-03-28 2015-01-21 Jsr株式会社 Protein-immobilized carrier and method for producing the same
US20080242559A1 (en) * 2007-03-28 2008-10-02 Northwestern University Protein and peptide arrays
US7863037B1 (en) 2007-04-04 2011-01-04 Maven Technologies, Llc Ligand binding assays on microarrays in closed multiwell plates
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
US20100190166A1 (en) * 2007-04-19 2010-07-29 The Governors Of The University Of Alberta Tissue rejection biomarkers
US20080299673A1 (en) * 2007-04-19 2008-12-04 Sru Biosystems, Inc. Method for Employing a Biosensor to Detect Small Molecules ("Fragments") that Bind Directly to Immobilized Protein Targets
US20080274458A1 (en) * 2007-05-01 2008-11-06 Latham Gary J Nucleic acid quantitation methods
CA2690281A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
US20090041633A1 (en) * 2007-05-14 2009-02-12 Dultz Shane C Apparatus and method for performing ligand binding assays on microarrays in multiwell plates
JP5067588B2 (en) * 2007-05-18 2012-11-07 富士レビオ株式会社 Chemical surface nanopatterns for increased activity of surface-immobilized biological molecules
US7799558B1 (en) 2007-05-22 2010-09-21 Dultz Shane C Ligand binding assays on microarrays in closed multiwell plates
EP2160478B1 (en) * 2007-06-06 2014-08-27 Siemens Healthcare Diagnostics Inc. Predictive diagnostics for kidney disease
KR100927886B1 (en) * 2007-06-18 2009-11-23 한국생명공학연구원 Protein shock-oligonucleotide conjugates
CA3056116A1 (en) 2007-06-22 2008-12-31 Randolph Watnick Methods and uses thereof of prosaposin
WO2009003273A1 (en) * 2007-06-29 2009-01-08 The Governors Of The University Of Alberta Assessing tissue rejection
CA2693700A1 (en) * 2007-07-11 2009-01-15 Sru Biosystems, Inc. Methods for identifying modulators of ion channels
US9134307B2 (en) * 2007-07-11 2015-09-15 X-Body, Inc. Method for determining ion channel modulating properties of a test reagent
WO2009014987A2 (en) * 2007-07-20 2009-01-29 The University Of Utah Research Foundation Identification and quantification of biomarkers for evaluating the risk of preterm birth
DE102007034993A1 (en) * 2007-07-26 2009-01-29 Hanna Diehl Soluble Cadherin 17 for the diagnosis and risk stratification of colon or colon cancer
US20090060786A1 (en) * 2007-08-29 2009-03-05 Gibum Kim Microfluidic apparatus for wide area microarrays
US8354280B2 (en) 2007-09-06 2013-01-15 Bioscale, Inc. Reusable detection surfaces and methods of using same
DK2548962T3 (en) 2007-09-19 2016-04-11 Applied Biosystems Llc Sirna sequence-independent modification formats to reduce off-target phenotype effects in RNAI and stabilized forms thereof
JP5743135B2 (en) 2007-09-28 2015-07-01 エックスアールプロ・サイエンシーズ・インコーポレーテッド Method and apparatus for measuring post-translational modifications of proteins
WO2009058867A2 (en) * 2007-10-29 2009-05-07 Primorigen Biosciences, Llc Affinity measurements using frameless multiplexed microarrays
CN101932728B (en) 2007-11-30 2013-06-19 西门子医疗保健诊断公司 Adiponectin receptor fragments and methods of use
US8004669B1 (en) 2007-12-18 2011-08-23 Plexera Llc SPR apparatus with a high performance fluid delivery system
US20090181857A1 (en) * 2008-01-15 2009-07-16 Academia Sinica System and method for producing a label-free micro-array biochip
KR100959831B1 (en) 2008-01-18 2010-05-28 포항공과대학교 산학협력단 Pattern-Recognition Type Plate Detecting Multi-Biomolecule
US20090196852A1 (en) * 2008-02-04 2009-08-06 Watkinson D Tobin Compositions and methods for diagnosing and treating immune disorders
DE102008011850A1 (en) 2008-02-29 2009-09-03 Michael Grzendowski Biomarker for the diagnosis of brain tumor
CA2718251A1 (en) 2008-03-10 2009-09-17 Lineagen, Inc. Copd biomarker signatures
CA2715914C (en) 2008-03-12 2019-01-22 Otago Innovation Limited Insulin signal peptide fragment biomarkers
CA2715921A1 (en) 2008-03-12 2009-09-17 Otago Innovation Limited Biomarkers
WO2009118343A1 (en) * 2008-03-27 2009-10-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and kits for determining the occurrence of a liver disease in a subject
WO2009121034A2 (en) * 2008-03-28 2009-10-01 Pelican Group Holdings, Inc. Multicapillary sample preparation devices and methods for processing analytes
US8257936B2 (en) 2008-04-09 2012-09-04 X-Body Inc. High resolution label free analysis of cellular properties
WO2009137244A1 (en) * 2008-04-15 2009-11-12 Charles River Laboratories, Inc. Cartridge and method for sample analysis
US20090263905A1 (en) * 2008-04-18 2009-10-22 Kim Scheuringer Detection test assembly for detecting the presence of a substance in a sample
BRPI0910482A2 (en) 2008-04-29 2019-09-24 Abbott Lab double variable domain immunoglobins and their uses
US7981664B1 (en) 2008-05-22 2011-07-19 Maven Technologies, Llc Apparatus and method for performing ligand binding assays on microarrays in multiwell plates
US8039270B2 (en) * 2008-05-22 2011-10-18 Maven Technologies, Llc Apparatus and method for performing ligand binding assays on microarrays in multiwell plates
AU2009253675A1 (en) 2008-05-28 2009-12-03 Genomedx Biosciences, Inc. Systems and methods for expression-based discrimination of distinct clinical disease states in prostate cancer
US20090298082A1 (en) * 2008-05-30 2009-12-03 Klee George G Biomarker panels for predicting prostate cancer outcomes
US10407731B2 (en) 2008-05-30 2019-09-10 Mayo Foundation For Medical Education And Research Biomarker panels for predicting prostate cancer outcomes
EP2297209A4 (en) 2008-06-03 2012-08-01 Abbott Lab Dual variable domain immunoglobulins and uses thereof
WO2009149285A1 (en) * 2008-06-04 2009-12-10 Sru Biosystems, Inc. Detection of promiscuous small submicrometer aggregates
CN102057044B (en) * 2008-06-04 2013-05-01 泰勒克里斯生物治疗学公司 Composition, method and kit for preparing plasmin
JP2011526303A (en) 2008-06-27 2011-10-06 デューク ユニバーシティ A therapeutic agent containing an elastin-like peptide
WO2010123720A1 (en) 2009-04-23 2010-10-28 Siemens Healthcare Diagnostics Inc. Monomeric and dimeric forms of adiponectin receptor fragments and methods of use
US8021850B2 (en) * 2008-07-14 2011-09-20 Ribo Guo Universal tandem solid-phases based immunoassay
WO2010123608A2 (en) 2009-01-29 2010-10-28 The Regents Of The University Of California A spatial biomarker of disease and detection of spatial organization of cellular recptors
WO2010011860A1 (en) * 2008-07-23 2010-01-28 Diabetomics, Llc Methods for detecting pre-diabetes and diabetes
US10384203B2 (en) 2008-09-24 2019-08-20 First Light Biosciences, Inc. Kits and devices for detecting analytes
JP2012508577A (en) 2008-11-12 2012-04-12 カリス ライフ サイエンシズ ルクセンブルク ホールディングス Method and system for using exosomes to determine phenotype
US9598491B2 (en) * 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
JP4911639B2 (en) * 2008-12-02 2012-04-04 学校法人早稲田大学 Biosensing method and immobilization method
US8349325B2 (en) 2008-12-23 2013-01-08 Abbott Laboratories Soluble FMS-like tyrosine kinase-1 (sFLT-1) antibody and related composition, kit, methods of using, and materials and method for making
WO2010078403A2 (en) 2008-12-30 2010-07-08 Lipella Pharmaceuticals Inc. Methods and compositions for diagnosing urological disorders
AU2009333489A1 (en) 2008-12-30 2010-07-08 Centocor Ortho Biotech Inc. Serum markers predicting clinical response to anti-TNF antibodies in patients with ankylosing spondylitis
AU2010208394A1 (en) 2009-01-27 2011-09-08 Hologic, Inc. Biomarkers for detection of neonatal sepsis in biological fluid
WO2010087985A2 (en) 2009-01-28 2010-08-05 Yale University Novel markers for detection of complications resulting from in utero encounters
WO2010096331A1 (en) 2009-02-11 2010-08-26 Duke University Sensors incorporating antibodies and methods of making and using the same
ES2552337T3 (en) 2009-03-03 2015-11-27 Grifols Therapeutics Inc. Procedures for plasminogen preparation
JP5836807B2 (en) 2009-03-05 2015-12-24 アッヴィ・インコーポレイテッド IL-17 binding protein
WO2010106535A1 (en) 2009-03-15 2010-09-23 Technion Research And Development Foundation Ltd. Soluble hla complexes for use in disease diagnosis
WO2010125566A2 (en) 2009-04-27 2010-11-04 Technion Research And Development Foundation Ltd. Markers for cancer detection
US20100273185A1 (en) * 2009-04-27 2010-10-28 Sru Biosystems, Inc. Detection of Biased Agonist Activation
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)
US20100291575A1 (en) * 2009-05-15 2010-11-18 Sru Biosystems, Inc. Detection of Changes in Cell Populations and Mixed Cell Populations
CN102449481B (en) * 2009-05-29 2016-05-25 德克萨斯大学系统董事会 For separating of with the class peptide part of processing autoimmune T cell
EP2438444A1 (en) * 2009-06-02 2012-04-11 The Board of Regents of The University of Texas System Identification of small molecules recognized by antibodies in subjects with neurodegenerative diseases
WO2010148365A2 (en) * 2009-06-19 2010-12-23 The Arizona Board Of Regents, A Body Corporate Of The State Of Arizona For And On Behalf Of Arizona State University Compound arrays for sample profiling
GB0912231D0 (en) * 2009-07-14 2009-08-26 Imp Innovations Ltd Method and apparatus for determining an analyte parameter
US8685755B2 (en) * 2009-07-20 2014-04-01 The Board Of Regents Of The University Of Texas System Combinatorial multidomain mesoporous chips and a method for fractionation, stabilization, and storage of biomolecules
HUE032703T2 (en) 2009-08-14 2017-10-30 Phasebio Pharmaceuticals Inc Modified vasoactive intestinal peptides
EP2504024A2 (en) 2009-09-27 2012-10-03 Ruhr-Universität Bochum Method for the therapy and diagnosis of alzheimer's disease
US8759259B2 (en) * 2009-10-16 2014-06-24 The Board Of Regents Of The University Of Texas System Compositions and methods for producing cyclic peptoid libraries
KR101105328B1 (en) * 2009-11-23 2012-01-16 한국표준과학연구원 Apparatus and method for quantifying the binding and dissociation kinetics of molecular interactions
ES2677944T3 (en) 2009-11-25 2018-08-07 Hologic Inc. Detection of intraamniotic infection
EP2513137B1 (en) 2009-12-17 2018-02-28 Children's Medical Center Corporation Saposin-a derived peptides and uses thereof
US8355133B2 (en) * 2009-12-30 2013-01-15 Maven Technologies, Llc Biological testing with sawtooth-shaped prisms
WO2011088226A2 (en) 2010-01-13 2011-07-21 Caris Life Sciences Luxembourg Holdings Detection of gastrointestinal disorders
WO2011089903A1 (en) * 2010-01-25 2011-07-28 Panasonic Corporation A method for immobilizing protein a on a self-assembled monolayer
JP6063378B2 (en) 2010-01-26 2017-01-18 バンヤン・バイオマーカーズ・インコーポレイテッド Compositions and methods related to argininosuccinate synthase
CA2791905A1 (en) 2010-03-01 2011-09-09 Caris Life Sciences Luxembourg Holdings, S.A.R.L. Biomarkers for theranostics
JP5828195B2 (en) 2010-03-11 2015-12-02 ユニバーシティ オブ ルイスビル リサーチ ファンデーション,インコーポレイテッドUniversity Of Louisville Research Foundation,Inc. Methods for predicting pregnancy loss risk and methods for reducing pregnancy loss risk
US20110229921A1 (en) * 2010-03-18 2011-09-22 Abbott Laboratories METHODS OF ASSAYING URINARY NEUTROPHIL GELATINASE-ASSOCIATED LIPOCALIN (uNGAL) IN THE PROGNOSIS OF CADAVERIC KIDNEY TRANSPLANT FUNCTION IN A PATIENT, INCLUDING A PATIENT DIAGNOSED WITH DELAYED GRAFT FUNCTION (DGF), A METHOD OF ASSAYING uNGAL IN THE ASSESSMENT OF RISK OF DGF IN A PATIENT DIAGNOSED WITH EARLY GRAFT FUNCTION (EGF), AND RELATED KITS
US20110237535A1 (en) * 2010-03-26 2011-09-29 Sru Biosystems, Inc. Use of Induced Pluripotent Cells and other Cells for Screening Compound Libraries
EP2556172A4 (en) 2010-04-06 2013-10-30 Caris Life Sciences Luxembourg Holdings Circulating biomarkers for disease
EP3508854A1 (en) 2010-04-27 2019-07-10 The Regents of The University of California Cancer biomarkers and methods of use thereof
TWI615405B (en) 2010-05-14 2018-02-21 艾伯維有限公司 Il-1 binding proteins
US9211542B2 (en) * 2010-05-21 2015-12-15 Eidgenossische Technische Hochschule Zurich High-density sample support plate for automated sample aliquoting
US9551714B2 (en) 2010-06-25 2017-01-24 Abbott Laboratories Materials and methods for assay of anti-hepatitis C virus (HCV) antibodies
US20120009196A1 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
ES2629850T3 (en) 2010-07-19 2017-08-16 Otago Innovation Limited Signal biomarkers
KR101045209B1 (en) * 2010-07-26 2011-06-30 삼성전자주식회사 An array device comprising a photoresist film having immobilization regions and a method using the same
CA2807014A1 (en) 2010-08-03 2012-02-09 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
CN102918064B (en) * 2010-08-30 2015-03-11 松下健康医疗控股株式会社 A method for immobilizing streptavidin on a self-assembled monolayer
CN103459611B (en) 2010-09-17 2016-11-02 哈佛大学校长及研究员协会 The functional genomics research that effectiveness and the safety of pluripotent stem cell are characterized
WO2012051519A2 (en) 2010-10-14 2012-04-19 The Johns Hopkins University Biomarkers of brain injury
WO2012053138A1 (en) * 2010-10-19 2012-04-26 パナソニック株式会社 Method for immobilizing glucose oxidase on self-assembled film
US20120115244A1 (en) 2010-11-09 2012-05-10 Abbott Laboratories Materials and methods for immunoassay of pterins
CN102486474B (en) * 2010-12-06 2014-05-28 北京大学人民医院 Protein chip for chronic hepatitis C outcome prediction in chronic hepatitis C interferon treatment
TW201249865A (en) 2010-12-21 2012-12-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
BR112013023722A2 (en) 2011-03-15 2016-11-22 Univ Utah Res Found methods for predicting the risk of an individual having or developing a disease, and for diagnosing an individual with a disease, kit, nonhuman animal model, and system for identifying aberrant coriocapillary lobes
US8735175B2 (en) * 2011-03-18 2014-05-27 Chris D. Geddes Multicolor microwave-accelerated metal-enhanced fluorescence (M-MAMEF)
WO2012139089A2 (en) 2011-04-08 2012-10-11 The General Hospital Corporation Fungal-derived carbohydrate-conjugated scaffold
CA2833257A1 (en) 2011-04-15 2012-10-18 Children's Medical Center Corporation Diagnostic markers and therapeutic targets of kawasaki disease
KR101167649B1 (en) 2011-04-20 2012-07-20 한국과학기술원 Apparatus for analyzing single protein-protein interactions in whole cell lysates
EP2707389B1 (en) 2011-05-12 2019-10-30 The Johns Hopkins University Assay reagents for a neurogranin diagnostic kit
EP2717902B1 (en) 2011-06-06 2018-01-24 Phasebio Pharmaceuticals, Inc. Use of modified vasoactive intestinal peptides in the treatment of hypertension
CN103492879B (en) * 2011-06-10 2015-04-01 松下健康医疗控股株式会社 Method for affixing antibodies to self-assembled monolayer
WO2012174014A2 (en) * 2011-06-13 2012-12-20 Indevr, Inc. Low density microarrays for vaccine related protein quantification, potency determination and efficacy evaluation
CN102279261B (en) * 2011-06-20 2013-09-18 东南大学 Inkjet printing preparation method of pattern code microcarrier
EP2724156B1 (en) 2011-06-27 2017-08-16 The Jackson Laboratory Methods and compositions for treatment of cancer and autoimmune disease
WO2013005269A1 (en) * 2011-07-05 2013-01-10 パナソニック株式会社 Method for immobilizing albumin on self-assembled monolayer
WO2013008280A1 (en) * 2011-07-08 2013-01-17 パナソニック株式会社 Method for immobilizing protein on self-assembled film
US20140235492A1 (en) * 2011-09-20 2014-08-21 Institut National De La Sante Et De La Recherche Medicate (Inserm) Methods for preparing single domain antibody microarrays
WO2013055829A1 (en) * 2011-10-11 2013-04-18 Nestec S.A. Proximity-based assays for the detection of signaling protein expression and activation
RU2014121043A (en) 2011-10-24 2015-12-10 Эббви Инк. BISPECIFIC IMMUNO-BINDING AGENTS AIMED AGAINST TNF AND IL-17
US8999331B2 (en) 2011-10-24 2015-04-07 Abbvie Inc. Immunobinders directed against sclerostin
ES2785973T3 (en) 2011-11-07 2020-10-08 Rapid Micro Biosystems Inc Sterility test cassette
BR112014011491A2 (en) 2011-11-14 2017-05-09 Nestec Sa trials and methods for selecting a treatment regimen for an individual with depression
AU2012352153B2 (en) 2011-12-13 2018-07-26 Veracyte, Inc. Cancer diagnostics using non-coding transcripts
AU2012358269B2 (en) 2011-12-22 2017-11-02 Children's Medical Center Corporation Saposin-A derived peptides and uses thereof
US8993248B2 (en) 2011-12-31 2015-03-31 Abbott Laboratories Truncated human vitamin D binding protein and mutation and fusion thereof and related materials and methods of use
US20150010914A1 (en) 2012-01-20 2015-01-08 Adelaide Research & Innovation Pty Ltd. Biomarkers for gastric cancer and uses thereof
CA2864080C (en) * 2012-02-07 2023-04-25 Vibrant Holdings, Llc Substrates, peptide arrays, and methods
EP2828282B1 (en) 2012-03-20 2017-12-27 Otago Innovation Limited Biomarkers
US10407707B2 (en) 2012-04-16 2019-09-10 Rapid Micro Biosystems, Inc. Cell culturing device
EP2866928A1 (en) 2012-06-29 2015-05-06 Danmarks Tekniske Universitet A method of charging a test carrier and a test carrier
UY34905A (en) 2012-07-12 2014-01-31 Abbvie Inc IL-1 UNION PROTEINS
WO2014028907A1 (en) 2012-08-16 2014-02-20 The Trustees Of Columbia University In The City Of New York Diagnostic markers of indolent prostate cancer
WO2014028884A2 (en) 2012-08-16 2014-02-20 Genomedx Biosciences, Inc. Cancer diagnostics using biomarkers
US10006909B2 (en) 2012-09-28 2018-06-26 Vibrant Holdings, Llc Methods, systems, and arrays for biomolecular analysis
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
EP2912182B1 (en) 2012-10-23 2021-12-08 Caris Science, Inc. Aptamers and uses thereof
WO2014100434A1 (en) 2012-12-19 2014-06-26 Caris Science, Inc. Compositions and methods for aptamer screening
AU2013202668B2 (en) 2012-12-24 2014-12-18 Adelaide Research & Innovation Pty Ltd Inhibition of cancer growth and metastasis
US9086412B2 (en) 2012-12-31 2015-07-21 University Of Louisville Research Foundation, Inc. Extracellular vesicle-associated protein markers of cancer
US9790478B2 (en) 2013-03-14 2017-10-17 Abbott Laboratories HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
CA2906421C (en) 2013-03-14 2022-08-16 George J. Dawson Hcv antigen-antibody combination assay and methods and compositions for use therein
MX2015012825A (en) 2013-03-14 2016-06-10 Abbott Lab Hcv core lipid binding domain monoclonal antibodies.
US9157910B2 (en) 2013-03-15 2015-10-13 Abbott Laboratories Assay with increased dynamic range
US9005901B2 (en) 2013-03-15 2015-04-14 Abbott Laboratories Assay with internal calibration
US20160109455A1 (en) 2013-05-10 2016-04-21 Vermillion, Inc. Compositions for ovarian cancer assessment having improved specificty
DE102013210138A1 (en) 2013-05-30 2014-12-04 Boehringer Ingelheim Vetmedica Gmbh Method for generating a plurality of measuring ranges on a chip and chip with measuring ranges
JP2014235115A (en) 2013-06-04 2014-12-15 ウシオ電機株式会社 Microchip and method for forming metal thin film in microchip
WO2015009907A1 (en) 2013-07-17 2015-01-22 The Johns Hopkins University A multi-protein biomarker assay for brain injury detection and outcome
WO2015031694A2 (en) 2013-08-28 2015-03-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
CA2940653A1 (en) 2014-02-27 2015-09-03 Vijay Kuchroo T cell balance gene expression, compositions of matters and methods of use thereof
WO2015187227A2 (en) 2014-03-13 2015-12-10 Duke University Electronic platform for sensing and control of electrochemical reactions
ES2818824T3 (en) 2014-05-08 2021-04-14 Phasebio Pharmaceuticals Inc Compositions comprising a VIP-ELP fusion protein for use in the treatment of cystic fibrosis
EP3152347A4 (en) 2014-06-04 2018-04-11 Indevr, Inc. Universal capture array for multiplexed subtype-specific quantification and stability determination of influenza proteins
SG11201610610YA (en) 2014-06-19 2017-01-27 Sloan Kettering Inst Cancer Biomarkers for response to ezh2 inhibitors
JP2017521654A (en) 2014-06-27 2017-08-03 アボット・ラボラトリーズAbbott Laboratories Compositions and methods for detecting human pegivirus 2 (HPgV-2)
US20170227541A1 (en) 2014-07-17 2017-08-10 The Trustees Of The University Of Pennsylvania Methods for using exosomes to monitor transplanted organ status
US20160032281A1 (en) * 2014-07-31 2016-02-04 Fei Company Functionalized grids for locating and imaging biological specimens and methods of using the same
US10222386B2 (en) 2014-09-19 2019-03-05 The Johns Hopkins University Biomarkers of congnitive dysfunction
ES2828713T3 (en) 2014-10-29 2021-05-27 Abbott Lab Target Anti-HCV Antibody Detection Assays Using NS3 Capture Peptides
KR101687950B1 (en) * 2014-11-14 2016-12-21 연세대학교 산학협력단 Surface modified micropate, menufacturing method for the surface modified micropate and immunoassays using the surface modified micropate
US11168369B2 (en) 2014-11-25 2021-11-09 The Brigham And Women's Hospital, Inc. Method of identifying and treating a person having a predisposition to or afflicted with a cardiometabolic disease
US10683552B2 (en) 2014-11-25 2020-06-16 Presidents And Fellows Of Harvard College Clonal haematopoiesis
US11543411B2 (en) 2014-12-05 2023-01-03 Prelude Corporation DCIS recurrence and invasive breast cancer
WO2016109378A1 (en) 2014-12-29 2016-07-07 North Carolina State University Multiplexed diagnostic to recognize concentrations of related proteins and peptides
AU2016219513B2 (en) 2015-02-09 2021-09-30 Immunoforge Co., Ltd. Methods and compositions for treating muscle disease and disorders
EP3259594A4 (en) 2015-02-20 2018-12-26 The Johns Hopkins University Biomarkers of myocardial injury
EP3262193A2 (en) 2015-02-26 2018-01-03 The Broad Institute Inc. T cell balance gene expression, compositions of matters and methods of use thereof
CN107533069A (en) * 2015-03-06 2018-01-02 泰摩拉分析运营有限责任公司 Array for the chemically functionalization of analyzing proteins modification
AU2016229076B2 (en) 2015-03-09 2022-01-20 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10294451B2 (en) 2015-04-22 2019-05-21 University Of Maryland, Baltimore County Flow and static lysing systems and methods for ultra-rapid isolation and fragmentation of biological materials by microwave irradiation
EP3314027A4 (en) 2015-06-29 2019-07-03 Caris Science, Inc. Therapeutic oligonucleotides
GB2556004A (en) 2015-07-10 2018-05-16 Univ West Virginia Markers of stroke and stroke severity
WO2017015531A1 (en) 2015-07-22 2017-01-26 University Of Maryland, Baltimore County Hydrophilic coatings of plasmonic metals to enable low volume metal-enhanced fluorescence
CA2993652A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
DE102015114026A1 (en) 2015-08-24 2017-03-02 Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V. Biomarker panel for the diagnosis of recurrent prostate cancer
US10758886B2 (en) 2015-09-14 2020-09-01 Arizona Board Of Regents On Behalf Of Arizona State University Conditioned surfaces for in situ molecular array synthesis
US11884978B2 (en) 2015-09-30 2024-01-30 Immunexpress Pty Ltd Pathogen biomarkers and uses therefor
CA3003767A1 (en) 2015-12-02 2017-06-08 Boehringer Ingelheim Vetmedica Gmbh Method for producing a plurality of measurement regions on a chip, and chip having a plurality of measurement regions
US20190000831A1 (en) 2015-12-11 2019-01-03 The General Hospital Corporation Compositions and methods for treating drug-tolerant glioblastoma
EP3394291B1 (en) 2015-12-24 2021-10-06 Immunexpress Pty Ltd Triage biomarkers and uses therefor
IL303936A (en) 2016-03-18 2023-08-01 Caris Science Inc Oligonucleotide probes and uses thereof
IL298701B2 (en) 2016-05-25 2024-03-01 Caris Science Inc Oligonucleotide probes and uses thereof
US11747334B2 (en) 2016-06-20 2023-09-05 Cowper Sciences Inc. Methods for differential diagnosis of autoimmune diseases
AU2017281040A1 (en) 2016-06-20 2019-01-24 Healthtell Inc. Methods for diagnosis and treatment of autoimmune diseases
JP6420281B2 (en) * 2016-07-04 2018-11-07 花王株式会社 Solid phase carrier for protein analysis and method for producing the same
US11414708B2 (en) 2016-08-24 2022-08-16 Decipher Biosciences, Inc. Use of genomic signatures to predict responsiveness of patients with prostate cancer to post-operative radiation therapy
CA3043264A1 (en) 2016-11-11 2018-05-17 Healthtell Inc. Methods for identifying candidate biomarkers
US11448648B2 (en) 2016-11-11 2022-09-20 Ascendant Diagnostics, LLC Compositions and methods for diagnosing and differentiating systemic juvenile idiopathic arthritis and Kawasaki disease
US11208697B2 (en) 2017-01-20 2021-12-28 Decipher Biosciences, Inc. Molecular subtyping, prognosis, and treatment of bladder cancer
AU2018230784A1 (en) 2017-03-09 2019-10-10 Decipher Biosciences, Inc. Subtyping prostate cancer to predict response to hormone therapy
EP3615694B1 (en) 2017-04-25 2022-03-30 The Brigham and Women's Hospital, Inc. Il-8, il-6, il-1b and tet2 and dnmt3a in atherosclerosis
EP3622087A4 (en) 2017-05-12 2021-06-16 Decipher Biosciences, Inc. Genetic signatures to predict prostate cancer metastasis and identify tumor agressiveness
WO2018218250A2 (en) 2017-05-26 2018-11-29 Vibrant Holdings, Llc Photoactive compounds and methods for biomolecule detection and sequencing
MA51647A (en) 2017-08-16 2020-06-24 Medimmune Llc COMPOSITIONS AND METHODS FOR ATOPIC DERMATITIS TREATMENT AND TREATMENT SELECTION
US11709155B2 (en) 2017-09-18 2023-07-25 Waters Technologies Corporation Use of vapor deposition coated flow paths for improved chromatography of metal interacting analytes
US11709156B2 (en) 2017-09-18 2023-07-25 Waters Technologies Corporation Use of vapor deposition coated flow paths for improved analytical analysis
WO2019094798A1 (en) 2017-11-10 2019-05-16 The Trustees Of Columbia University In The City Of New York Methods and compositions for promoting or inducing hair growth
EP3791188A1 (en) 2018-05-10 2021-03-17 The Methodist Hospital Methods for prognosis and management of disease
CN112272775A (en) 2018-06-04 2021-01-26 雅芳产品公司 Protein biomarkers for identifying and treating aging skin and skin conditions
AU2019339508A1 (en) 2018-09-14 2021-04-15 Prelude Corporation Method of selection for treatment of subjects at risk of invasive breast cancer
EP3877400A4 (en) 2018-11-07 2022-09-07 Seer, Inc. Compositions, methods and systems for protein corona analysis and uses thereof
GB201904697D0 (en) 2019-04-03 2019-05-15 Vib Vzw Means and methods for single molecule peptide sequencing
CN117129704A (en) 2019-08-05 2023-11-28 禧尔公司 Systems and methods for sample preparation, data generation, and protein crown analysis
US11918936B2 (en) 2020-01-17 2024-03-05 Waters Technologies Corporation Performance and dynamic range for oligonucleotide bioanalysis through reduction of non specific binding
EP4074820A1 (en) 2021-04-16 2022-10-19 The Trustees of The University of Pennsylvania Micro-engineered models of the human eye and methods of use
WO2023288108A1 (en) * 2021-07-16 2023-01-19 The University Of Chicago Biocompatible surface for quantum sensing and methods thereof
WO2023122723A1 (en) 2021-12-23 2023-06-29 The Broad Institute, Inc. Panels and methods for diagnosing and treating lung cancer

Citations (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4071409A (en) * 1976-05-20 1978-01-31 Corning Glass Works Immobilization of proteins on inorganic support materials
US4444879A (en) * 1981-01-29 1984-04-24 Science Research Center, Inc. Immunoassay with article having support film and immunological counterpart of analyte
US4514508A (en) * 1982-07-06 1985-04-30 Biond Inc. Assaying for a multiplicity of antigens or antibodies with a detection compound
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US4722896A (en) * 1981-01-26 1988-02-02 The Beth Israel Hospital Association Method for affinity purification of hybridoma antibodies
US4802951A (en) * 1986-03-07 1989-02-07 Trustees Of Boston University Method for parallel fabrication of nanometer scale multi-device structures
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4894146A (en) * 1986-01-27 1990-01-16 University Of Utah Thin channel split flow process and apparatus for particle fractionation
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US4987032A (en) * 1987-06-26 1991-01-22 Fuji Photo Film Co., Ltd. Functional organic thin film and method of manufacture thereof
US4994373A (en) * 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US5079600A (en) * 1987-03-06 1992-01-07 Schnur Joel M High resolution patterning on solid substrates
US5096807A (en) * 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
US5281540A (en) * 1988-08-02 1994-01-25 Abbott Laboratories Test array for performing assays
US5283173A (en) * 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5294369A (en) * 1990-12-05 1994-03-15 Akzo N.V. Ligand gold bonding
US5296114A (en) * 1991-12-06 1994-03-22 Ciba-Geigy Corporation Electrophoretic separating device and electrophoretic separating method
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5384261A (en) * 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US5405766A (en) * 1992-03-26 1995-04-11 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of National Defence Immobilization of biologically active protein on a support with a 7-18 carbon spacer and a bifunctional phospholipid
US5405783A (en) * 1989-06-07 1995-04-11 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of an array of polymers
US5412087A (en) * 1992-04-24 1995-05-02 Affymax Technologies N.V. Spatially-addressable immobilization of oligonucleotides and other biological polymers on surfaces
US5424186A (en) * 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5466589A (en) * 1990-11-19 1995-11-14 Biotechnology Research & Development Corporation Coated substrates employing oriented layers of mutant heme proteins and methods of making and using same
US5489678A (en) * 1989-06-07 1996-02-06 Affymax Technologies N.V. Photolabile nucleoside and peptide protecting groups
US5498545A (en) * 1994-07-21 1996-03-12 Vestal; Marvin L. Mass spectrometer system and method for matrix-assisted laser desorption measurements
US5506121A (en) * 1992-11-03 1996-04-09 Institut Fur Bioanalytik Gemeinnutzige Gesellschaft MBH Fusion peptides with binding activity for streptavidin
US5512492A (en) * 1993-05-18 1996-04-30 University Of Utah Research Foundation Waveguide immunosensor with coating chemistry providing enhanced sensitivity
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5514501A (en) * 1994-06-07 1996-05-07 The United States Of America As Represented By The Secretary Of Commerce Process for UV-photopatterning of thiolate monolayers self-assembled on gold, silver and other substrates
US5516635A (en) * 1991-10-15 1996-05-14 Ekins; Roger P. Binding assay employing labelled reagent
US5520787A (en) * 1994-02-09 1996-05-28 Abbott Laboratories Diagnostic flow cell device
US5532142A (en) * 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
US5594111A (en) * 1994-01-28 1997-01-14 Prolinx, Inc. Phenylboronic acid complexes for bioconjugate preparation
US5593838A (en) * 1994-11-10 1997-01-14 David Sarnoff Research Center, Inc. Partitioned microelectronic device array
US5603351A (en) * 1995-06-07 1997-02-18 David Sarnoff Research Center, Inc. Method and system for inhibiting cross-contamination in fluids of combinatorial chemistry device
US5605662A (en) * 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US5620850A (en) * 1994-09-26 1997-04-15 President And Fellows Of Harvard College Molecular recognition at surfaces derivatized with self-assembled monolayers
US5623055A (en) * 1994-01-28 1997-04-22 Prolinx, Inc. Phenylboronic acid complexes derived from aminosalicylic acid for bioconjugate preparation
US5622826A (en) * 1994-12-22 1997-04-22 Houston Advanced Research Center Method for immobilization of molecules on platinum solid support surfaces
US5624711A (en) * 1995-04-27 1997-04-29 Affymax Technologies, N.V. Derivatization of solid supports and methods for oligomer synthesis
US5627369A (en) * 1995-05-19 1997-05-06 Perseptive Biosystems, Inc. Time-of-flight mass spectrometry analysis of biomolecules
US5629213A (en) * 1995-03-03 1997-05-13 Kornguth; Steven E. Analytical biosensor
US5637469A (en) * 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5643948A (en) * 1986-06-11 1997-07-01 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. K.
US5719060A (en) * 1993-05-28 1998-02-17 Baylor College Of Medicine Method and apparatus for desorption and ionization of analytes
US5720928A (en) * 1988-09-15 1998-02-24 New York University Image processing and analysis of individual nucleic acid molecules
US5726026A (en) * 1992-05-01 1998-03-10 Trustees Of The University Of Pennsylvania Mesoscale sample preparation device and systems for determination and processing of analytes
US5731152A (en) * 1996-05-13 1998-03-24 Motorola, Inc. Methods and systems for biological reagent placement
US5741700A (en) * 1993-08-11 1998-04-21 University Of Chicago Method of immobilizing water-soluble bioorganic compounds on a capillary-porous carrier
US5763170A (en) * 1991-04-16 1998-06-09 Amersham International Plc Method for forming an array of biological particles
US5763263A (en) * 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
US5858188A (en) * 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
US5861242A (en) * 1993-06-25 1999-01-19 Affymetrix, Inc. Array of nucleic acid probes on biological chips for diagnosis of HIV and methods of using the same
US5861254A (en) * 1997-01-31 1999-01-19 Nexstar Pharmaceuticals, Inc. Flow cell SELEX
US5866362A (en) * 1985-03-28 1999-02-02 Chiron Corporation Enhanced purification and expression of insoluble recombinant proteins
US5866363A (en) * 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
US5874219A (en) * 1995-06-07 1999-02-23 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US5885793A (en) * 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5905024A (en) * 1996-12-17 1999-05-18 University Of Chicago Method for performing site-specific affinity fractionation for use in DNA sequencing
US5948621A (en) * 1997-09-30 1999-09-07 The United States Of America As Represented By The Secretary Of The Navy Direct molecular patterning using a micro-stamp gel
US6040193A (en) * 1991-11-22 2000-03-21 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US6051380A (en) * 1993-11-01 2000-04-18 Nanogen, Inc. Methods and procedures for molecular biological analysis and diagnostics
US6061476A (en) * 1997-11-24 2000-05-09 Cognex Corporation Method and apparatus using image subtraction and dynamic thresholding
US6064754A (en) * 1996-11-29 2000-05-16 Oxford Glycosciences (Uk) Ltd. Computer-assisted methods and apparatus for identification and characterization of biomolecules in a biological sample
US6066448A (en) * 1995-03-10 2000-05-23 Meso Sclae Technologies, Llc. Multi-array, multi-specific electrochemiluminescence testing
US6190908B1 (en) * 1998-08-12 2001-02-20 The Scripps Research Institute Modulation of polypeptide display on modified filamentous phage
US6190619B1 (en) * 1997-06-11 2001-02-20 Argonaut Technologies, Inc. Systems and methods for parallel synthesis of compounds
US6194612B1 (en) * 1995-10-17 2001-02-27 The Scripps Research Institute Template for solution phase synthesis of combination libraries
US6197506B1 (en) * 1989-06-07 2001-03-06 Affymetrix, Inc. Method of detecting nucleic acids
US6197599B1 (en) * 1998-07-30 2001-03-06 Guorong Chin Method to detect proteins
US6225047B1 (en) * 1997-06-20 2001-05-01 Ciphergen Biosystems, Inc. Use of retentate chromatography to generate difference maps
US6228326B1 (en) * 1996-11-29 2001-05-08 The Board Of Trustees Of The Leland Stanford Junior University Arrays of independently-addressable supported fluid bilayer membranes
US6232066B1 (en) * 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6346413B1 (en) * 1989-06-07 2002-02-12 Affymetrix, Inc. Polymer arrays
US6350369B1 (en) * 1998-04-14 2002-02-26 California Institute Of Technology Method and system for determining analyte activity
US6365418B1 (en) * 1998-07-14 2002-04-02 Zyomyx, Incorporated Arrays of protein-capture agents and methods of use thereof
US6391625B1 (en) * 1999-06-28 2002-05-21 Lg Electronics Inc. Biochip and method for patterning and measuring biomaterial of the same
US20030017149A1 (en) * 1996-10-10 2003-01-23 Hoeffler James P. Single chain monoclonal antibody fusion reagents that regulate transcription in vivo
US6531283B1 (en) * 2000-06-20 2003-03-11 Molecular Staging, Inc. Protein expression profiling
US6544739B1 (en) * 1990-12-06 2003-04-08 Affymetrix, Inc. Method for marking samples
US20030073811A1 (en) * 2000-08-17 2003-04-17 Sense Proteomic Limited Method
US6682942B1 (en) * 1998-07-14 2004-01-27 Zyomyx, Inc. Microdevices for screening biomolecules
US6692751B1 (en) * 1988-05-06 2004-02-17 New York Blood Center Methods and systems for producing recombinant viral antigens
US6699665B1 (en) * 2000-11-08 2004-03-02 Surface Logix, Inc. Multiple array system for integrating bioarrays
US6720149B1 (en) * 1995-06-07 2004-04-13 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US6720157B2 (en) * 2000-02-23 2004-04-13 Zyomyx, Inc. Chips having elevated sample surfaces
US20050026215A1 (en) * 2003-07-17 2005-02-03 Predki Paul F. Method for the prediction of an epitope
US20050095646A1 (en) * 2001-11-19 2005-05-05 Sherman Michael I. Method of using a non-antibody protein to detect and measure an analyte
US20050100947A1 (en) * 1998-07-14 2005-05-12 Zyomyx, Inc. Array devices and methods of use thereof
US6897073B2 (en) * 1998-07-14 2005-05-24 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US6899137B2 (en) * 1999-06-28 2005-05-31 California Institute Of Technology Microfabricated elastomeric valve and pump systems

Family Cites Families (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690715A (en) 1982-06-18 1987-09-01 American Telephone And Telegraph Company, At&T Bell Laboratories Modification of the properties of metals
US4973493A (en) 1982-09-29 1990-11-27 Bio-Metric Systems, Inc. Method of improving the biocompatibility of solid surfaces
GB8314523D0 (en) 1983-05-25 1983-06-29 Lowe C R Diagnostic device
US4608112A (en) * 1984-05-16 1986-08-26 The United States Of America As Represented By The Secretary Of The Air Force Mask aligner for solar cell fabrication
AU600885B2 (en) 1984-05-25 1990-08-30 Zymogenetics Inc. Stable DNA constructs
US5637489A (en) * 1986-08-23 1997-06-10 Hoechst Aktiengesellschaft Phosphinothricin-resistance gene, and its use
US4859538A (en) 1986-11-20 1989-08-22 Ribi Hans O Novel lipid-protein compositions and articles and methods for their preparation
US4928112A (en) * 1987-03-23 1990-05-22 Howtek, Inc. Ink curing apparatus
US5154808A (en) 1987-06-26 1992-10-13 Fuji Photo Film Co., Ltd. Functional organic thin film and process for producing the same
DE3733190A1 (en) * 1987-10-01 1989-04-13 Kugelfischer G Schaefer & Co MULTI-ROW BALL OR ROLLER BEARING OR COMBINED BALL ROLLER BEARING
SE462454B (en) 1988-11-10 1990-06-25 Pharmacia Ab METHOD FOR USE IN BIOSENSORS
JPH02272081A (en) * 1989-04-14 1990-11-06 Fuji Photo Film Co Ltd Functional organic thin film
IT1229691B (en) 1989-04-21 1991-09-06 Eniricerche Spa SENSOR WITH ANTIGEN CHEMICALLY LINKED TO A SEMICONDUCTIVE DEVICE.
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
DE3939973A1 (en) 1989-12-02 1991-06-06 Basf Ag TWO-DIMENSIONAL CRYSTALLIZED MACROMOLECULAR LAYERS
WO1991016425A1 (en) 1990-04-12 1991-10-31 Hans Sigrist Method for the light-induced immobilization of biomolecules on chemically 'inert' surfaces
US5665582A (en) * 1990-10-29 1997-09-09 Dekalb Genetics Corp. Isolation of biological materials
US5384886A (en) * 1991-04-01 1995-01-24 Xerox Corporation Process for electronically printing envelopes
IL103674A0 (en) * 1991-11-19 1993-04-04 Houston Advanced Res Center Method and apparatus for molecule detection
US5756355A (en) 1992-04-22 1998-05-26 Ecole Polytechnique Federale De Lausanne Lipid membrane sensors
JPH0641183A (en) 1992-07-23 1994-02-15 Mitsubishi Kasei Corp Monomolecular film of oligonucleotide
CA2108705A1 (en) * 1992-11-06 1994-05-07 Richard Barner Biologically recognizing layers on new ti02 waveguide for biosensors
US5472881A (en) 1992-11-12 1995-12-05 University Of Utah Research Foundation Thiol labeling of DNA for attachment to gold surfaces
US5677196A (en) 1993-05-18 1997-10-14 University Of Utah Research Foundation Apparatus and methods for multi-analyte homogeneous fluoro-immunoassays
US5837832A (en) 1993-06-25 1998-11-17 Affymetrix, Inc. Arrays of nucleic acid probes on biological chips
US5441876A (en) 1993-07-30 1995-08-15 The United States Of America As Represented By The Secretary Of The Navy Process for the preparation of headgroup-modified phospholipids using phosphatidylhydroxyalkanols as intermediates
US5874239A (en) * 1993-07-30 1999-02-23 Affymax Technologies N.V. Biotinylation of proteins
JPH0784372A (en) 1993-09-17 1995-03-31 Res Dev Corp Of Japan Organic silane modified oxide and modified surface light patterning oxide
DE4332003C2 (en) 1993-09-21 1996-02-22 Seeger Stefan Process for coating surfaces with biomolecules and other receptor molecules
WO1995011755A1 (en) * 1993-10-28 1995-05-04 Houston Advanced Research Center Microfabricated, flowthrough porous apparatus for discrete detection of binding reactions
US5429708A (en) 1993-12-22 1995-07-04 The Board Of Trustees Of The Leland Stanford Junior University Molecular layers covalently bonded to silicon surfaces
EP0664452B1 (en) * 1994-01-19 2002-07-31 Roche Diagnostics GmbH Biotin-silane compounds and binding matrix containing these compounds
DE4435728A1 (en) * 1994-01-19 1995-07-20 Boehringer Mannheim Gmbh Biotin silane compounds and binding matrix containing these compounds
US6287850B1 (en) * 1995-06-07 2001-09-11 Affymetrix, Inc. Bioarray chip reaction apparatus and its manufacture
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
AU2897595A (en) 1994-07-14 1996-02-16 Technobiochip Biosensor and method and instrument for deposition of alternating monomolecular layers
SE9403245D0 (en) 1994-09-26 1994-09-26 Pharmacia Biosensor Ab Improvements relating to bilayer lipid membranes
US5571410A (en) 1994-10-19 1996-11-05 Hewlett Packard Company Fully integrated miniaturized planar liquid sample handling and analysis device
US5688642A (en) 1994-12-01 1997-11-18 The United States Of America As Represented By The Secretary Of The Navy Selective attachment of nucleic acid molecules to patterned self-assembled surfaces
US5814565A (en) 1995-02-23 1998-09-29 University Of Utah Research Foundation Integrated optic waveguide immunosensor
EP0812434B1 (en) 1995-03-01 2013-09-18 President and Fellows of Harvard College Microcontact printing on surfaces and derivative articles
WO1996027614A1 (en) 1995-03-08 1996-09-12 Elias Klein Surface modified affinity separation membrane
US6140045A (en) * 1995-03-10 2000-10-31 Meso Scale Technologies Multi-array, multi-specific electrochemiluminescence testing
US6309820B1 (en) * 1995-04-07 2001-10-30 University Of North Carolina At Chapel Hill Polypeptides having a functional domain of interest and methods of identifying and using same
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
WO1996038726A1 (en) 1995-05-30 1996-12-05 Ecole Polytechnique Federale De Lausanne (Epfl) Covalently immobilized phospholipid bilayers on solid surfaces
US5776674A (en) 1995-06-05 1998-07-07 Seq, Ltd Chemical biochemical and biological processing in thin films
WO1996039937A1 (en) 1995-06-07 1996-12-19 The Regents Of The University Of California Microfabricated devices for diagnostic applications
US6518168B1 (en) 1995-08-18 2003-02-11 President And Fellows Of Harvard College Self-assembled monolayer directed patterning of surfaces
DE19543232A1 (en) 1995-11-07 1997-05-15 Knoell Hans Forschung Ev Production of matrix-bound miniaturised combinatorial polymer and oligomer library
ATE271219T1 (en) 1995-12-01 2004-07-15 Innogenetics Nv SYSTEM AND METHOD FOR DETERMINING IMPEDANCE AND PRODUCTION PROCESS
DE19548152A1 (en) 1995-12-22 1997-06-26 Boehringer Mannheim Gmbh Process for covering a surface with a film of an oligoethylene glycol derivative
ES2166987T3 (en) 1996-03-15 2002-05-01 Harvard College METHOD OF FORMING ARTICLES AND MODELED SURFACES THROUGH CAPILLARY MICROMOLDING.
DE69700499T2 (en) 1996-04-03 2000-03-23 Perkin Elmer Corp DEVICE AND METHOD FOR DETECTING SEVERAL ANALYZES
US5942443A (en) 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US6165335A (en) 1996-04-25 2000-12-26 Pence And Mcgill University Biosensor device and method
US5925552A (en) 1996-04-25 1999-07-20 Medtronic, Inc. Method for attachment of biomolecules to medical devices surfaces
DE69719817T2 (en) 1996-04-25 2003-12-24 Pence Inc BIOSENSOR DEVICE AND METHOD
US6075875A (en) * 1996-09-30 2000-06-13 Microsoft Corporation Segmentation of image features using hierarchical analysis of multi-valued image data and weighted averaging of segmentation results
US5837860A (en) 1997-03-05 1998-11-17 Molecular Tool, Inc. Covalent attachment of nucleic acid molecules onto solid-phases via disulfide bonds
US6180288B1 (en) * 1997-03-21 2001-01-30 Kimberly-Clark Worldwide, Inc. Gel sensors and method of use thereof
WO1998050773A2 (en) 1997-05-08 1998-11-12 University Of Minnesota Microcantilever biosensor
WO1999019510A1 (en) * 1997-10-10 1999-04-22 President And Fellows Of Harvard College Surface-bound, double-stranded dna protein arrays
AU2583899A (en) 1998-02-04 1999-08-23 Invitrogen Corporation Microarrays and uses therefor
US6087103A (en) * 1998-03-04 2000-07-11 Lifespan Biosciences, Inc. Tagged ligand arrays for identifying target-ligand interactions
US6287765B1 (en) * 1998-05-20 2001-09-11 Molecular Machines, Inc. Methods for detecting and identifying single molecules
US6576478B1 (en) * 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
AU3387700A (en) 1999-03-02 2000-09-21 Chiron Corporation Microarrays for identifying pathway activation or induction
EP1159615A2 (en) 1999-03-10 2001-12-05 National Institutes of Health, as represented by the Secretary, Department of Health and Human Services of the Government Universal protein array system
WO2000053625A2 (en) 1999-03-11 2000-09-14 Combimatrix Corporation Microarrays of peptide affinity probes for analyzing gene products and methods for analyzing gene products
US6406840B1 (en) * 1999-12-17 2002-06-18 Biomosaic Systems, Inc. Cell arrays and the uses thereof

Patent Citations (102)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4071409A (en) * 1976-05-20 1978-01-31 Corning Glass Works Immobilization of proteins on inorganic support materials
US4722896A (en) * 1981-01-26 1988-02-02 The Beth Israel Hospital Association Method for affinity purification of hybridoma antibodies
US4444879A (en) * 1981-01-29 1984-04-24 Science Research Center, Inc. Immunoassay with article having support film and immunological counterpart of analyte
US4514508A (en) * 1982-07-06 1985-04-30 Biond Inc. Assaying for a multiplicity of antigens or antibodies with a detection compound
US4994373A (en) * 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5096807A (en) * 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
US5866362A (en) * 1985-03-28 1999-02-02 Chiron Corporation Enhanced purification and expression of insoluble recombinant proteins
US5866363A (en) * 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
US4894146A (en) * 1986-01-27 1990-01-16 University Of Utah Thin channel split flow process and apparatus for particle fractionation
US4802951A (en) * 1986-03-07 1989-02-07 Trustees Of Boston University Method for parallel fabrication of nanometer scale multi-device structures
US5643948A (en) * 1986-06-11 1997-07-01 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. K.
US5079600A (en) * 1987-03-06 1992-01-07 Schnur Joel M High resolution patterning on solid substrates
US4987032A (en) * 1987-06-26 1991-01-22 Fuji Photo Film Co., Ltd. Functional organic thin film and method of manufacture thereof
US6692751B1 (en) * 1988-05-06 2004-02-17 New York Blood Center Methods and systems for producing recombinant viral antigens
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5281540A (en) * 1988-08-02 1994-01-25 Abbott Laboratories Test array for performing assays
US5720928A (en) * 1988-09-15 1998-02-24 New York University Image processing and analysis of individual nucleic acid molecules
US5744305A (en) * 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US6197506B1 (en) * 1989-06-07 2001-03-06 Affymetrix, Inc. Method of detecting nucleic acids
US5405783A (en) * 1989-06-07 1995-04-11 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of an array of polymers
US5424186A (en) * 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US6346413B1 (en) * 1989-06-07 2002-02-12 Affymetrix, Inc. Polymer arrays
US5489678A (en) * 1989-06-07 1996-02-06 Affymax Technologies N.V. Photolabile nucleoside and peptide protecting groups
US5510270A (en) * 1989-06-07 1996-04-23 Affymax Technologies N.V. Synthesis and screening of immobilized oligonucleotide arrays
US5283173A (en) * 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5858188A (en) * 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
US5466589A (en) * 1990-11-19 1995-11-14 Biotechnology Research & Development Corporation Coated substrates employing oriented layers of mutant heme proteins and methods of making and using same
US5294369A (en) * 1990-12-05 1994-03-15 Akzo N.V. Ligand gold bonding
US6544739B1 (en) * 1990-12-06 2003-04-08 Affymetrix, Inc. Method for marking samples
US5763170A (en) * 1991-04-16 1998-06-09 Amersham International Plc Method for forming an array of biological particles
US5516635A (en) * 1991-10-15 1996-05-14 Ekins; Roger P. Binding assay employing labelled reagent
US5384261A (en) * 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US6040193A (en) * 1991-11-22 2000-03-21 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US5885793A (en) * 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5296114A (en) * 1991-12-06 1994-03-22 Ciba-Geigy Corporation Electrophoretic separating device and electrophoretic separating method
US5405766A (en) * 1992-03-26 1995-04-11 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of National Defence Immobilization of biologically active protein on a support with a 7-18 carbon spacer and a bifunctional phospholipid
US5412087A (en) * 1992-04-24 1995-05-02 Affymax Technologies N.V. Spatially-addressable immobilization of oligonucleotides and other biological polymers on surfaces
US5866345A (en) * 1992-05-01 1999-02-02 The Trustees Of The University Of Pennsylvania Apparatus for the detection of an analyte utilizing mesoscale flow systems
US5726026A (en) * 1992-05-01 1998-03-10 Trustees Of The University Of Pennsylvania Mesoscale sample preparation device and systems for determination and processing of analytes
US5635358A (en) * 1992-05-01 1997-06-03 Trustees Of The University Of Pennsylvania Fluid handling methods for use in mesoscale analytical devices
US5637469A (en) * 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5506121A (en) * 1992-11-03 1996-04-09 Institut Fur Bioanalytik Gemeinnutzige Gesellschaft MBH Fusion peptides with binding activity for streptavidin
US5532142A (en) * 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
US5512492A (en) * 1993-05-18 1996-04-30 University Of Utah Research Foundation Waveguide immunosensor with coating chemistry providing enhanced sensitivity
US5719060A (en) * 1993-05-28 1998-02-17 Baylor College Of Medicine Method and apparatus for desorption and ionization of analytes
US5861242A (en) * 1993-06-25 1999-01-19 Affymetrix, Inc. Array of nucleic acid probes on biological chips for diagnosis of HIV and methods of using the same
US5741700A (en) * 1993-08-11 1998-04-21 University Of Chicago Method of immobilizing water-soluble bioorganic compounds on a capillary-porous carrier
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5605662A (en) * 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US6051380A (en) * 1993-11-01 2000-04-18 Nanogen, Inc. Methods and procedures for molecular biological analysis and diagnostics
US5623055A (en) * 1994-01-28 1997-04-22 Prolinx, Inc. Phenylboronic acid complexes derived from aminosalicylic acid for bioconjugate preparation
US5594111A (en) * 1994-01-28 1997-01-14 Prolinx, Inc. Phenylboronic acid complexes for bioconjugate preparation
US5520787A (en) * 1994-02-09 1996-05-28 Abbott Laboratories Diagnostic flow cell device
US5514501A (en) * 1994-06-07 1996-05-07 The United States Of America As Represented By The Secretary Of Commerce Process for UV-photopatterning of thiolate monolayers self-assembled on gold, silver and other substrates
US5498545A (en) * 1994-07-21 1996-03-12 Vestal; Marvin L. Mass spectrometer system and method for matrix-assisted laser desorption measurements
US5620850A (en) * 1994-09-26 1997-04-15 President And Fellows Of Harvard College Molecular recognition at surfaces derivatized with self-assembled monolayers
US5858804A (en) * 1994-11-10 1999-01-12 Sarnoff Corporation Immunological assay conducted in a microlaboratory array
US5593838A (en) * 1994-11-10 1997-01-14 David Sarnoff Research Center, Inc. Partitioned microelectronic device array
US5622826A (en) * 1994-12-22 1997-04-22 Houston Advanced Research Center Method for immobilization of molecules on platinum solid support surfaces
US5629213A (en) * 1995-03-03 1997-05-13 Kornguth; Steven E. Analytical biosensor
US6066448A (en) * 1995-03-10 2000-05-23 Meso Sclae Technologies, Llc. Multi-array, multi-specific electrochemiluminescence testing
US5624711A (en) * 1995-04-27 1997-04-29 Affymax Technologies, N.V. Derivatization of solid supports and methods for oligomer synthesis
US5627369A (en) * 1995-05-19 1997-05-06 Perseptive Biosystems, Inc. Time-of-flight mass spectrometry analysis of biomolecules
US5603351A (en) * 1995-06-07 1997-02-18 David Sarnoff Research Center, Inc. Method and system for inhibiting cross-contamination in fluids of combinatorial chemistry device
US5874219A (en) * 1995-06-07 1999-02-23 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US6720149B1 (en) * 1995-06-07 2004-04-13 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US6194612B1 (en) * 1995-10-17 2001-02-27 The Scripps Research Institute Template for solution phase synthesis of combination libraries
US5763263A (en) * 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
US5731152A (en) * 1996-05-13 1998-03-24 Motorola, Inc. Methods and systems for biological reagent placement
US20030017149A1 (en) * 1996-10-10 2003-01-23 Hoeffler James P. Single chain monoclonal antibody fusion reagents that regulate transcription in vivo
US6064754A (en) * 1996-11-29 2000-05-16 Oxford Glycosciences (Uk) Ltd. Computer-assisted methods and apparatus for identification and characterization of biomolecules in a biological sample
US6228326B1 (en) * 1996-11-29 2001-05-08 The Board Of Trustees Of The Leland Stanford Junior University Arrays of independently-addressable supported fluid bilayer membranes
US5905024A (en) * 1996-12-17 1999-05-18 University Of Chicago Method for performing site-specific affinity fractionation for use in DNA sequencing
US5861254A (en) * 1997-01-31 1999-01-19 Nexstar Pharmaceuticals, Inc. Flow cell SELEX
US6190619B1 (en) * 1997-06-11 2001-02-20 Argonaut Technologies, Inc. Systems and methods for parallel synthesis of compounds
US6225047B1 (en) * 1997-06-20 2001-05-01 Ciphergen Biosystems, Inc. Use of retentate chromatography to generate difference maps
US6881586B2 (en) * 1997-06-20 2005-04-19 Ciphergen Biosystems, Inc. Retentate chromatography and protein chip arrays with applications in biology and medicine
US6844165B2 (en) * 1997-06-20 2005-01-18 Ciphergen Biosystems, Inc. Retentate chromatography and protein chip arrays with applications in biology and medicine
US5948621A (en) * 1997-09-30 1999-09-07 The United States Of America As Represented By The Secretary Of The Navy Direct molecular patterning using a micro-stamp gel
US6061476A (en) * 1997-11-24 2000-05-09 Cognex Corporation Method and apparatus using image subtraction and dynamic thresholding
US6232066B1 (en) * 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6350369B1 (en) * 1998-04-14 2002-02-26 California Institute Of Technology Method and system for determining analyte activity
US20050014292A1 (en) * 1998-07-14 2005-01-20 Peter Wagner Protein arrays for high-throughput screening
US20060035387A1 (en) * 1998-07-14 2006-02-16 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US6682942B1 (en) * 1998-07-14 2004-01-27 Zyomyx, Inc. Microdevices for screening biomolecules
US6897073B2 (en) * 1998-07-14 2005-05-24 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US20050100947A1 (en) * 1998-07-14 2005-05-12 Zyomyx, Inc. Array devices and methods of use thereof
US6365418B1 (en) * 1998-07-14 2002-04-02 Zyomyx, Incorporated Arrays of protein-capture agents and methods of use thereof
US20030003599A1 (en) * 1998-07-14 2003-01-02 Peter Wagner Arrays of protein-capture agents and methods of use thereof
US6197599B1 (en) * 1998-07-30 2001-03-06 Guorong Chin Method to detect proteins
US6190908B1 (en) * 1998-08-12 2001-02-20 The Scripps Research Institute Modulation of polypeptide display on modified filamentous phage
US6391625B1 (en) * 1999-06-28 2002-05-21 Lg Electronics Inc. Biochip and method for patterning and measuring biomaterial of the same
US6899137B2 (en) * 1999-06-28 2005-05-31 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6720157B2 (en) * 2000-02-23 2004-04-13 Zyomyx, Inc. Chips having elevated sample surfaces
US6531283B1 (en) * 2000-06-20 2003-03-11 Molecular Staging, Inc. Protein expression profiling
US20030073811A1 (en) * 2000-08-17 2003-04-17 Sense Proteomic Limited Method
US6699665B1 (en) * 2000-11-08 2004-03-02 Surface Logix, Inc. Multiple array system for integrating bioarrays
US20050095646A1 (en) * 2001-11-19 2005-05-05 Sherman Michael I. Method of using a non-antibody protein to detect and measure an analyte
US20050026215A1 (en) * 2003-07-17 2005-02-03 Predki Paul F. Method for the prediction of an epitope

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Wagner et al., Formation and in situ modification of monolayer chemisorbed on ultraflat template-stripped gold surfaces, 1995, Langmuir, 11, 3867,3875 *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637264B2 (en) 1998-02-04 2014-01-28 Life Technologies Corporation Microarrays and uses therefor
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
US8012703B2 (en) 1998-02-04 2011-09-06 Life Technologies Corporation Microarrays and uses therefor
US20050014292A1 (en) * 1998-07-14 2005-01-20 Peter Wagner Protein arrays for high-throughput screening
US20030003599A1 (en) * 1998-07-14 2003-01-02 Peter Wagner Arrays of protein-capture agents and methods of use thereof
US20030138973A1 (en) * 1998-07-14 2003-07-24 Peter Wagner Microdevices for screening biomolecules
US20040241751A1 (en) * 1998-07-14 2004-12-02 Peter Wagner Arrays of protein-capture agents and methods of use thereof
US20020106702A1 (en) * 1998-07-14 2002-08-08 Peter Wagner Protein arrays for high-throughput screening
US20050100947A1 (en) * 1998-07-14 2005-05-12 Zyomyx, Inc. Array devices and methods of use thereof
US20020115225A1 (en) * 1998-07-14 2002-08-22 Peter Wagner Microdevices for high-throughput screening of biomolecules
US20020110933A1 (en) * 1998-07-14 2002-08-15 Peter Wagner Arrays of proteins and methods of use thereof
US20030207467A1 (en) * 2000-05-04 2003-11-06 Michael Snyder Protein chips for high throughput screening of protein activity
US8399383B2 (en) 2000-05-04 2013-03-19 Yale University Protein chips for high throughput screening of protein activity
US20050048580A1 (en) * 2001-01-23 2005-03-03 President And Fellows Of Harvard College Nucleic-acid programmable protein arrays
US8609344B2 (en) 2001-01-23 2013-12-17 President And Fellows Of Harvard College Nucleic-acid programmable protein arrays
US20060073593A1 (en) * 2001-02-07 2006-04-06 Invitrogen Corporation Compositions and methods for molecular biology
US7687437B2 (en) 2001-07-13 2010-03-30 Nanosphere, Inc. Method for immobilizing molecules onto surfaces
US20030082588A1 (en) * 2001-07-13 2003-05-01 Viswanadham Garimella Method for immobilizing molecules onto surfaces
US20050287560A1 (en) * 2001-07-13 2005-12-29 Nanosphere, Inc. Method for preparing substrates having immobilized molecules and substrates
US20070134714A1 (en) * 2002-05-28 2007-06-14 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
US20070172872A1 (en) * 2002-05-28 2007-07-26 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
US20070292843A1 (en) * 2002-05-28 2007-12-20 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
US20070141615A1 (en) * 2002-05-28 2007-06-21 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
US20080097115A1 (en) * 2002-05-28 2008-04-24 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
US20050233473A1 (en) * 2002-08-16 2005-10-20 Zyomyx, Inc. Methods and reagents for surface functionalization
US20050118665A1 (en) * 2003-06-09 2005-06-02 Zhou Fang X. Methods for conducting assays for enzyme activity on protein microarrays
US20050260653A1 (en) * 2004-04-14 2005-11-24 Joshua Labaer Nucleic-acid programmable protein arrays
US8178316B2 (en) 2006-06-29 2012-05-15 President And Fellows Of Harvard College Evaluating proteins
US20080071071A1 (en) * 2006-06-29 2008-03-20 President And Fellows Of Harvard College Evaluating proteins
US20080124719A1 (en) * 2006-07-25 2008-05-29 Samsung Electronics Co., Ltd. Patterned spot microarray using photocatalyst and method of manufacturing the same
US8273533B2 (en) 2006-07-25 2012-09-25 Samsung Electronics Co., Ltd. Patterned spot microarray using photocatalyst and method of manufacturing the same
EP1882520A1 (en) * 2006-07-25 2008-01-30 Samsung Electronics Co., Ltd. Patterned spot microarray using photocatalyst and a method of manufacturing the same
US9694518B2 (en) 2014-06-20 2017-07-04 The Regents Of The University Of Michigan Breath-activated images and anti-counterfeit authentication features formed of nanopillar arrays

Also Published As

Publication number Publication date
CA2337075A1 (en) 2000-01-27
JP2002520620A (en) 2002-07-09
US6630358B1 (en) 2003-10-07
US6475808B1 (en) 2002-11-05
AU5102599A (en) 2000-02-07
EP1097377A2 (en) 2001-05-09
US6365418B1 (en) 2002-04-02
AU773068B2 (en) 2004-05-13
US6406921B1 (en) 2002-06-18
US20030003599A1 (en) 2003-01-02
AU5102399A (en) 2000-02-07
US20090131278A1 (en) 2009-05-21
DE69938867D1 (en) 2008-07-17
US20020110933A1 (en) 2002-08-15
WO2000004389A3 (en) 2000-04-27
WO2000004389A2 (en) 2000-01-27
US20110086779A1 (en) 2011-04-14
CA2337654A1 (en) 2000-01-27
EP1097377B1 (en) 2008-06-04
JP2002520618A (en) 2002-07-09
AU765508B2 (en) 2003-09-18
US20020106702A1 (en) 2002-08-08
US6475809B1 (en) 2002-11-05
US20050014292A1 (en) 2005-01-20
ATE397752T1 (en) 2008-06-15
WO2000004382A1 (en) 2000-01-27
EP1097380A1 (en) 2001-05-09
WO2000004382A8 (en) 2001-03-15
US6329209B1 (en) 2001-12-11
US20060228701A1 (en) 2006-10-12

Similar Documents

Publication Publication Date Title
US6475809B1 (en) Protein arrays for high-throughput screening
US6576478B1 (en) Microdevices for high-throughput screening of biomolecules
US6682942B1 (en) Microdevices for screening biomolecules
US7247469B2 (en) Non-specific binding resistant protein arrays and methods for making the same
US20090042744A1 (en) Microdevices for screening biomolecules
US6780582B1 (en) Arrays of protein-capture agents and methods of use thereof
US20020119579A1 (en) Arrays devices and methods of use thereof
AU2003262452B2 (en) Arrays of proteins and methods of use thereof I
AU2003257898A1 (en) Microdevices for screening biomolecules

Legal Events

Date Code Title Description
AS Assignment

Owner name: BILL & MELINDA GATES FOUNDATION, WASHINGTON

Free format text: SECURITY AGREEMENT;ASSIGNOR:ZYOMYX, INC.;REEL/FRAME:026240/0824

Effective date: 20110506

STCB Information on status: application discontinuation

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