US20040023366A1 - Surface for the immobilization of ligands - Google Patents

Surface for the immobilization of ligands Download PDF

Info

Publication number
US20040023366A1
US20040023366A1 US10/297,244 US29724403A US2004023366A1 US 20040023366 A1 US20040023366 A1 US 20040023366A1 US 29724403 A US29724403 A US 29724403A US 2004023366 A1 US2004023366 A1 US 2004023366A1
Authority
US
United States
Prior art keywords
binding
anchor
measuring
interaction
molecules
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
US10/297,244
Other versions
US20040235146A9 (en
Inventor
Stefan Dickopf
Holger Ottleben
Harald Rau
Renate Sekul
Kristina Schmidt
Dirk Vetter
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
Publication of US20040023366A1 publication Critical patent/US20040023366A1/en
Publication of US20040235146A9 publication Critical patent/US20040235146A9/en
Priority to US11/980,015 priority Critical patent/US7955638B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/551Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being inorganic
    • G01N33/553Metal or metal coated
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y15/00Nanotechnology for interacting, sensing or actuating, e.g. quantum dots as markers in protein assays or molecular motors
    • 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
    • G01N33/54353Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals with ligand attached to the carrier via a chemical coupling agent

Definitions

  • the present invention relates to binding surfaces for the immobilization of ligands, ligand surfaces and structured surface arrays which present a plurality of identical or different ligands.
  • the invention further relates to a process for the production and the use of such surfaces and to specific binder molecules which can be used for the preparation thereof.
  • receptor-ligand systems wherein the receptor is usually a biomacromolecule (e.g. a protein or single-stranded DNA) and the ligand is a “probe”, a molecule usually of low molecular weight and of biological or synthetic origin (peptides, oligonucleotides or what are referred to as small organic molecules).
  • a biomacromolecule e.g. a protein or single-stranded DNA
  • the ligand is a “probe”, a molecule usually of low molecular weight and of biological or synthetic origin (peptides, oligonucleotides or what are referred to as small organic molecules).
  • ligands exhibit highly specific structural properties which can interact with a receptor provided it exhibits corresponding structures.
  • the binding to the receptor can take place via one or several ligands.
  • systems are studied wherein both interaction partners are macromolecules (e.g. protein-protein interaction).
  • Interaction analysis is used in the pharmaceutical and agrochemical industries for researching active substances. The goal is to analyze as large a number of different samples as possible in as short a time period as possible. There is a growing interest to develop testing systems that are able to simultaneously subject a large number of both identical and different molecules to a certain test method for detecting their biospecific binding behavior in order to identify molecules with a specific effect (High-Throughput Screening, HTS). Such a parallelization is closely linked to a simultaneous miniaturization of the testing arrangement and automation of the testing procedure. Furthermore, interaction analysis is used in the research of the genome (polymorphisms (SNP) analysis or expression pattern analysis) and also in food analysis.
  • SNP polymorphisms
  • the generation of a bioactive surface using such anchors can be carried out in one or several steps.
  • the interaction partner is bonded to the head group of the anchor prior to its immobilization.
  • Suitable anchors for the single-step process are described in DE 199 24 606.8.
  • LAC ligand-anchor conjugate
  • a binding surface in the form of an organic boundary layer is generated which does not yet exhibit the desired specific structural features, however, which is suitable for binding or can be activated to bind the interaction partners to be immobilized, e.g. ligands.
  • immobilization of an interaction partner takes place after the generation of a first boundary layer that is not bond-specific by means of a covalent, ionic or complex bond via the head group of the binding component.
  • the head group can be used directly or after it has been activated. Examples of such head groups are described in EP 485 874 A2.
  • An important advantage of the multi-step process is the fact that the same surface can be used for every interaction partner to be immobilized. Therefore, the resulting concentration of the interaction partners to be immobilized is the same on the surface as well, which is not or not always a given in the single-step process. This is particularly important for the comparability and reproducibility of the different measuring results.
  • the chemical homogeneity of the non-biospecific binding surface decreases again as the number of potentially necessary activation steps increases.
  • a step-by-step formation of a diluted binding layer is described in WO 98/40739 A1, wherein in a first step cystamine is applied onto a gold surface and the provision of maleimide as head group is realized in a second chemical reaction.
  • the disadvantage of this process is that another activation step is required for obtaining a binding surface.
  • reaction partner is often a macromolecule carrying a maleimidyl group.
  • the product of the reaction of the thiol and the maleimidyl group is a thio succinimide (Michael addition) (G. T. Hermanson, Bioconjugate Techniques, Academic Press 1996, 229-252; Suda et al. Biochem. Biophys. Res. Comm. (1999), 261, 276-282).
  • reaction partners for the thiol group are known from the prior art (referred to in the following as mercaptophiles), which also allow its selective and quantitative reaction.
  • maleimides include the following compounds:
  • the thiol/mercaptophile system offers the essential advantage that the binding of the interaction partner to be immobilized can be carried out under gentle reaction conditions (ambient temperature, neutral pH, buffer solutions can be used). This is of particular importance when unstable compounds or proteins that denature easily are used. Another advantage is that compared to carboxylic acid, amine or amide groups, a thiol functionality hardly ever occurs in active substances. Thus, an undesired reaction of mercaptophiles that may remain after the immobilization of one interaction partner and the target can largely be avoided.
  • the thiol/maleimide system is furthermore particularly known for its high degree of selectivity compared to other functionalities such as hydroxyl, amine, carboxyl or hydroxylamine groups.
  • the formation of the covalent bond is characterized by a high reaction rate (cf. Schelté et al., Bioconj. Chem. (2000), 11, 118-123).
  • anchors comprising both a thiol group for binding to the solid phase surface and a mercaptophilic head group for immobilizing an interaction partner are not disclosed in the prior art since both during synthesis and during the generation of the binding surface both intra- and intermolecular side reactions, and even polymerization, may occur (with respect to the purposeful polymerization of oligothiols with bis-maleimides, cf. L. R. Dix et al., Eur. Polym. J. 31 (1995), 653-658).
  • EP 485 874 A2 refers to this problem, which is avoided by the exclusive use of disulfide and sulfide groups in the anchor, in order to use for example maleimide as head group for the immobilization of proteins (reaction with —SH group of a Fab′ fragment).
  • EP 698 787 A1 uses short-chain, maleimide-carrying disulfide anchors for the immobilization of proteins.
  • anchors based on disulfide and sulfide groups show the disadvantage that an undesired spatial proximity of the head groups is generated. In this case, the head groups can interfere with each other in the immobilization reaction.
  • EP 485 874 A2 and EP 698 787 A1 do not only apply anchor molecules onto the solid phase surface. Rather, the anchor molecules are “diluted”. This is due to the fact that if one partner, for example a ligand is bound to a solid phase carrier for interaction analysis, adjacent ligands on the surface can interfere with each other or with the interaction between the adjacent ligand and the interaction partner to be detected.
  • mixed surfaces as shown in FIG. 1—can be applied, which are composed of ligand-carrying anchor molecules and so-called diluting molecules that do not carry any ligands and thus dilute the measuring surface.
  • the structural nature of the diluting component has to meet the prerequisite that it will not influence the interaction of the immobilized interaction partner and the free interaction partner. In particular, if possible, no specific or unspecific binding between the free interaction partner and the diluting component should occur (e.g. diluant with as high a resistance to protein adsorption as possible). Furthermore, the anchor molecule and the diluting component should be as structurally similar as possible to ensure that their mixing behavior on the solid phase surface is as homogeneous as possible.
  • binding surfaces presenting reactive head groups which are capable of entering into selective and highly quantitative reactions with interaction partners to be immobilized.
  • the present invention relates to novel anchor molecules comprising these head groups which can be used, together with diluting molecules, to provide such binding surfaces.
  • the invention further relates to sensor or measuring surfaces obtainable by immobilizing specific interaction partners on the binding surfaces of the present invention, as well as to surface arrays comprising a plurality of such identical or different measuring surfaces.
  • a binding surface according to the present invention comprises a solid phase carrier on which an organic monolayer can assemble spontaneously, preferably comprising at least two components, a binding component and a diluting component (anchor and diluant).
  • the solid phase carrier consists of a substrate formed by a metal, preferably a noble metal, especially preferred gold, or carries a layer of such a metal.
  • the metal layer can be applied with the help of an intermediate layer which serves as a primer.
  • the material used for the substrate depends on measuring method employed. If optical reflection processes such as surface plasmone resonance (SPR) are used, the substrate preferably consists of glass or a plastic material. Due to the use of sulfur-containing compounds for the immobilization of ligands, a gold layer and a chromium layer as a primer are preferably applied onto such substrates.
  • the spatial design of the solid phase carrier to be used in accordance with the present invention is not restricted, although planar, two-dimensional structures are preferably used for sensor applications. However, depending on the field of application, three-dimensional shapes such as spheres or hollow bodies may be used as well.
  • Processes for the Generation of the binding layers according to the present invention start from a solution of the anchor molecules which is brought into contact with the solid phase carrier.
  • Preferred diluted measuring surfaces are obtained when a solution is used that contains a mixture of anchor molecules and diluting components in a certain molar ratio.
  • the solution has a molar ratio of anchor/diluant between 1:2 and 1:10,000.
  • Especially preferred dilution ratios range from 1:10 to 1:100.
  • the total concentration of anchor and diluting components preferably lies in the range of from 0.001 to 100 mmol/l, especially preferred about 0.1 to 1 mmol/l.
  • the entire surface of the solid phase carrier is coated, for example by immersing in a tub, with the (diluted) binding layer.
  • the solution for applying the solution in a spatially defined manner, commercially available pipetting or spotting devices, as well as micropipetting devices or ink-jet processes can be used.
  • a spatial structure of the measuring surface is achieved by the selective application of the interaction partners to be immobilized onto the large-area binding surface.
  • Suitable solvents include aqueous (e.g. buffer solutions) or organic solvents (e.g. methanol, ethanol, acetonitrile. N,N-dimethylformamide, ethylene glycol) or mixtures thereof.
  • the anchor molecule solutions preferably comprise an acid such as trifluoroacetic acid, hydrochloric acid or phosphoric acid.
  • Anchor molecules suitable for preparing the binding surface according to the present invention comprise at least two functional units present at opposite ends of the anchor. As a group allowing the bonding of the anchor with the surface of a carrier, these anchors comprise a thiol group. For binding the interaction partner to be immobilized to the anchor, the anchor comprises a mercaptophilic head group M as a second functional unit.
  • the anchors according to the present invention have the following general structure:
  • Suitable structural elements R that both promote the formation of a monolayer and optionally also allow the adjustment of suitable distances between the head groups and the carrier surface with the help of spacer groups are described with respect to the anchor molecules in the German patent application DE 199 24 606.8, whose entire corresponding disclosure content is hereby incorporated by reference.
  • the moiety R is a branched or straight-chain, optionally substituted, saturated or unsaturated hydrocarbon chain which may be interrupted by heteroatoms, aromatic and heterocyclic compounds and comprises 5 to 2,000 atoms, including heteroatoms.
  • the anchor should comprise structures that impede or prevent a passive adsorption of the free interaction partner both on the anchor structure and the measuring surface.
  • the anchor comprises a spacer group which allows adjustment of the total chain length and of the flexibility of the LAC.
  • R a causes the formation of an SAM and for this purpose, it should essentially be hydrophobic. It comprises a branched or straight hydrocarbon chain with 5 to 50 carbon atoms, which may be saturated or partially unsaturated and which may be interrupted by aromatic or heterocyclic groups or heteroatoms. A fully saturated hydrocarbon chain without heteroatoms is preferred. In a preferred embodiment, it has the general formula —(CH 2 ) n 13 , wherein n is an integer of 5 to 50, preferably 5 to 25, especially preferred 5 to 18 and most preferred 8 to 12.
  • R b is preferably a spacer that allows adjustment of the total chain length and of the flexibility of the ligand-anchor conjugate.
  • R b is a hydrocarbon chain which is interrupted by heteroatoms and therefore hydrophilic and impedes a passive adsorption of the receptor.
  • the chain comprises 2 to 1,000 atoms, including heteroatoms; chain lengths of 5 to 500 are preferred, and especially preferred are chain lengths between 10 and 100 atoms.
  • R b is an oligoether of the general formula —(OAlk) y — wherein y is a natural number and Alk represents an alkylene group.
  • y is a natural number and Alk represents an alkylene group.
  • Alk represents an alkylene group.
  • the Alk group preferably comprises 1 to 20, more preferably 2 to 10 and particularly preferred 2 to 5 carbon atoms. Especially preferred is —(OC 2 H 4 ) y —.
  • R b is an oligoamide consisting of dicarboxylic acids and diamines and/or aminocarboxylic acids, wherein the amines independently preferably comprise 1 to 20, more preferably 1 to 10 carbon atoms, and may also be interrupted by further heteroatoms, in particular oxygen atoms.
  • the carboxylic acid monomers independently preferably comprise 1 to 20, more preferably 1 to 10 carbon atoms and may also be interrupted by further heteroatoms, in particular oxygen atoms.
  • Suitable compounds for the introduction of R b in an especially preferred embodiment include commercially available compounds such as in particular glycol ethers such as e.g. triethylene glycol, triethylene glycol monomethylether, tetraethylene glycol, ⁇ -diamines such as ethylene-, propylene-, butylene-, or pentylenediamine or 1,8-diamino-3,6-dioxaoctane, but also dicarboxylic acids such as succinic acid, 1,13-diamino-4,7,10-triooxatridecane, 3,6,9-trioxaundecanedioic acid, 8-amino-3,6-dioxaoctanoic acid or 4-aminobenzoic acid as well as their derivatives or combinations of identical components (such as e.g.
  • glycol ethers such as e.g. triethylene glycol, triethylene glycol monomethylether, tetraethylene glycol,
  • 4-aminobenzoic acid in the case of 8-amino-3,6-dioxaoctanoic acid or 4-aminobenzoic acid) or combinations of different components (such as e.g. 1,13-diamino-4,7,10-triooxatridecane and 3,6,9-trioxaundecanedioic acid in an alternating sequence).
  • 4-aminobenzoic acid is the fact that it can easily be detected spectroscopically, for example by means of a UV spectroscopy.
  • Especially preferred moieties R have the following general formula:
  • Q 1 ,Q 5 are —NH—C(O)—, —C(O)—NH— or a bond
  • Q 2 ,Q 3 ,Q 4 are —NH—C(O)— or —C(O)—NH—
  • a is 5 to 20, preferably 8 to 12, particularly preferred 10;
  • b is 0 to 5, preferably 0 if Q 1 is a bond and 1 to 10, preferably 2 to 7, particularly preferred 3 to 5 in all other cases;
  • c, c′ are 1 to 5, preferably 1 to 3, particularly preferred 1;
  • d, d′ are 1 to 5, preferably 1 to 3, particularly preferred 2;
  • e, e′ are 1 to 5, preferably 1 to 3, particularly preferred 2;
  • f, f′ are 1 to 5, preferably 1 to 3, particularly preferred 1;
  • i is 1 to 3, preferably 1 to 2, particularly preferred 1;
  • j is 0 to 5, preferably 1 to 3, particularly preferred 2;
  • k is 0 to 5.
  • Mercaptophilic head groups M are for example iodo- or bromoacetamide, pyridylthio compounds, Michael acceptors in general, acrylic acid derivatives such as its esters, amides, lactones or lactames, methylene-gem-difluorocyclopropanes, ⁇ , ⁇ -unsaturated aldehydes and ketones as well as ⁇ , ⁇ -unsaturated sulfones and sulfonamides.
  • Preferred head groups M are those of the general formula
  • R 1 and R 2 are independently hydrogen or C 1 -C 5 alkyl, preferably methyl, ethyl or n-propyl,
  • R 3 and R 4 are independently hydrogen or C 1 -C 5 alkyl, preferably methyl, ethyl or n-propyl, or R 3 and R 4 together form ⁇ O and
  • R 3 and R 4 together form ⁇ O, and most preferred, the head group is a maleimidyl group.
  • Processes for the synthesis of an anchor molecule can be carried out in a solid phase or in solution.
  • the thiol group is preferably provided with a protective group. Suitable protective groups are described in T. W. Greene, P. G. M. Wuts, Protective Groups in Organic Synthesis, Wiley 1999 3 , Chapter 6 Protection for the Thiol Group.
  • the coupling to the resin preferably takes place via the thiol group so that the thiol group remains masked during synthesis.
  • the cleaving from 25 the solid phase or the protective group is carried out in an acidic environment (for example 1% TFA in dichloromethane). It has been found that this way the side reactions of the thiol and the mercaptophilic compound described in EP 485 874 A2 can be prevented.
  • the diluting component comprises diluting molecules of the general formula
  • the diluting component as well includes a preferred moiety R comprising the subunits R a and R b and, in a preferred embodiment, corresponding to the structural formula given above.
  • the diluting molecules must not influence the interaction between the immobilized interaction partner and the free interaction partner. In particular, there should be no specific or unspecific binding between the free interaction partner and the diluting molecule.
  • the anchor molecule and the diluting molecule should be as structurally similar as possible in order to ensure that their chemical behavior on the solid phase surface is similar as well (homogeneous miscibility on the solid phase surface). It is advantageous if the total chain length of the diluting molecule is shorter than that of the anchor molecule, for example by one structural unit, such as an ethylene glycol unit. This can for example be achieved by leaving out the last component in the diluant structure when forming the anchor and the diluant from commercially available synthesis components.
  • the head group of the diluting molecule should be different from that of the anchor molecule.
  • Preferred head groups for the diluting molecule include methoxy and acylamide groups. Acetamide is especially preferred, in particular if maleimidyl is used as the anchor head group.
  • a (diluted) measuring surface according to the present invention is generated by a chemical reaction of the interaction partner to be immobilized and the mercaptophilic head group M of the anchor.
  • the interaction partner to be immobilized carries a thiol group which reacts in as selective and quantitative a manner as possible while forming a covalent bond with the head group of the anchor (cf. C. Boeckler et al., J. Immun. Meth. (1996) 191, 1-10). If no suitable functionality is present, it can be generated by converting available functional groups in the interaction partner to be immobilized, e.g. by reductively cleaving a disulfide bond to form a thiol (P. Parham, J. Immunol.
  • the additional molecule can furthermore function as a spacer between the head group of the anchor and the interaction partner to be immobilized, wherein the latter is kept away from the head group and thus the electronic and steric properties of the head group do not affect the binding properties of the interaction partner.
  • Interaction partners to be immobilized/immobilized interaction partners or ligands in the present invention are chemically unmodified or modified, i.e. provided with a thiol functionality, interaction partners or ligands.
  • immobilized interaction partners generally refers to structural elements which due to their structural characteristics can enter into specific interaction with test substances or subunits thereof. With the help of such immobilized structures, targets can be bound. e.g. in screening processes, which exhibit corresponding compatible structural units. Knowledge of the structure of the ligand then inter alia allows conclusions as to the possible structure or specific structural elements of the receptor.
  • ligand is not used consistently in the prior art literature. It should therefore be emphasized that in the present invention the term “ligand” is used in particular for so-called “small molecules” which are preferably bound covalently to the binding surface.
  • ligands include oligomers or small organic molecules, such as peptides, oligonucleotides, carbohydrates (glycosides), isoprenoids or lipid structures.
  • the molecular weight is usually the basis for the definition of such small molecules.
  • the mercaptophilic head group M should preferably serve to immobilize such a ligand, for example a so-called “small molecule” or “low molecular weight molecule”.
  • WO 89/03041 and WO 89/03042 describe molecules with molecular masses of up to 7,000 g/mol as small molecules. Usually, however, molecular masses between 50 and 3,000 g/mol, more frequently between 75 and 2,000 g/mol and most of the time in the range of 100 to 1000 g/mol are mentioned.
  • the molecular weight of a lioand/small molecule should be between 50 and 3,000 g/mol, preferably between 75 and 1,500 g/mol.
  • ligands can optionally specifically bind to the receptors. They can be synthesized using reactions known in the prior art (cf. e.g. J. S. Complementary aromatic compound, a terpene, an organophosphorous compound, a chelate complex, a neurotransmitter, a substituted amine, an alcohol, an ester, an ether or a carboxylic acid and its derivatives and combinations thereof. They can be synthesized using reactions known in the prior art (cf. e.g. J. S. Computel, G. Jung, Angew. Chem. Int. Ed. 35, (1996) 17-42).
  • ligands may have to be chemically modified such that they are able to react with the mercaptophilic head group of the anchors according to the present invention.
  • a list of exemplary compounds that can be bound as ligands, optionally after modification of a functional group or addition of an additional molecule, to the binding surfaces of the present invention can be found in WO 00/73796 A2, which was filed on May 26, 2000 with the German Patent & Trademark Office and whose corresponding disclosure content is hereby incorporated by reference.
  • the terms “immobilized interaction partner” or “interaction partner to be immobilized” also encompass macromolecules, preferably biomolecules or receptors such as proteins, oligo- or polypeptides, DNA, RNA, oligo- or polynucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- and polysaccarides, as well as their modifications, and synthetic molecules such as e.g. fusion proteins and synthesized primers.
  • These macromolecules as well may have to be chemically modified for use in the present invention in order to guarantee their covalent binding to the mercaptophilic head group of the anchor molecules.
  • Molecules acting as free interaction partners are preferably molecules occurring in biological systems or molecules interacting with such molecules, in particular receptors such as proteins, DNA, RNA, oligonucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- and polysaccarides, as well as synthetic molecules such as e.g. fusion proteins and synthesized primers.
  • receptors such as proteins, DNA, RNA, oligonucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- and polysaccarides, as well as synthetic molecules such as e.g. fusion proteins and synthesized primers.
  • a solution containing the interaction partner to be immobilized is applied onto the binding surface. It is an advantage of the present invention that the concentration of the ligand (if one is used) on the surface is only determined by the dilution of the binding surface and not by the concentration of the ligand in the solution to be applied. This is in particular advantageous if many interaction partners to be immobilized of which only an approximate concentration is known are processed simultaneously, which is often the case with ligands obtained from combinatorial synthesis. This increases the reproducibility and comparability of different test measurements.
  • the application of the interaction partner to be immobilized is not restricted to specific processes; however, conventional pipetting or spotting devices, but also stamping or ink-jet processes can be employed for a more precise localization of the active areas on the binding surface.
  • Another aspect of the present invention relates to the provision of an array comprising a plurality of fields on a planar solid phase carrier.
  • Each individual field can be used as a separate measuring surface.
  • these measuring surfaces differ in the type of interaction partner immobilized on each one, whereby a single measuring surface can both represent a single type of interaction partner and a mixture of different interaction partners.
  • a variety of interaction partners to be immobilized are applied onto the binding surface and subjected to a measuring process.
  • the spatial structure of the resulting array can be predetermined by mechanical structuring the carrier.
  • structured carrier plates When structured carrier plates are used as sensor surfaces in the present invention, they preferably comprise a plurality of evenly positioned fields with addressable positions for the generation of binding surfaces wherein these fields are located in low-depth cavities. This provides a barrier for the liquid while at the same time the surface is kept as small as possible in order to minimize possible unspecific adsorption phenomena.
  • these fields comprise a layer of the material allowing the binding of the thiol-functionalized anchors.
  • the cavities Preferably, the cavities have a depth of 20 to 100 ⁇ m and the anchors are immobilized at their bottom which is formed for example by a metal or metal oxide, preferably a noble metal such as gold.
  • FIGS. 7 to 9 each show a schematic detail of a cross-section of a preferred carrier plate.
  • a copper-clad base material ( 4 ) can be used which preferably already carries a metal layer ( 3 ) such as copper and which is provided with said carrier layer ( 2 ) by means of a galvanic deposition process.
  • the thickness of the carrier layer is only a few micrometers, the exact thickness necessary for generating a closed layer.
  • an UV-exposable protective layer ( 1 ) For this purpose, either photoresists common in semiconductor production or other protective paints that are UV-exposable and can therefore be structured can be used.
  • the paint layers preferably have a thickness of 20 ⁇ m to 100 ⁇ m.
  • an image of a mask is projected onto the protective layer.
  • the mask preferably exhibits round or rectangular/square patterns.
  • defined openings are formed in the protective layer which expose the carrier layer underneath.
  • the protective layer that can be photostructured also forms the walls of the cavities ( 6 ) and thus the form of the cavity ( 6 ) and its opening.
  • FIG. 8 shows a carrier plate ( 5 ) with deeper cavities ( 6 ) wherein, however, the portion of unprotected wall surface is not increased.
  • This carrier plate preferably has a basis material ( 4 ) on whose surface a metallic coating ( 3 ) is provided, which in turn is provided with a protective layer ( 1 ).
  • At least one cavity ( 6 ) is provided in the protective layer ( 1 ) and the metal layer ( 3 ), which in the area of the metal layer ( 3 ) well-shaped and is provided with a carrier layer ( 2 ) and which in the area of the protective layer ( 1 ) tapers towards the depression, wherein the lower rim of the cavity portion provided in the protective layer ( 1 ) has a smaller radius than the upper rim of the cavity portion formed in the metal layer ( 3 ).
  • the production of a carrier plate as shown in FIG. 9 also starts from a coated plate.
  • the thickness of the layer already present ( 3 ) is preferably 100 ⁇ m to 150 ⁇ m.
  • the layer ( 3 ) is structured such that it already exhibits wells.
  • the plate is coated with the carrier layer ( 2 ) by means of a galvanic process.
  • a protective layer ( 1 ) is structured such that a structure is formed in the protective layer ( 1 ) above the cavities ( 6 ) etched into the layer ( 3 ).
  • the depth of the thus formed cavity ( 6 ) is the sum of the depth of the etched structure and the thickness of the protective layer ( 1 ).
  • the cavities are arranged such that a regular, preferably Cartesian, grid of columns and lines is formed on the carrier plate.
  • the size and shape of the carrier plate can be selected as desired and can easily be adapted to the detection system used. If spotting robots are used for immobilizing the anchors or interaction partners, or if the binding surface is present on microtitration plates, the distance between the fields should preferably be adjusted to the microtitration format or the spotting device used.
  • the number of fields can also exceed the number of subunits of the microtitration plate, i.e. multiple fields can be used per surface.
  • a square carrier plate of about 12 ⁇ 12 cm may comprise a total of 9216 fields which can be covered by means of a pipetting robot from six conventional 1536 microtitration plates.
  • a spatially structured presentation of identical or different immobilized to interaction partners can also be achieved by covalently binding the interaction partner to a predetermined portion of the binding surface after an amount of liquid has selectively been applied, without it being necessary to physically divide the surface into individual compartments.
  • techniques for applying reagent spots onto metal or metal oxide surfaces as described in EP-A-0 872 735 can analogously be applied to the binding surfaces of the present invention. If solutions of the interaction partners/ligands are applied onto a homogeneous binding surface, the concentration of the ligand/interaction partner on the finished measuring surface is determined by the dilution of the binding surface. Consequently, there is the additional advantage compared to EP 872 735 A1 that precisely defined amounts of liquid only have to be applied to the solid phase carrier surface once to generate a measuring array (namely when the binding layer is generated).
  • the present invention can be employed in HTS, in the research of active substances or in medical diagnostics.
  • Suitable measuring methods for detecting interaction between an immobilized (surface-bound) interaction partner/ligand and a free interaction partner/receptor wherein the solid phase carrier only serves for the immobilization of one interaction partner are based on the verification of the specific binding reaction by means of electrochemical (electro immunoassays), radiochemical (e.g. radioimmunoassay), mass-sensitive or optical processes such as fluorescence or luminescence measurements, in particular enzyme assays.
  • electrochemical electrochemical
  • radiochemical e.g. radioimmunoassay
  • mass-sensitive or optical processes such as fluorescence or luminescence measurements
  • enzyme assays in particular enzyme assays.
  • the so-called ELISA technique enzyme-linked immunosorbent assay, immunoassays using solid-phase technique
  • Reflecto-optical processes such as surface plasmone resonance are suitable for a marker-free detection of the interactions.
  • the solid phase carrier is part of the sensor system.
  • the surfaces of the present invention can also be used in classic processes such as affinity chromatography.
  • the resin was washed once with N,N-dimethylformamide (DMF), once with 5% water in DMF, four times with DMF, three times with dichloromethane and twice with hexane and dried under vacuum.
  • N,N-dimethylformamide DMF
  • the loading of the resin with N-(N 5 -Fmoc-5-aminopentyl)-11-mercaptoundecaneamide was determined by means of Fmoc analysis (G. B. Fields, R. L. Noble, Int. J. Peptide Protein Res. 1990, 35, 161-214) to be 0.35 mmol/g (yield: 60% of the theoretical value).
  • Fmoc-8-amino-3,6-dioxa-octanoic acid was coupled to 500 mg resin (0.175 mmol) from b) and then the Fmoc protective group was cleaved off as described in b). Subsequently, the free amino groups were acetylated by incubating the resin for 30 min with 10 ml 1/1/2 (v/v/v) acetic acid anhydride/pyridine/DMF. Then the resin was washed five times with DMF and three times with dichloromethane.
  • the binding surface consists of the anchor component from Example 2 and the diluting component from Example 1.
  • stock solutions of the individual components are prepared by dissolving the solids in ethylene glycol/0.1% TFA with the desired molar concentration which is verified by means of Ellman's test (G. L. Ellman, Arch. Biochem. Biophys. 82 (1959), 70-77) or based on the extinction coefficient at 295 nm. Then the stock solutions of anchor and diluting components are mixed in the desired ratio (1:10, v/v) and the gold surface is incubated with this 100 ⁇ M to 1 mM solution for 1 hour at room temperature. It is then washed with methanol/0.1% TFA and several times with water/acetic acid (2 ppm) and dried in a nitrogen stream.
  • the preparation of the protein surface is carried out by incubating the binding surface from Example 3 (1:10 dilution) with a 17 ⁇ M solution of caspase 7T7 (modified by means of a T7 and an oligo-His tag) in 0.2 M NaH 2 PO 4 /Na 2 HPO 4 , pH 7, (20 min, room temperature). Then the chip is washed with acetic acid/water (1 ⁇ l/500 ml), treated for 5 min with 10 mM 2-mercaptoethanol in 0.2 M NaH 2 PO 4 /Na 2 HPO 4 , pH 7, washed with acetic acid/water (see above) and dried in a nitrogen stream.
  • the goal of this measuring method is the vibrational spectroscopy of a thin layer located on a metal surface.
  • the IR light at a grazing incidence is reflected off the metal surface.
  • Measurements are only carried out with IR light polarized in the incidence plane since light with a perpendicular polarization direction cannot contribute to the measurement.
  • transition dipole moments which comprise portions at least parallel to the E-field of the radiation contribute to the absorption.
  • the upper plot in FIG. 2 shows the FTIR spectrum of the pure diluting component (A).
  • the CH 2 bands can be seen in the area of 2.900 cm ⁇ 1 , the CONH bands in the area of 1.500 to 1,700 cm ⁇ 1 and the C—O—C band at 1.122 cm ⁇ 1 .
  • the bottom-most plot shows the spectrum of the pure anchor component with ligand (B). Compared to the spectrum of the diluant, the band at 1.716 cm ⁇ 1 stands out. This band can be assigned to the imide group of the anchor component. When surfaces are prepared with an increasing anchor component concentration (spectra from top to bottom), a systematic increase in the imide band can be observed.
  • the area of the peak at 1.716 cm ⁇ 1 was determined and plotted against the concentration of the anchor component (cf FIG. 3).
  • a linear fit to the measuring data clearly shows the connection and thus the conformity of the anchor molecule concentration on the gold surface and the concentration in the solution during gold coating.
  • XPS X-ray photoelectron spectroscopy
  • the layer thicknesses obtained from XPS measurements and ellipsometric measurements correspond to the length of the diluting molecule from Example 1.
  • the molecular length of the diluting component is 44 ⁇ . Since the molecules do not protrude at a right angle from the surface but are usually bound at a specific angle, the somewhat shorter measured layer thickness (34 ⁇ ) indicates that these surfaces are SAiMs. A bond angle of the molecules of about 50° with respect to the surface is therefore conceivable.
  • the protein adsorption resistance was determined by means of surface plasmone resonance on Biacore® 3000 with the following proteins: ribonuclease A (bovine) 0.1 mg/ml lysozyme (chicken) 0.1 mg/ml egg albumin (chicken) 0.1 mg/ml D-amino-acid oxidase (procine) 0.1 mg/ml pyruvate kinase (rabbit) 0.1 mg/ml fibriogen (human) 1 mg/ml glutathione reductase (wheat germ) 1 mg/ml
  • the values shown in FIG. 4 are average values from 4 measurements.
  • a short-chain disulfide (compound A of EP 698 787), the diluting molecule from Example 1 and 12,15,18-trioxa-20-hydroxyicosan-1-thiol (Whitesides et al. J.Am.Chem.Soc. 1995, 117, 12009-10) were used as a diluants.
  • the chain length of the molecule influences the unspecific adsorption resistance, which increasingly occurs in short-chain diluting molecules.
  • Example 4 Analogously to Example 4, pY with cysteine as additional molecule is used to bind pY to the binding surface from Example 3 (“pY tag”).
  • a gold chip was incubated with a 1:10 mixture of the anchor component from Example 2 and the diluting component from Example 1 in ethylene glycol and 1% TFA at a total concentration of 0.5 mM. Then the chip was washed several times in methanol/0.1% TFA and then in water pH 7.0. Chips pretreated in this manner were dried in a nitrogen stream.
  • Such chips were then incubated overnight in BSA blocking solution (50 mM Tris/HCl, 150 mM NaCl, 5 g/l BSA, 0.05% (v/v) Tween-20®, pH 7.3 ).
  • BSA blocking solution 50 mM Tris/HCl, 150 mM NaCl, 5 g/l BSA, 0.05% (v/v) Tween-20®, pH 7.3 ).
  • the verification of the pY groups on the surface of the chip was carried out by means of an immunoassay wherein the chip was first incubated with a 1:5,000 a-pY antibody in blocking solution and then with 1:5,000 anti-mouse-Fab-POD. Between the incubation steps, the blocking solution was washed (2 ⁇ 1 min). Prior to the detection of the luminescence signal with the Super-Signal-Plus Substrate from Roche Diagnostics, it was again washed in TBS buffer. The luminescence reaction was observed in a Lumi-Imager (Roche
  • a 12 ⁇ 12 cm gold chip (40 nm gold/1 nm chromium on glass) was incubated with a 1:10 mixture (dilution) of anchor component from Example 2 and diluting component from Example 1 in ethylene glycol and 1% trifluoroacetic acid (TFA) at a total concentration of 1.0 mM. Then the chip was washed several times in methanol/1% TFA and then in water pH 7.0. Chips pretreated in this manner were dried in a nitrogen stream.
  • TFA trifluoroacetic acid
  • a library of 9216 thiol-containing ligands were deposited on these chips by means of a spotting device in an arrangement of 96 ⁇ 96 spots with a distance of 1.125 mm between the spots.
  • the thiol-containing ligands applied onto the surface are dissolved in a 40 ⁇ M solution of 0.2 M Pi, 5 mM EDTA and 10% (v/v) ethylene glycol pH 7.0.
  • the spotting device releases about 10 nl per spot so that in each spot a high excess of the thiol-containing ligand compared to the surface-bound maleimide group is guaranteed and a complete reaction of the maleimide groups can be achieved.
  • the maleimide groups were then saturated by incubating the chip in 0.2 M phosphate buffer pH 7.0, 10 mM mercaptoethanol for 30 min.
  • Such chips were then incubated overnight in BSA blocking solution (50 mM Tris/HCl, 150 mM NaCl, 5 g/1 BSA, 0.05% (v/v) Tween-20®, pH 7.3).
  • BSA blocking solution 50 mM Tris/HCl, 150 mM NaCl, 5 g/1 BSA, 0.05% (v/v) Tween-20®, pH 7.3.
  • the analysis of potential binding partners of the target protein thrombin was carried out by means of an immunoassay.
  • the chip was incubated for 4 hours in 10 nM thrombin in blocking solution. After washing twice for two minutes in blocking solution, a 1:1,000 dilution of a polyclonal anti-thrombin antibody was incubated with the chip for 2 hours. After again washing twice in blocking solution, an anti-rabbit antibody POD conjugate was incubated with the chip for 2 hours in order to determine the binding activity. Finally, the chip was washed twice for 2 min in TBST. The chemical luminescence reaction was verified by reacting the Lumi-Light Plus Substrate in the Lumi Imager (Roche Diagnostics, Germany).
  • FIG. 6 shows a photograph of the chemical luminescence reaction (10 nM thrombin). Black spots (represented in the Imager as light spots) indicate binding of the thrombin. Discrete intensities can be seen as spots in certain positions. Since every compound on the array has certain spatial coordinates, specific chemical structures can be assigned to the spots.
  • the products were dissolved in ethyl acetate and were each washed three times with 10% sodium carbonate solution, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were dried over sodium sulfate and concentrated in a rotary evaporator until dry.
  • the mixtures were stirred for 3 hours at room temperature, then the urea was filtered off and the solvent was concentrated until dry.
  • the products (4), (5) and (6) were dissolved in ethyl acetate and were each washed three times with 0.1 M HCl, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were then dried over sodium sulfate and concentrated until dry.
  • the products (7) and (8) were dissolved in ethyl acetate (EE) and were each washed three times with 10% sodium carbonate solution, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were dried over sodium sulfate and concentrated until dry.
  • EE ethyl acetate

Abstract

The present invention relates to binding surfaces for the immobilization of ligands, ligand surfaces and structured surface arrays which present a plurality of identical or different ligands. The invention further relates to a process for the production and the use of such surfaces and to specific binder molecules which can be used for the preparation thereof.

Description

  • The present invention relates to binding surfaces for the immobilization of ligands, ligand surfaces and structured surface arrays which present a plurality of identical or different ligands. The invention further relates to a process for the production and the use of such surfaces and to specific binder molecules which can be used for the preparation thereof. [0001]
  • In known processes of interaction analysis, biomolecular interaction is studied with receptor-ligand systems wherein the receptor is usually a biomacromolecule (e.g. a protein or single-stranded DNA) and the ligand is a “probe”, a molecule usually of low molecular weight and of biological or synthetic origin (peptides, oligonucleotides or what are referred to as small organic molecules). Such ligands exhibit highly specific structural properties which can interact with a receptor provided it exhibits corresponding structures. The binding to the receptor can take place via one or several ligands. Furthermore, systems are studied wherein both interaction partners are macromolecules (e.g. protein-protein interaction). [0002]
  • Interaction analysis is used in the pharmaceutical and agrochemical industries for researching active substances. The goal is to analyze as large a number of different samples as possible in as short a time period as possible. There is a growing interest to develop testing systems that are able to simultaneously subject a large number of both identical and different molecules to a certain test method for detecting their biospecific binding behavior in order to identify molecules with a specific effect (High-Throughput Screening, HTS). Such a parallelization is closely linked to a simultaneous miniaturization of the testing arrangement and automation of the testing procedure. Furthermore, interaction analysis is used in the research of the genome (polymorphisms (SNP) analysis or expression pattern analysis) and also in food analysis. [0003]
  • It constitutes a practical advantage to bind one of the interaction or binding partners, i.e. the ligand or the receptor, to an organic or inorganic surface, thereby imparting bioactivity should preferably be such that it allows at least the formation of a highly ordered monolayer on the basis of the anchor molecules. [0004]
  • The generation of a bioactive surface using such anchors can be carried out in one or several steps. In the single-step process, the interaction partner is bonded to the head group of the anchor prior to its immobilization. Suitable anchors for the single-step process are described in DE 199 24 606.8. For providing a measuring surface, a complete ligand-anchor conjugate (LAC) is applied onto the surface. This is a molecule connecting the interaction partner to be immobilized and the functional group necessary for binding to the surface by means of a structure capable of forming an SAM. [0005]
  • In multi-step processes, first a binding surface in the form of an organic boundary layer is generated which does not yet exhibit the desired specific structural features, however, which is suitable for binding or can be activated to bind the interaction partners to be immobilized, e.g. ligands. Thus, immobilization of an interaction partner takes place after the generation of a first boundary layer that is not bond-specific by means of a covalent, ionic or complex bond via the head group of the binding component. For this purpose, the head group can be used directly or after it has been activated. Examples of such head groups are described in EP 485 874 A2. [0006]
  • An important advantage of the multi-step process is the fact that the same surface can be used for every interaction partner to be immobilized. Therefore, the resulting concentration of the interaction partners to be immobilized is the same on the surface as well, which is not or not always a given in the single-step process. This is particularly important for the comparability and reproducibility of the different measuring results. However, it also has to be kept in mind that the chemical homogeneity of the non-biospecific binding surface decreases again as the number of potentially necessary activation steps increases. [0007]
  • A step-by-step formation of a diluted binding layer is described in WO 98/40739 A1, wherein in a first step cystamine is applied onto a gold surface and the provision of maleimide as head group is realized in a second chemical reaction. The disadvantage of this process is that another activation step is required for obtaining a binding surface. [0008]
  • Furthermore, reactive amino functions remain that can be responsible for an unspecific binding of macromolecules such as proteins. [0009]
  • It has to be made sure in all multi-step processes that the reaction binding the interaction partner to be immobilized to the anchor via the head group proceeds selectively and in a controlled manner. In this connection, covalent binding of the interaction partner to be immobilized is advantageous since the bioactive surface generated in this manner is chemically more stable, for example with respect to regeneration or storage conditions that can be applied. These advantages particular apply in comparison with the non-covalent binding system streptavidin/biotin which is used frequently. [0010]
  • In particular in the case of immobilization of a small ligand, for instance a so-called “small molecule”, the selectivity and the quantitative course of the reaction is of utmost importance for the quantitative evaluation of binding studies or interaction analyses. Here, in contrast to e.g. macromolecular recognition structures, slight differences in the binding strength of the ligand to the receptor often have to be resolved. A ligand of higher affinity offered at a lower density (concentration) than another ligand with a lower binding affinity can lead to an artificially reduced signal, while the other way round a weaker ligand offered at a higher density (concentration) can lead to an artificially increased signal compared to another ligand. [0011]
  • In order to ensure a high degree of selectivity and the possibility of quantitative reactions, reactions with molecules carrying a thiol group or modified by means of a thiol group are employed in biochemistry. The reaction partner is often a macromolecule carrying a maleimidyl group. The product of the reaction of the thiol and the maleimidyl group is a thio succinimide (Michael addition) (G. T. Hermanson, Bioconjugate Techniques, Academic Press 1996, 229-252; Suda et al. Biochem. Biophys. Res. Comm. (1999), 261, 276-282). [0012]
  • A number of reaction partners for the thiol group are known from the prior art (referred to in the following as mercaptophiles), which also allow its selective and quantitative reaction. In addition to maleimides, they include the following compounds: [0013]
  • Iodo- and bromoacetamides, pyridyldithio compounds (G. T. Hermanson, Bioconjugate Techniques, Academic Press 1996, 229-252; C. Boeckler et al., J. Immun.Meth. (1996) 191, 1-10), Michael acceptors in general, acrylic acid derivatives such as the esters, amides, lactones or lactames thereof (K. Matsuura et al. J. Mass Spectrom. (1998) 33, 1199-1208; W. Adam et al. J. Org. Chem. (1995) 60, 578-584), methylene-gem-difluorocyclopropanes (T. Taguchi et al., Tetrahedron (1997). 53, 9497-9508), α,β-unsaturated aldehydes and ketones (Chen et al., J. Am. Chem. Soc. (1994) 116, 2661-2662) and α,β-unsaturated sulfones and sulfonamides. [0014]
  • The thiol/mercaptophile system offers the essential advantage that the binding of the interaction partner to be immobilized can be carried out under gentle reaction conditions (ambient temperature, neutral pH, buffer solutions can be used). This is of particular importance when unstable compounds or proteins that denature easily are used. Another advantage is that compared to carboxylic acid, amine or amide groups, a thiol functionality hardly ever occurs in active substances. Thus, an undesired reaction of mercaptophiles that may remain after the immobilization of one interaction partner and the target can largely be avoided. [0015]
  • The thiol/maleimide system is furthermore particularly known for its high degree of selectivity compared to other functionalities such as hydroxyl, amine, carboxyl or hydroxylamine groups. In addition, the formation of the covalent bond is characterized by a high reaction rate (cf. Schelté et al., Bioconj. Chem. (2000), 11, 118-123). [0016]
  • Although such reactions have proven successful in bioconjugate chemistry, anchors comprising both a thiol group for binding to the solid phase surface and a mercaptophilic head group for immobilizing an interaction partner are not disclosed in the prior art since both during synthesis and during the generation of the binding surface both intra- and intermolecular side reactions, and even polymerization, may occur (with respect to the purposeful polymerization of oligothiols with bis-maleimides, cf. L. R. Dix et al., Eur. Polym. J. 31 (1995), 653-658). [0017]
  • EP 485 874 A2 refers to this problem, which is avoided by the exclusive use of disulfide and sulfide groups in the anchor, in order to use for example maleimide as head group for the immobilization of proteins (reaction with —SH group of a Fab′ fragment). EP 698 787 A1 as well uses short-chain, maleimide-carrying disulfide anchors for the immobilization of proteins. However, anchors based on disulfide and sulfide groups show the disadvantage that an undesired spatial proximity of the head groups is generated. In this case, the head groups can interfere with each other in the immobilization reaction. [0018]
  • In order to be able to control the spatial proximity of adjacent ligands, EP 485 874 A2 and EP 698 787 A1 do not only apply anchor molecules onto the solid phase surface. Rather, the anchor molecules are “diluted”. This is due to the fact that if one partner, for example a ligand is bound to a solid phase carrier for interaction analysis, adjacent ligands on the surface can interfere with each other or with the interaction between the adjacent ligand and the interaction partner to be detected. Whitesides and his colleagues were able to verify this effect in the biospecific adsorption of carbonic anhydrase on benzosulfonamide groups, which were immobilized by means of alkane thiolates on gold, by demonstrating that the degree of undesired irreversible binding of carbonic anhydrase to a mixture of ligand-carrying anchor (immobilized sulfonamide) and diluting component (12,15,.18-trioxa-20-hydroxyicosan-1-thiol) decreases as the amount of diluting component in the mixture increases (J. Am. Chem. Soc. 1995, 117, 12009-10). [0019]
  • In order to avoid this disadvantage, mixed surfaces—as shown in FIG. 1—can be applied, which are composed of ligand-carrying anchor molecules and so-called diluting molecules that do not carry any ligands and thus dilute the measuring surface. [0020]
  • The structural nature of the diluting component has to meet the prerequisite that it will not influence the interaction of the immobilized interaction partner and the free interaction partner. In particular, if possible, no specific or unspecific binding between the free interaction partner and the diluting component should occur (e.g. diluant with as high a resistance to protein adsorption as possible). Furthermore, the anchor molecule and the diluting component should be as structurally similar as possible to ensure that their mixing behavior on the solid phase surface is as homogeneous as possible. [0021]
  • On principle, prior art anchors that are based on disulfides and sulfides and thus present two head groups cannot guarantee a stochastic distribution of head-group-carrying molecules and diluting molecules. However, this is necessary in order to ensure a purposeful interaction of the bioactive surface with the free interaction partner later on. [0022]
  • An immobilization of small ligands (in addition to proteins) in a multi-step process is described by Lahiri et al. in Anal. Chem. 1999, 71, 777-90. 12,15,18-Trioxa-20-hydroxyicosan-1-thiol is used as diluting component. For immobilizing the ligand, the anchor molecule carries a carboxylic acid function as a head group, which in a first step is activated with NHS and then reacted with an amino function in the protein or ligand that was either present or had been introduced. However, this reaction is not complete, and its to yield depends on the degree of dilution on the surface. [0023]
  • Thus, it is the object of the present invention to provide binding surfaces presenting reactive head groups which are capable of entering into selective and highly quantitative reactions with interaction partners to be immobilized. Furthermore, the present invention relates to novel anchor molecules comprising these head groups which can be used, together with diluting molecules, to provide such binding surfaces. The invention further relates to sensor or measuring surfaces obtainable by immobilizing specific interaction partners on the binding surfaces of the present invention, as well as to surface arrays comprising a plurality of such identical or different measuring surfaces. [0024]
  • A binding surface according to the present invention comprises a solid phase carrier on which an organic monolayer can assemble spontaneously, preferably comprising at least two components, a binding component and a diluting component (anchor and diluant). [0025]
  • The solid phase carrier consists of a substrate formed by a metal, preferably a noble metal, especially preferred gold, or carries a layer of such a metal. Optionally, the metal layer can be applied with the help of an intermediate layer which serves as a primer. The material used for the substrate depends on measuring method employed. If optical reflection processes such as surface plasmone resonance (SPR) are used, the substrate preferably consists of glass or a plastic material. Due to the use of sulfur-containing compounds for the immobilization of ligands, a gold layer and a chromium layer as a primer are preferably applied onto such substrates. [0026]
  • The spatial design of the solid phase carrier to be used in accordance with the present invention is not restricted, although planar, two-dimensional structures are preferably used for sensor applications. However, depending on the field of application, three-dimensional shapes such as spheres or hollow bodies may be used as well. [0027]
  • Processes for the Generation of the binding layers according to the present invention start from a solution of the anchor molecules which is brought into contact with the solid phase carrier. Preferred diluted measuring surfaces are obtained when a solution is used that contains a mixture of anchor molecules and diluting components in a certain molar ratio. Preferably, the solution has a molar ratio of anchor/diluant between 1:2 and 1:10,000. Especially preferred dilution ratios range from 1:10 to 1:100. The total concentration of anchor and diluting components preferably lies in the range of from 0.001 to 100 mmol/l, especially preferred about 0.1 to 1 mmol/l. For generating a homogeneous, large-area binding surface, the entire surface of the solid phase carrier is coated, for example by immersing in a tub, with the (diluted) binding layer. For applying the solution in a spatially defined manner, commercially available pipetting or spotting devices, as well as micropipetting devices or ink-jet processes can be used. Preferably, however, a spatial structure of the measuring surface is achieved by the selective application of the interaction partners to be immobilized onto the large-area binding surface. Suitable solvents include aqueous (e.g. buffer solutions) or organic solvents (e.g. methanol, ethanol, acetonitrile. N,N-dimethylformamide, ethylene glycol) or mixtures thereof. [0028]
  • During the application of the anchors onto the carrier surface, the problems of intramolecular reactions between the anchor molecules described in the prior art can be avoided by working in an acidic medium. For obtaining an acidic environment, the anchor molecule solutions preferably comprise an acid such as trifluoroacetic acid, hydrochloric acid or phosphoric acid. [0029]
  • Anchor molecules suitable for preparing the binding surface according to the present invention comprise at least two functional units present at opposite ends of the anchor. As a group allowing the bonding of the anchor with the surface of a carrier, these anchors comprise a thiol group. For binding the interaction partner to be immobilized to the anchor, the anchor comprises a mercaptophilic head group M as a second functional unit. [0030]
  • The two functionalities are connected via structural element R, which provides for the formation of a self-assembling monolayer on the surface of the carrier. Consequently, the anchors according to the present invention have the following general structure: [0031]
  • HS—R-M
  • Suitable structural elements R that both promote the formation of a monolayer and optionally also allow the adjustment of suitable distances between the head groups and the carrier surface with the help of spacer groups are described with respect to the anchor molecules in the German patent application DE 199 24 606.8, whose entire corresponding disclosure content is hereby incorporated by reference. [0032]
  • Accordingly, the moiety R is a branched or straight-chain, optionally substituted, saturated or unsaturated hydrocarbon chain which may be interrupted by heteroatoms, aromatic and heterocyclic compounds and comprises 5 to 2,000 atoms, including heteroatoms. Preferably, the anchor should comprise structures that impede or prevent a passive adsorption of the free interaction partner both on the anchor structure and the measuring surface. Furthermore, it is advantageous if the anchor comprises a spacer group which allows adjustment of the total chain length and of the flexibility of the LAC. These objects are achieved in a preferred embodiment of the anchor according to the present invention wherein R comprises at least two structural subunits R[0033] a and Rb. However, even if only one of these subunits is present, preferably Ra, anchors can be provided that can be used in the present invention to a satisfactory degree.
  • Preferably, R[0034] a causes the formation of an SAM and for this purpose, it should essentially be hydrophobic. It comprises a branched or straight hydrocarbon chain with 5 to 50 carbon atoms, which may be saturated or partially unsaturated and which may be interrupted by aromatic or heterocyclic groups or heteroatoms. A fully saturated hydrocarbon chain without heteroatoms is preferred. In a preferred embodiment, it has the general formula —(CH2)n 13 , wherein n is an integer of 5 to 50, preferably 5 to 25, especially preferred 5 to 18 and most preferred 8 to 12.
  • Commercially available compounds, in particular functionalized alkanes carrying functional units such as hydroxyl groups, halogen atoms, carboxylic acid groups or mercapto groups at their end groups are suitable for the introduction of R[0035] a in an especially preferred embodiment. These terminal functional units facilitate for example the connection to the adjacent structural units during the synthesis of the anchor. Optionally, they can be used to help introduce necessary components of the anchor, in particular —SH. Examples include 11-mercaptoundecanoic acid and its derivatives.
  • R[0036] b is preferably a spacer that allows adjustment of the total chain length and of the flexibility of the ligand-anchor conjugate. Preferably, Rb is a hydrocarbon chain which is interrupted by heteroatoms and therefore hydrophilic and impedes a passive adsorption of the receptor. The chain comprises 2 to 1,000 atoms, including heteroatoms; chain lengths of 5 to 500 are preferred, and especially preferred are chain lengths between 10 and 100 atoms.
  • In a preferred embodiment, R[0037] b is an oligoether of the general formula —(OAlk)y— wherein y is a natural number and Alk represents an alkylene group. A structure wherein y lies between 1 and 100, preferably between 1 and 20 and most preferred between 2 and 10 is preferred. The Alk group preferably comprises 1 to 20, more preferably 2 to 10 and particularly preferred 2 to 5 carbon atoms. Especially preferred is —(OC2H4)y—.
  • In a second preferred embodiment, R[0038] b is an oligoamide consisting of dicarboxylic acids and diamines and/or aminocarboxylic acids, wherein the amines independently preferably comprise 1 to 20, more preferably 1 to 10 carbon atoms, and may also be interrupted by further heteroatoms, in particular oxygen atoms. The carboxylic acid monomers independently preferably comprise 1 to 20, more preferably 1 to 10 carbon atoms and may also be interrupted by further heteroatoms, in particular oxygen atoms.
  • Suitable compounds for the introduction of R[0039] b in an especially preferred embodiment include commercially available compounds such as in particular glycol ethers such as e.g. triethylene glycol, triethylene glycol monomethylether, tetraethylene glycol, αω-diamines such as ethylene-, propylene-, butylene-, or pentylenediamine or 1,8-diamino-3,6-dioxaoctane, but also dicarboxylic acids such as succinic acid, 1,13-diamino-4,7,10-triooxatridecane, 3,6,9-trioxaundecanedioic acid, 8-amino-3,6-dioxaoctanoic acid or 4-aminobenzoic acid as well as their derivatives or combinations of identical components (such as e.g. in the case of 8-amino-3,6-dioxaoctanoic acid or 4-aminobenzoic acid) or combinations of different components (such as e.g. 1,13-diamino-4,7,10-triooxatridecane and 3,6,9-trioxaundecanedioic acid in an alternating sequence). An advantage of the use of 4-aminobenzoic acid is the fact that it can easily be detected spectroscopically, for example by means of a UV spectroscopy.
  • Especially preferred moieties R have the following general formula: [0040]
  • —(CH2)a-Q1-(CH2)b-{[Q2-(CH2)c—[O—(CH2)d]e—O—(CH2)f]g-[Q3-(CH2)c′—[O—(CH2)d′]e′—O—(CH2)f′]h}i-Q4-(CH2)j-Q5-(CH2)k
  • wherein the variables are defined independently of each other as follows and the numeric ranges comprise their respective limiting values as well as the integers contained therein: [0041]
  • Q[0042] 1,Q5 are —NH—C(O)—, —C(O)—NH— or a bond
  • Q[0043] 2,Q3,Q4 are —NH—C(O)— or —C(O)—NH—
  • a is 5 to 20, preferably 8 to 12, particularly preferred 10; [0044]
  • b is 0 to 5, preferably 0 if Q[0045] 1 is a bond and 1 to 10, preferably 2 to 7, particularly preferred 3 to 5 in all other cases;
  • c, c′ are 1 to 5, preferably 1 to 3, particularly preferred 1; [0046]
  • d, d′ are 1 to 5, preferably 1 to 3, particularly preferred 2; [0047]
  • e, e′ are 1 to 5, preferably 1 to 3, particularly preferred 2; [0048]
  • f, f′ are 1 to 5, preferably 1 to 3, particularly preferred 1; [0049]
  • g,h are 0 to 3, provided that g+h≧1, preferably g+h=2; [0050]
  • i is 1 to 3, preferably 1 to 2, particularly preferred 1; [0051]
  • j is 0 to 5, preferably 1 to 3, particularly preferred 2; and [0052]
  • k is 0 to 5. [0053]
  • Mercaptophilic head groups M are for example iodo- or bromoacetamide, pyridylthio compounds, Michael acceptors in general, acrylic acid derivatives such as its esters, amides, lactones or lactames, methylene-gem-difluorocyclopropanes, α,β-unsaturated aldehydes and ketones as well as α,β-unsaturated sulfones and sulfonamides. [0054]
  • Preferred head groups M are those of the general formula [0055]
    Figure US20040023366A1-20040205-C00001
  • wherein [0056]
  • R[0057] 1 and R2 are independently hydrogen or C1-C5 alkyl, preferably methyl, ethyl or n-propyl,
  • R[0058] 3 and R4 are independently hydrogen or C1-C5 alkyl, preferably methyl, ethyl or n-propyl, or R3 and R4 together form ═O and
  • the binding to the remaining anchor takes place via the nitrogen atom. [0059]
  • Preferably, R[0060] 3 and R4 together form ═O, and most preferred, the head group is a maleimidyl group.
  • Processes for the synthesis of an anchor molecule can be carried out in a solid phase or in solution. In the case of a preparation in solution, the thiol group is preferably provided with a protective group. Suitable protective groups are described in T. W. Greene, P. G. M. Wuts, Protective Groups in Organic Synthesis, Wiley 1999[0061] 3, Chapter 6 Protection for the Thiol Group. In a solid phase synthesis, the coupling to the resin preferably takes place via the thiol group so that the thiol group remains masked during synthesis. The cleaving from 25 the solid phase or the protective group is carried out in an acidic environment (for example 1% TFA in dichloromethane). It has been found that this way the side reactions of the thiol and the mercaptophilic compound described in EP 485 874 A2 can be prevented.
  • The diluting component comprises diluting molecules of the general formula [0062]
  • HS—R—X
  • wherein R is as defined above and X represents a non-mercaptophilic head group. Thus, the diluting component as well includes a preferred moiety R comprising the subunits R[0063] a and Rb and, in a preferred embodiment, corresponding to the structural formula given above.
  • Ideally, the diluting molecules must not influence the interaction between the immobilized interaction partner and the free interaction partner. In particular, there should be no specific or unspecific binding between the free interaction partner and the diluting molecule. Furthermore, the anchor molecule and the diluting molecule should be as structurally similar as possible in order to ensure that their chemical behavior on the solid phase surface is similar as well (homogeneous miscibility on the solid phase surface). It is advantageous if the total chain length of the diluting molecule is shorter than that of the anchor molecule, for example by one structural unit, such as an ethylene glycol unit. This can for example be achieved by leaving out the last component in the diluant structure when forming the anchor and the diluant from commercially available synthesis components. [0064]
  • In particular, the head group of the diluting molecule should be different from that of the anchor molecule. Preferred head groups for the diluting molecule include methoxy and acylamide groups. Acetamide is especially preferred, in particular if maleimidyl is used as the anchor head group. [0065]
  • A (diluted) measuring surface according to the present invention is generated by a chemical reaction of the interaction partner to be immobilized and the mercaptophilic head group M of the anchor. For this purpose, the interaction partner to be immobilized carries a thiol group which reacts in as selective and quantitative a manner as possible while forming a covalent bond with the head group of the anchor (cf. C. Boeckler et al., J. Immun. Meth. (1996) 191, 1-10). If no suitable functionality is present, it can be generated by converting available functional groups in the interaction partner to be immobilized, e.g. by reductively cleaving a disulfide bond to form a thiol (P. Parham, J. Immunol. 131 (1983), 2895-2902). Furthermore, there is the possibility of providing the interaction partner to be immobilized with an additional molecule, such as for example cysteine, by way of a chemical reaction, which carries the suitable functional group for binding to the head group of the anchor. (With respect to the introduction of a protected 2-aminoethyl thiol in oligonucleotides, see D. Gottschling et al., Bioconjugate Chem. 1998 (9) 831-837.) [0066]
  • The additional molecule can furthermore function as a spacer between the head group of the anchor and the interaction partner to be immobilized, wherein the latter is kept away from the head group and thus the electronic and steric properties of the head group do not affect the binding properties of the interaction partner. [0067]
  • Interaction partners to be immobilized/immobilized interaction partners or ligands in the present invention are chemically unmodified or modified, i.e. provided with a thiol functionality, interaction partners or ligands. [0068]
  • Furthermore, in the context of the present invention, the term immobilized interaction partners generally refers to structural elements which due to their structural characteristics can enter into specific interaction with test substances or subunits thereof. With the help of such immobilized structures, targets can be bound. e.g. in screening processes, which exhibit corresponding compatible structural units. Knowledge of the structure of the ligand then inter alia allows conclusions as to the possible structure or specific structural elements of the receptor. [0069]
  • The term “ligand” is not used consistently in the prior art literature. It should therefore be emphasized that in the present invention the term “ligand” is used in particular for so-called “small molecules” which are preferably bound covalently to the binding surface. [0070]
  • Examples of such ligands include oligomers or small organic molecules, such as peptides, oligonucleotides, carbohydrates (glycosides), isoprenoids or lipid structures. In the relevant literature, the molecular weight is usually the basis for the definition of such small molecules. [0071]
  • The mercaptophilic head group M should preferably serve to immobilize such a ligand, for example a so-called “small molecule” or “low molecular weight molecule”. [0072]
  • WO 89/03041 and WO 89/03042 describe molecules with molecular masses of up to 7,000 g/mol as small molecules. Usually, however, molecular masses between 50 and 3,000 g/mol, more frequently between 75 and 2,000 g/mol and most of the time in the range of 100 to 1000 g/mol are mentioned. Reference is made for example to the documents WO 86/02736, WO 97/31269, U.S. Pat No. 5,928,868,. U.S. Pat. No. 5,242,902, U.S. Pat. No. 5,468,651, U.S. Pat. No. 5,547,853, U.S. Pat. No. 5,616,562, U.S. Pat. No. 5,641,690, U.S. Pat. No. 4,956,303 and U.S. Pat. No. 5,928,643. [0073]
  • In the present invention, the molecular weight of a lioand/small molecule (without anchor and without additional molecule) should be between 50 and 3,000 g/mol, preferably between 75 and 1,500 g/mol. [0074]
  • Due to their characteristic spatial or electronic structures, caused for example by the functionality of an amino acid, a nucleoside, a heterocyclic compound, an alicyclic compound, an aromatic compound, a terpene, an organophosphorous compound, a chelate complex, a neurotransmitter, a substituted amine, an alcohol, an ester, an ether or a carboxylic acid and its derivatives and combinations thereof, ligands can optionally specifically bind to the receptors. They can be synthesized using reactions known in the prior art (cf. e.g. J. S. Früchtel, G. Jung, Angew. Chem. Int. Ed. 35, (1996) 17-42). These ligands may have to be chemically modified such that they are able to react with the mercaptophilic head group of the anchors according to the present invention. A list of exemplary compounds that can be bound as ligands, optionally after modification of a functional group or addition of an additional molecule, to the binding surfaces of the present invention can be found in WO 00/73796 A2, which was filed on May 26, 2000 with the German Patent & Trademark Office and whose corresponding disclosure content is hereby incorporated by reference. [0075]
  • However, in addition to ligands, the terms “immobilized interaction partner” or “interaction partner to be immobilized” also encompass macromolecules, preferably biomolecules or receptors such as proteins, oligo- or polypeptides, DNA, RNA, oligo- or polynucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- and polysaccarides, as well as their modifications, and synthetic molecules such as e.g. fusion proteins and synthesized primers. These macromolecules as well may have to be chemically modified for use in the present invention in order to guarantee their covalent binding to the mercaptophilic head group of the anchor molecules. [0076]
  • Molecules acting as free interaction partners are preferably molecules occurring in biological systems or molecules interacting with such molecules, in particular receptors such as proteins, DNA, RNA, oligonucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- and polysaccarides, as well as synthetic molecules such as e.g. fusion proteins and synthesized primers. [0077]
  • For preparing a measuring surface, a solution containing the interaction partner to be immobilized is applied onto the binding surface. It is an advantage of the present invention that the concentration of the ligand (if one is used) on the surface is only determined by the dilution of the binding surface and not by the concentration of the ligand in the solution to be applied. This is in particular advantageous if many interaction partners to be immobilized of which only an approximate concentration is known are processed simultaneously, which is often the case with ligands obtained from combinatorial synthesis. This increases the reproducibility and comparability of different test measurements. [0078]
  • Thus, starting from the binding surface according to the present invention. only one step is required for preparing the biospecific measuring surface, namely contact with the thiol-carrying ligand. For this purpose, the ligand should be present in a slight excess to guarantee a quantitative reaction. Another advantage of the present invention thus lies in the low amount of ligand required for the preparation of a bioactive surface, in particular compared with single-step processes. [0079]
  • The application of the interaction partner to be immobilized is not restricted to specific processes; however, conventional pipetting or spotting devices, but also stamping or ink-jet processes can be employed for a more precise localization of the active areas on the binding surface. [0080]
  • Another aspect of the present invention relates to the provision of an array comprising a plurality of fields on a planar solid phase carrier. Each individual field can be used as a separate measuring surface. Preferably, these measuring surfaces differ in the type of interaction partner immobilized on each one, whereby a single measuring surface can both represent a single type of interaction partner and a mixture of different interaction partners. For this purpose, a variety of interaction partners to be immobilized are applied onto the binding surface and subjected to a measuring process. [0081]
  • The spatial structure of the resulting array can be predetermined by mechanical structuring the carrier. When structured carrier plates are used as sensor surfaces in the present invention, they preferably comprise a plurality of evenly positioned fields with addressable positions for the generation of binding surfaces wherein these fields are located in low-depth cavities. This provides a barrier for the liquid while at the same time the surface is kept as small as possible in order to minimize possible unspecific adsorption phenomena. Furthermore, these fields comprise a layer of the material allowing the binding of the thiol-functionalized anchors. Preferably, the cavities have a depth of 20 to 100 μm and the anchors are immobilized at their bottom which is formed for example by a metal or metal oxide, preferably a noble metal such as gold. [0082]
  • With the help of such fields it is possible to prevent or minimize disadvantages with respect to unspecific binding or spilling over. Furthermore, the manufacture of such a carrier plate can be made inexpensive by the use of processes and materials employed in photo lithography and etching techniques applied in semiconductor technology.[0083]
  • In the following, preferred embodiments of such a carrier plate are described in more detail with reference to the Figures. FIGS. [0084] 7 to 9 each show a schematic detail of a cross-section of a preferred carrier plate.
  • For preparing a carrier plate ([0085] 5) according to FIG. 7, a copper-clad base material (4) can be used which preferably already carries a metal layer (3) such as copper and which is provided with said carrier layer (2) by means of a galvanic deposition process. The thickness of the carrier layer is only a few micrometers, the exact thickness necessary for generating a closed layer. After electroplating the plate is provided with an UV-exposable protective layer (1). For this purpose, either photoresists common in semiconductor production or other protective paints that are UV-exposable and can therefore be structured can be used. The paint layers preferably have a thickness of 20 μm to 100 μm. In one exposure step, an image of a mask is projected onto the protective layer. The mask preferably exhibits round or rectangular/square patterns. After a developing step, defined openings are formed in the protective layer which expose the carrier layer underneath. Thus, after structuring, the protective layer that can be photostructured also forms the walls of the cavities (6) and thus the form of the cavity (6) and its opening.
  • to If application and structuring of the protective layer ([0086] 1) is carried out prior to the application of the carrier layer (2), a carrier (5) as shown in FIG. 8 is obtained.
  • FIG. 8 shows a carrier plate ([0087] 5) with deeper cavities (6) wherein, however, the portion of unprotected wall surface is not increased. This carrier plate preferably has a basis material (4) on whose surface a metallic coating (3) is provided, which in turn is provided with a protective layer (1). At least one cavity (6) is provided in the protective layer (1) and the metal layer (3), which in the area of the metal layer (3) well-shaped and is provided with a carrier layer (2) and which in the area of the protective layer (1) tapers towards the depression, wherein the lower rim of the cavity portion provided in the protective layer (1) has a smaller radius than the upper rim of the cavity portion formed in the metal layer (3).
  • The production of a carrier plate as shown in FIG. 9 also starts from a coated plate. In that case, however, the thickness of the layer already present ([0088] 3) is preferably 100 μm to 150 μm. By means of a photoresist (not shown in the Figure), the layer (3) is structured such that it already exhibits wells. Then the plate is coated with the carrier layer (2) by means of a galvanic process. In a second photolithographic step, a protective layer (1) is structured such that a structure is formed in the protective layer (1) above the cavities (6) etched into the layer (3). The depth of the thus formed cavity (6) is the sum of the depth of the etched structure and the thickness of the protective layer (1).
  • Preferably, the cavities are arranged such that a regular, preferably Cartesian, grid of columns and lines is formed on the carrier plate. The size and shape of the carrier plate can be selected as desired and can easily be adapted to the detection system used. If spotting robots are used for immobilizing the anchors or interaction partners, or if the binding surface is present on microtitration plates, the distance between the fields should preferably be adjusted to the microtitration format or the spotting device used. The number of fields can also exceed the number of subunits of the microtitration plate, i.e. multiple fields can be used per surface. For example, a square carrier plate of about 12×12 cm may comprise a total of 9216 fields which can be covered by means of a pipetting robot from six conventional 1536 microtitration plates. [0089]
  • However, a spatially structured presentation of identical or different immobilized to interaction partners can also be achieved by covalently binding the interaction partner to a predetermined portion of the binding surface after an amount of liquid has selectively been applied, without it being necessary to physically divide the surface into individual compartments. For example, techniques for applying reagent spots onto metal or metal oxide surfaces as described in EP-A-0 872 735 can analogously be applied to the binding surfaces of the present invention. If solutions of the interaction partners/ligands are applied onto a homogeneous binding surface, the concentration of the ligand/interaction partner on the finished measuring surface is determined by the dilution of the binding surface. Consequently, there is the additional advantage compared to EP 872 735 A1 that precisely defined amounts of liquid only have to be applied to the solid phase carrier surface once to generate a measuring array (namely when the binding layer is generated). [0090]
  • The present invention can be employed in HTS, in the research of active substances or in medical diagnostics. Suitable measuring methods for detecting interaction between an immobilized (surface-bound) interaction partner/ligand and a free interaction partner/receptor wherein the solid phase carrier only serves for the immobilization of one interaction partner, are based on the verification of the specific binding reaction by means of electrochemical (electro immunoassays), radiochemical (e.g. radioimmunoassay), mass-sensitive or optical processes such as fluorescence or luminescence measurements, in particular enzyme assays. For the latter, the so-called ELISA technique (enzyme-linked immunosorbent assay, immunoassays using solid-phase technique) is preferred. [0091]
  • Reflecto-optical processes such as surface plasmone resonance are suitable for a marker-free detection of the interactions. Here, the solid phase carrier is part of the sensor system. [0092]
  • However, due to the advantages described above, the surfaces of the present invention can also be used in classic processes such as affinity chromatography. [0093]
  • EXAMPLES Example 1
  • Synthesis of a Diluting Component [0094]
  • a) Immobilization of N-(N[0095] 5-Fmoc-5-aminopentyl)-11-mercaptoundecaneamide on chlorotrityl resin
    Figure US20040023366A1-20040205-C00002
  • 600 mg (1.15 mmol) N-(N[0096] 5-Fmoc-aminopentyl)-11-mercaptoundecaneamide, obtainable from S-protected 11-mercaptoundecaneamide and Fmoc-1,5-diaminopentanehydro-chloride, were dissolved in 15 ml DMF and 2 g methoxytrityl-chloride resin (1.6 mmol) (Novabiochem) were added. The suspension was carefully shaken for 1 hour. Then, 500 μl pyridine were added and the suspension was shaken for another 3 hours. Afterwards, the resin was washed once with N,N-dimethylformamide (DMF), once with 5% water in DMF, four times with DMF, three times with dichloromethane and twice with hexane and dried under vacuum. The loading of the resin with N-(N5-Fmoc-5-aminopentyl)-11-mercaptoundecaneamide was determined by means of Fmoc analysis (G. B. Fields, R. L. Noble, Int. J. Peptide Protein Res. 1990, 35, 161-214) to be 0.35 mmol/g (yield: 60% of the theoretical value).
  • b) General Protocol for Coupling Fmoc-8-amino-3,6-dioxa-octanoic acid (Fmoc-Ado) [0097]
    Figure US20040023366A1-20040205-C00003
  • For cleaving off the Fmoc protective group, 1 g of the loaded resin (0.35 mmol) was carefully stirred for 20 min in 15 ml ⅓ (v/v) piperidine/DMF and then washed six times with DMF. The coupling of Fmoc-8-amino-3,6-dioxa-octanoic acid was carried out by incubating the resin for 4 hours with a solution of 270 mg (0.70 mmol) Fmoc-8-amino-3,6-dioxa-octanoic acid. 270 mg (0.71 mmol) O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluroniumhexafluorophosphate (HATU) and 250 μl (1.44 mmol) ethyl-diisopropylamine (DIEA) in 7 ml DMF. Then the resin was washed live times with DMF, three times with dichloromethane and twice with hexane and then dried. [0098]
  • c) Synthesis of a Diluting Component [0099]
    Figure US20040023366A1-20040205-C00004
  • As was described in item b), Fmoc-8-amino-3,6-dioxa-octanoic acid was coupled to 500 mg resin (0.175 mmol) from b) and then the Fmoc protective group was cleaved off as described in b). Subsequently, the free amino groups were acetylated by incubating the resin for 30 min with 10 [0100] ml 1/1/2 (v/v/v) acetic acid anhydride/pyridine/DMF. Then the resin was washed five times with DMF and three times with dichloromethane. Cleaving of the product from the resin was carried out with 2/18/1 (v/v/v) trifluoroacetic acid/dichloromethane/triethylsilane. The product was purified by means of preparative RP-HPLC and analyzed by means of LC/MS.
  • LC-MS (calc.): [M+H][0101] + 635.5 (635.4), [M+Na]+ 657.5 (657.4)
  • Example 2
  • Synthesis of an Anchor Component [0102]
    Figure US20040023366A1-20040205-C00005
  • As described in 1b), Fmoc-8-amino-3,6-dioxa-octanoic acid was twice coupled to 100 mg of the resin of 1b) and then the Fmoc protective group was cleaved off. Then 3-maleinimidopropionic acid was coupled by a 1-hour incubation of the resin with 4 eq. 3-maleinimidopropionic acid and 4 eq. diisopropylcarbodiimide in DMF (c=0.15 M). [0103]
  • Then the resin was washed five times with DMF and three times with dichloromethane. Cleaving of the product from the resin was carried out with 2/18/1 (v/v/v) trifluoroacetic acid/dichloromethane/triethylsilane. The product was purified by means of preparative RP-HPLC and analyzed by means of LC/MS. [0104]
  • LC-MS (calc.): [M+H][0105] + 889.2 (889.5) [M+Na]+ 911.1 (911.5)
  • Example 3
  • Preparation of a Binding Surface [0106]
  • The binding surface consists of the anchor component from Example 2 and the diluting component from Example 1. [0107]
  • First of all, stock solutions of the individual components are prepared by dissolving the solids in ethylene glycol/0.1% TFA with the desired molar concentration which is verified by means of Ellman's test (G. L. Ellman, Arch. Biochem. Biophys. 82 (1959), 70-77) or based on the extinction coefficient at 295 nm. Then the stock solutions of anchor and diluting components are mixed in the desired ratio (1:10, v/v) and the gold surface is incubated with this 100 μM to 1 mM solution for 1 hour at room temperature. It is then washed with methanol/0.1% TFA and several times with water/acetic acid (2 ppm) and dried in a nitrogen stream. [0108]
  • Example 4
  • Immobilization of a Ligand (“Small Molecule”) [0109]
  • The coupling of Ac-p-Tyr-Cys-NH[0110] 2, to the binding surface from Example 3 is carried out by incubating a 10-100 μM solution of the compound for 15 to 30 min. Then the surface is rinsed with acetic acid (2 ppm) in water and dried. Subsequently, it is washed with acetic acid (2 ppm) in water and with isopropanol and the surface is dried in a nitrogen stream.
  • Example 5
  • Immobilization of Proteins on a Biacore® J1 Chip [0111]
  • The preparation of the protein surface is carried out by incubating the binding surface from Example 3 (1:10 dilution) with a 17 μM solution of caspase 7T7 (modified by means of a T7 and an oligo-His tag) in 0.2 M NaH[0112] 2PO4/Na2HPO4, pH 7, (20 min, room temperature). Then the chip is washed with acetic acid/water (1 μl/500 ml), treated for 5 min with 10 mM 2-mercaptoethanol in 0.2 M NaH2PO4/Na2HPO4, pH 7, washed with acetic acid/water (see above) and dried in a nitrogen stream.
  • The success of the protein immobilization is verified by means of SPR on [0113] Biacore® 3000 by spraying on a 6.7 nM solution (HBS, pH 7.4) of an anti-T7 antibody (Novagen) or a 11 nM solution (HBS, pH 7.4) of an anti-oligo-histidine antibody (Sigma).
  • The change in the SPR signal (in RU) after spraying on 100 μl antibody solution (flow rate: 10 μl/min, run buffer: HBS, pH 7.4) was as follows: [0114]
    antibody RU
    anti-T7 antibody, 6.7 nM 206
    anti-oligo-His antibody, 11 nM 1067
  • Example 6
  • Grazing-angle FTIR [0115]
  • The goal of this measuring method is the vibrational spectroscopy of a thin layer located on a metal surface. The IR light at a grazing incidence is reflected off the metal surface. Measurements are only carried out with IR light polarized in the incidence plane since light with a perpendicular polarization direction cannot contribute to the measurement. Furthermore, only the transition dipole moments which comprise portions at least parallel to the E-field of the radiation contribute to the absorption. [0116]
  • When the molecules located on the surface are oriented, additional selection rules for the appearance of the bands in the spectrum are available. This measuring method is now sensitive enough to measure IR spectra of even monomolecular films, typically resulting in absorption values of some mOD. In the following measurements, a commercially available FTIR device (Bio-Rad FTIR Spectrometer Model FTS 175C) was used. [0117]
  • The upper plot in FIG. 2 shows the FTIR spectrum of the pure diluting component (A). The CH[0118] 2 bands can be seen in the area of 2.900 cm−1, the CONH bands in the area of 1.500 to 1,700 cm−1 and the C—O—C band at 1.122 cm−1. The bottom-most plot shows the spectrum of the pure anchor component with ligand (B). Compared to the spectrum of the diluant, the band at 1.716 cm−1 stands out. This band can be assigned to the imide group of the anchor component. When surfaces are prepared with an increasing anchor component concentration (spectra from top to bottom), a systematic increase in the imide band can be observed. For a quantitative evaluation, the area of the peak at 1.716 cm−1 was determined and plotted against the concentration of the anchor component (cf FIG. 3). A linear fit to the measuring data clearly shows the connection and thus the conformity of the anchor molecule concentration on the gold surface and the concentration in the solution during gold coating.
  • Example 7
  • XPS and Ellipsometry Measurements [0119]
  • In X-ray photoelectron spectroscopy (XPS) the kinetic energy of the electrons formed after the interaction of atoms and mono-energetic X-rays according to the photoelectric effect is verified. As an X-ray source., use was made of MgK[0120] α1,1 line (h=1253.6 eV) which was aimed at the surface to be tested at an angle of 45°. The detector measures the number of electrons exiting at 45°, classified according to its energy. In the energy spectrum obtained there are peaks at energy levels that correspond to the difference between the energy of the incident X-rays and the binding energy of the electron in the atom. These binding energies are specific to every atom and it is therefore possible to determine the composition of the molecules on the surface. The peak stemming from the Au4f electrons is particularly large. Since from the point of view of the detector the gold is located behind the molecule layer, the electrons detached from the gold will be re-absorbed by the molecule layer. Therefore, the layer thickness d of the sample can be determined based on the Au4f peak of the sample (Isample) and the corresponding peak of a pure gold surface (I0) using the formula d=λ(1nI0)/1n(Isample). In this case λ=37 Å is the mean free path of the 1.169.6 eV electrons in the layer.
  • In ellipsometry, the change in the polarization state of light is measured when it is reflected off the system consisting of molecule layer, gold layer and substrate. According to the Fresnel formulae, there is a reflected and a transmitted light ray at every boundary layer of this system. Since these layers are very thin, multiple reflections occur within the molecule and the gold layers, i.e. the amplitudes of the light rays have to be summed up according to their phases. The result of this summation is a reflection coefficient for the light component polarized in the incidence plane and a second one for the component polarized in a perpendicular direction. These two mathematical expressions comprise the parameters of the layer thicknesses and the (complex) refractive indices of the materials. These two reflection coefficients are determined by means of a spectroscopic ellipsometer (in this case J. A. Woollam Co., Inc. M-44®), also as a function of the wavelength. It is then possible to determine the thickness of the molecule layer from the measured wavelength-dependent reflection coefficients taking into account the refractive index of the molecule layer (here 1.45). [0121]
  • The layer thicknesses obtained from XPS measurements and ellipsometric measurements correspond to the length of the diluting molecule from Example 1. In a stretched configuration, the molecular length of the diluting component is 44 Å. Since the molecules do not protrude at a right angle from the surface but are usually bound at a specific angle, the somewhat shorter measured layer thickness (34 Å) indicates that these surfaces are SAiMs. A bond angle of the molecules of about 50° with respect to the surface is therefore conceivable. [0122]
  • Example 8
  • Unspecific Protein Adsorption Resistance of Different Diluants [0123]
  • The protein adsorption resistance was determined by means of surface plasmone resonance on [0124] Biacore® 3000 with the following proteins:
    ribonuclease A (bovine) 0.1 mg/ml
    lysozyme (chicken) 0.1 mg/ml
    egg albumin (chicken) 0.1 mg/ml
    D-amino-acid oxidase (procine) 0.1 mg/ml
    pyruvate kinase (rabbit) 0.1 mg/ml
    fibriogen (human) 1 mg/ml
    glutathione reductase (wheat germ) 1 mg/ml
  • For the determination of the protein adsorption resistance, all proteins were dissolved in HBS pH 7.4 at the concentrations given above, and 100 μl of each were sprayed to Biacore® 3000 (flow rate: 20 μl/min, run buffer: HBS pH 7.4) to the corresponding surfaces and the change in the SPR signal (Response Units=RU) was measured. An increase of 1,000 RU corresponds to an amount of protein of 1 ng/mm[0125] 2.
  • The values shown in FIG. 4 are average values from 4 measurements. [0126]
  • A short-chain disulfide (compound A of EP 698 787), the diluting molecule from Example 1 and 12,15,18-trioxa-20-hydroxyicosan-1-thiol (Whitesides et al. J.Am.Chem.Soc. 1995, 117, 12009-10) were used as a diluants. As can be seen from the results, the chain length of the molecule influences the unspecific adsorption resistance, which increasingly occurs in short-chain diluting molecules. [0127]
  • Example 9
  • Quantitative Reaction of Phosphotyrosine (pY) at the Binding Surface [0128]
  • Analogously to Example 4, pY with cysteine as additional molecule is used to bind pY to the binding surface from Example 3 (“pY tag”). [0129]
  • A gold chip was incubated with a 1:10 mixture of the anchor component from Example 2 and the diluting component from Example 1 in ethylene glycol and 1% TFA at a total concentration of 0.5 mM. Then the chip was washed several times in methanol/0.1% TFA and then in water pH 7.0. Chips pretreated in this manner were dried in a nitrogen stream. [0130]
  • For determining in which concentration range a complete reaction of the presented maleimide functionalities takes place and thus a constant signal intensity of the luminescence can be observed, 0.15 μl of pY tag were applied in dilutions of 10 μM, 1 μM, 100 nM and 10 nM. Four measurements were carried out for each concentration. In the free areas, the maleimide groups were then saturated by incubating the chip in 0.2 M phosphate buffer pH 7.0, 10 mM mercaptoethanol for 30 min. [0131]
  • Such chips were then incubated overnight in BSA blocking solution (50 mM Tris/HCl, 150 mM NaCl, 5 g/l BSA, 0.05% (v/v) Tween-20®, pH 7.3 ). The verification of the pY groups on the surface of the chip was carried out by means of an immunoassay wherein the chip was first incubated with a 1:5,000 a-pY antibody in blocking solution and then with 1:5,000 anti-mouse-Fab-POD. Between the incubation steps, the blocking solution was washed (2×1 min). Prior to the detection of the luminescence signal with the Super-Signal-Plus Substrate from Roche Diagnostics, it was again washed in TBS buffer. The luminescence reaction was observed in a Lumi-Imager (Roche Diagnostics). Afterwards, the signal intensities were integrated and are shown in the Figure. The standard deviations of the four measurements were calculated and can be inferred from FIG. 5 as error bars. [0132]
  • The results show that the signal intensities are constant throughout a concentration range of three powers of ten within the limit of error. A decrease in the signal can only be observed when the pY tag is highly diluted. The consistency of the signal results from the fact that the reaction of the maleimide with the thiol is approximately quantitative as long as the thiol is present in a certain excess. [0133]
  • Example 10
  • Preparation of an Array with a Plurality of Measuring Surfaces [0134]
  • A 12×12 cm gold chip (40 nm gold/1 nm chromium on glass) was incubated with a 1:10 mixture (dilution) of anchor component from Example 2 and diluting component from Example 1 in ethylene glycol and 1% trifluoroacetic acid (TFA) at a total concentration of 1.0 mM. Then the chip was washed several times in methanol/1% TFA and then in water pH 7.0. Chips pretreated in this manner were dried in a nitrogen stream. [0135]
  • Subsequently, a library of 9216 thiol-containing ligands were deposited on these chips by means of a spotting device in an arrangement of 96×96 spots with a distance of 1.125 mm between the spots. The thiol-containing ligands applied onto the surface are dissolved in a 40 μM solution of 0.2 M Pi, 5 mM EDTA and 10% (v/v) ethylene glycol pH 7.0. The spotting device releases about 10 nl per spot so that in each spot a high excess of the thiol-containing ligand compared to the surface-bound maleimide group is guaranteed and a complete reaction of the maleimide groups can be achieved. In the free areas, the maleimide groups were then saturated by incubating the chip in 0.2 M phosphate buffer pH 7.0, 10 mM mercaptoethanol for 30 min. [0136]
  • Such chips were then incubated overnight in BSA blocking solution (50 mM Tris/HCl, 150 mM NaCl, 5 g/1 BSA, 0.05% (v/v) Tween-20®, pH 7.3). The analysis of potential binding partners of the target protein thrombin was carried out by means of an immunoassay. [0137]
  • First of all, the chip was incubated for 4 hours in 10 nM thrombin in blocking solution. After washing twice for two minutes in blocking solution, a 1:1,000 dilution of a polyclonal anti-thrombin antibody was incubated with the chip for 2 hours. After again washing twice in blocking solution, an anti-rabbit antibody POD conjugate was incubated with the chip for 2 hours in order to determine the binding activity. Finally, the chip was washed twice for 2 min in TBST. The chemical luminescence reaction was verified by reacting the Lumi-Light Plus Substrate in the Lumi Imager (Roche Diagnostics, Germany). FIG. 6 shows a photograph of the chemical luminescence reaction (10 nM thrombin). Black spots (represented in the Imager as light spots) indicate binding of the thrombin. Discrete intensities can be seen as spots in certain positions. Since every compound on the array has certain spatial coordinates, specific chemical structures can be assigned to the spots. [0138]
  • Example 11
  • Synthesis of Further Anchor and Diluting Components [0139]
  • 1) Structure of Further Anchor and Diluting Compounds (Components) Based on the Reaction of 4-methoxytrityl-protected mercaptoundecanoic acid with Different Diamines. [0140]
  • Reaction Mixtures: [0141]
  • 1 g (2.04 mmol) S-(4-methoxytrityl)-mercaptoundecanoic acid [0142]
  • 462 mg (2.24 mmol) dicyclohexylcarbodiimide (DCC) [0143]
  • 236 mg (2.04 mmol) n-hydroxysuccinimide (NHS) [0144]
  • a) 755.3 mg (850.6 μl; 10.2 mmol) 1.3-diaminopropane [0145]
  • b) 1.51 g (1.49 ml; 10.2 mmol) 1.8-diamino-3,6-dioxaoctane [0146]
  • c) 1.04 g (1.19 ml; 10.2 mmol) 1,-diaminopentane [0147]
    Figure US20040023366A1-20040205-C00006
  • Preparation: [0148]
  • 1 g S-(4-methoxytrityl)-mercaptoundecanoic acid, 462 mg (2.24 mmol) DCC and 236 mg (2.04 mmol) NHS were each dissolved in 20 ml dichloromethane (DCM) and stirred at room temperature for 30 min. The three diamines a) to c) were each dissolved in 20 ml DCM and the S-(4-methoxytrityl)-mercaptoundecanoic acid NHS-ester was added in 10 portions over a time period of 30 min to the diamine solutions under stirring. The reaction mixtures were stirred for 3 hours, the urea was filtered off and the solvent was evacuated in a vacuum. The products were dissolved in ethyl acetate and were each washed three times with 10% sodium carbonate solution, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were dried over sodium sulfate and concentrated in a rotary evaporator until dry. [0149]
  • 2) Extension of the Amines (1) to (3) with Trioxaundecanoic Acid. [0150]
    Figure US20040023366A1-20040205-C00007
    Figure US20040023366A1-20040205-C00008
  • Reaction Mixtures: [0151]
  • 1.27 g (5.71 mmol) 3,6,9-trioxaundecanedioic acid [0152]
  • 431.6 mg (2.09 mmol) DCC [0153]
  • 220.2 mg (1.90 mmol) NHS [0154]
  • 1.23 g (1.62 ml) diisopropylethylamine (DIEA) [0155]
  • a) 1.04 g (1) (1.90 mmol) [0156]
  • b) 1.17 g (2) (1.90 mmol) [0157]
  • c) 1.09 g (3) (1.90 mmol) [0158]
  • Preparation for a) to c): [0159]
  • 1.27 g (5.71 mmol) 3,6,9-trioxaundecanedioic acid, 431.6 mg (2.09 mmol) DCC, 220.2 mg (1.90 mmol) NHS and 1.23 g (1.62 ml) diisopropylethylamine (DIEA) were each dissolved in 30 ml DCM and stirred at room temperature for 30 min. Then, 1.90 mmol of amines (1) (1.04 g), (2) (1.17 g) and (3) (1.09 g) were each dissolved in 20 ml DCM and added in 10 portions over a time period of 30 min to the three reaction mixtures under stirring. The mixtures were stirred for 3 hours at room temperature, then the urea was filtered off and the solvent was concentrated until dry. The products (4), (5) and (6) were dissolved in ethyl acetate and were each washed three times with 0.1 M HCl, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were then dried over sodium sulfate and concentrated until dry. [0160]
  • Preparation of the Anchor (13) and the Corresponding Diluants (7) and (8) [0161]
  • Reaction Mixtures: [0162]
  • 0.72 g (0.873 mmol) (5) [0163]
  • 198 mg (0.96 mmol) DCC [0164]
  • 101.8 mg (0.873 mmol) NHS [0165]
  • a) 5 ml 26% aqueous ammonia [0166]
  • b) 266.5 mg (263.4 μL) (4.36 mmol) 2-aminoethanol [0167]
    Figure US20040023366A1-20040205-C00009
  • Preparation: [0168]
  • 0.72 g (0.873 mmol) (5), 198 mg (0.96 mmol) DCC and 101.8 mg (0.873 mmol) NHS were each dissolved in 30 ml DCM and then stirred for 30 min at room temperature. Then a) 5 ml 29% aqueous ammonia and b) 266.5 mg (263.4 μL) (4.36 mmol) 2-aminoethanol were added under vigorous stirring, and stirring was continued for another 2 hours. Then the urea was filtered off and the solvent was concentrated until dry. The products (7) and (8) were dissolved in ethyl acetate (EE) and were each washed three times with 10% sodium carbonate solution, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were dried over sodium sulfate and concentrated until dry. [0169]
  • For cleaving off the 4-methoxytrityl protective group, compounds (7) and (8) were each dissolved in 20 ml trifluoroacetic acid, 2 ml triethylsilane was added and the mixture was stirred at room temperature for 30 min. The solutions were concentrated until dry and the products were purified by means of preparative HPLC and analyzed by means of LC/MS. [0170]
  • LC/MS (calc.): [0171]
  • [M+H]+ 552.3(552.7), [M+Na]+ 574.3 (574.7)   (7):
  • [M+H]+ 596.3 (596.8), [M+Na]+ 618.3 (618.8)   (8):
  • 200 mg of (5) were reacted with 1,2-diaminoethane analogously to 1a. Then 3-maleinimidopropionic acid was added as described in 4 and the 4-methoxytrityl protective group was cleaved off. The product (13) was purified by means of preparative HPLC and analyzed by means of LC/MS. [0172]
  • LC/MS (calc.): [M+H][0173] + 746.6 (746.9), [M+Na]+ 748.5 (748.9)
  • 3) Preparation of the Anchors (11) and (12) and the Accompanying Diluants (9) and (10) [0174]
  • Formulation: [0175]
  • a) 0.73 g (6) (0.94 mmol) [0176]
  • b) 0.45 g (4) (0.94 mmol) [0177]
  • 213.3 mg (1.034 mmol) DCC [0178]
  • 108.9 mg (0.940 mmol) NHS [0179]
  • 417.9 mg (411.8 μl; 2.82 mmol) 1,8-diamino-3,6-dioxaoctane [0180]
    Figure US20040023366A1-20040205-C00010
  • Preparation: [0181]
  • 0.73g (0.94 mmol) (6) and 0.45 g (0.94 mmol) (4) were each dissolved in 20 ml DCM and 213.3 mg (1.034 mmol) DCC and 108.9 mg (0.940 mmol) NHS were added to each solution, which were stirred at room temperature for 30 min. Then 418 mg (412 μl; 2.82 mmol) 1,8-diamino-3,6-dioxaoctane were dissolved in two batches of 20 ml DCM and added to the two reaction mixtures under vigorous stirring. After 3 hours of stirring at room temperature, the urea was filtered off and the solvent was concentrated. The products were dissolved in ethyl acetate and were each washed three times with 10% sodium carbonate solution, twice with a saturated sodium chloride solution and twice with distilled water. The solutions were dried over sodium sulfate and again concentrated until dry. [0182]
  • 500 mg each of (4a) and (6a) were dissolved in 20 ml tetrahydrofuran. 2 ml acetic acid anhydride and 2 ml pyridine were added to each solution, which were stirred for 30 min. Then the solvent was concentrated, the products were taken up in ethyl acetate and washed three times with 0.1 M HCl, twice with a saturated NaCl solution and twice with distilled to water. The solutions were dried over sodium sulfate and concentrated until dry. [0183]
  • For cleaving off the 4-methoxytrityl protective group, the products were each dissolved in 20 ml TFA and 2 ml triethylsilane and the mixture was stirred at room temperature for 30 min and then concentrated until dry. The resulting compounds (9) and (10) were purified by means of preparative HPLC and analyzed by means of LC/MS. [0184]
  • LC/MS (calc.): [0185]
  • [M+H]+ 651.5 (651.9), [M+Na]+ 673.5 (673.9)   (9)
  • [M+H]+ 679.6 (679.9), [M+Na]+ 701.6 (701.9)   (10)
  • 200 mg each of (4a) and (6a) (0.22 mmol) were dissolved in 10 ml DCM and to each was added a solution of 43 mg (0.25 mmol) 3-maleinimidopropionic acid and 52 mg (0.25 mmol) DCC in 5 ml DCM, and stirred at room temperature for 3 hours. Then the urea was filtered off the solutions were concentrated until dry, and for cleaving off the protective group the products were each dissolved in 10 ml TFA with 2 ml triethylsilane. After 30 min stirring at room temperature, the solutions were concentrated until dry; the products (11) and (12) were purified by means of preparative HPLC and analyzed by means of LC/MS. [0186]
  • LC/MS (calc.): [0187]
  • [M+H]+ 788.6 (788.9), [M+Na]+ 810.5 (810.9)   (11)
  • [M+H]+ 760.5 (760.9), [M+Na]+ 782.3 (782.9)   (12)

Claims (21)

1. Process for the generation of a binding layer on the metal surface of a solid phase carrier, comprising the step of contacting a solution of a plurality of identical or different anchor molecules represented by the formula
HS—R-M
wherein the structural moiety R provides for the formation of a self-assembling monolayer on the surface and M represents a mercaptophilic head group, with the surface under acidic conditions.
2. Process according to claim 1, wherein a plurality of identical or different diluting molecules represented by the general formula
HS—R—X
wherein R is defined as in claim 1 and X represents a non-mercaptophilic head group, is contacted with the surface together with the anchor molecules.
3. Process according to claim 2, wherein the ratio of anchor molecules to diluting molecules ranges from 1:2 to 1:10.000.
4. Binding surface comprising a binding layer obtainable according to the process of any of claims 1 to 3 and a solid phase support.
5. Binding surface according to claim 4, wherein a planar, non-structured carrier plate or a physically structured carrier plate having a plurality of separate fields for binding the anchors and diluants is used as the solid phase support.
6. Measuring surface obtainable by covalently binding a plurality of identical or different biospecific interaction partners which carry a thiol group or which are modified by means of a thiol group to a binding surface according to any of claims 4 or 5.
7. Measuring surface according to claim 6, wherein the bound biospecific interaction partners are selected from proteins, peptides, oligonucleotides, carbohydrates, isoprenoids, enzymes, lipid structures, saccharides, antibodies, peptide hormones, cytokines, antibiotics and small organic molecules, the small organic molecules being characterized by having a molecular weight ranging from 50 to 3,000 g/mol.
8. Measuring array comprising a plurality of measuring surfaces according to claim 6 or 7, wherein the individual measuring surfaces differ from each other at least in the structure of the bound interaction partners.
9. Process for providing a measuring array according to claim 8, wherein solutions of the interaction partners are applied onto defined, spatially limited areas of the binding surface.
10. Process according to claim 9, wherein the solution of the interaction partners is applied by means of a pipetting device, a spotting device, a micropipetting device or an ink-jet process.
11. Process for the detection and/or quantification of an interaction between surface-bound and free interaction partners, comprising contacting the free interaction partners with a measuring surface or a measuring array according to any of claims 6 to 8.
12. Process according to claim 11, wherein an interaction of the surface-bound interaction partners takes place with one or more receptors as free interaction partners, selected from proteins, DNA, RNA, oligonucleotides, prosthetic groups, vitamins, lipids, mono-, oligo- or polysaccharides or fusion proteins or synthetic primers.
13. Use of a measuring surface or a measuring array according to any of claims 6 to 8 for interaction analysis, in a screening process or in affinity chromatography.
14. Use of a measuring surface or a measuring array according to any of claims 6 to 8 in medical diagnostics.
15. Anchor molecule of the formula
HS—R-M
wherein the structural moiety R provides for the formation of a self assembling to monolayer on the surface and M represents a mercaptophilic head group suitable for covalently binding to a thiol-functionalized ligand.
16. Anchor molecule according to claim 15, wherein R comprises a hydrophobic structural moiety Ra, formed by a branched or straight-chain hydrocarbon chain of 5 to 50 carbon atoms, which may be saturated or partially unsaturated.
17. Anchor molecule according to claim 15 or 16, wherein R comprises a branched or non-branched hydrophilic spacer Rb, formed by hydrocarbon chain of 5 to 1,000 carbon atoms which is interrupted by heteroatoms.
18. Anchor molecule according to one of claims 15 to 17, wherein R has the following general formula
—(CH2)a-Q1-(CH2)b-{[Q2-(CH2)c—[O—(CH2)d]e—O—(CH2)f]g-[Q3-(CH2)c—[O—(CH2)d′]e′—O—(CH2)f′]h}i-Q4-(CH2)j-Q5-(CH2)k
wherein the variables are defined independently of each other as follows and the numeric ranges comprise their respective limiting values as well as the integers contained therein:
Q1,Q5 are —NH—C(O)—, —C(O)—NH— or a bond
Q2,Q3,Q4 are —NH—C(O)— or —C(O)—NH—
a is 5 to 20, preferably 8 to 12, particularly preferred 10;
b is 0 to 5, preferably 0 if Q1 is a bond and 1 to 10, preferably 2 to 7, particularly preferred 3 to 5in all other cases;
c, c′ are 1 to 5, preferably 1 to 3, particularly preferred 1;
d, d′ are 1 to 5, preferably 1 to 3, particularly preferred 2;
e, e′ are 1 to 5, preferably 1 to 3, particularly preferred 2;
f, f′ are 1 to 5, preferably 1 to 3, particularly preferred 1;
g, h are 0 to 3, provided that g+h≧1, preferably g+h=2;
i is 1 to 3, preferably 1 to 2, particularly preferred 1;
j is 0 to 5, preferably 1 to 3, particularly preferred 2; and
k is 0 to 5.
19. Anchor molecule according to any of claims 15 to 18, wherein M is selected from iodo- or bromoacetamide, pyridylthio compounds, Michael acceptors, acrylic acid, (meth)acrylic acid esters, -amides, -lactones, -lactames, methylene-gem-difluorocyclopropanes, α,β-unsaturated aldehydes and ketones and α,β-unsaturated sulfones and sulfonamides.
20. Anchor molecule according to one of claims 15 to 18, wherein M has the following structure
Figure US20040023366A1-20040205-C00011
wherein
R1 and R2 are independently hydrogen or C1-C5 alkyl,
R3 and R4 are independently hydrogen or C1-C5 alkyl or
R3 and R4together form ═O.
21. Process for the solid phase synthesis of an anchor molecule according to any of claims 15 to 20, wherein the anchor molecule is coupled to the synthesis phase via the thiol group and is cleaved off in an acidic medium to provide the free anchor molecule.
US10/297,244 2000-06-02 2001-06-01 Surface for the immobilization of ligands Abandoned US20040235146A9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/980,015 US7955638B2 (en) 2000-06-02 2007-10-30 Surface for the immobilization of ligands

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE100-27-397.1 2000-06-02
DE10027397A DE10027397A1 (en) 2000-06-02 2000-06-02 Surface for the immobilization of ligands
PCT/DE2001/002072 WO2001092883A2 (en) 2000-06-02 2001-06-01 Surface for the immobilisation of ligands

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/980,015 Division US7955638B2 (en) 2000-06-02 2007-10-30 Surface for the immobilization of ligands

Publications (2)

Publication Number Publication Date
US20040023366A1 true US20040023366A1 (en) 2004-02-05
US20040235146A9 US20040235146A9 (en) 2004-11-25

Family

ID=7644474

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/297,244 Abandoned US20040235146A9 (en) 2000-06-02 2001-06-01 Surface for the immobilization of ligands
US11/980,015 Expired - Fee Related US7955638B2 (en) 2000-06-02 2007-10-30 Surface for the immobilization of ligands

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/980,015 Expired - Fee Related US7955638B2 (en) 2000-06-02 2007-10-30 Surface for the immobilization of ligands

Country Status (8)

Country Link
US (2) US20040235146A9 (en)
EP (1) EP1287361B1 (en)
JP (1) JP2003536056A (en)
AT (1) ATE374368T1 (en)
AU (2) AU7232801A (en)
CA (1) CA2413855A1 (en)
DE (3) DE10027397A1 (en)
WO (1) WO2001092883A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020127577A1 (en) * 1999-05-28 2002-09-12 Jutta Eichler Bioactive sensors
WO2006002141A2 (en) * 2004-06-18 2006-01-05 Wisconsin Alumni Research Foundation Detection of post-translationally modified peptides with liquid crystals
US20080054256A1 (en) * 2006-09-05 2008-03-06 Hyoyoung Lee Molecular electronic device including organic dielectric thin film and method of fabricating the same

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2435608A1 (en) * 2001-02-05 2002-08-15 Graffinity Pharmaceutical Design Gmbh Low affinity screening method
EP1360493A1 (en) * 2001-02-07 2003-11-12 Graffinity Pharmaceutical Design GmbH Screening method using solid supports modified with self-assembled monolayers
DE10245651A1 (en) * 2002-09-30 2004-04-08 Graffinity Pharmaceuticals Ag Quality control of microarray sensor fields
EP1439394A1 (en) * 2002-12-20 2004-07-21 Graffinity Pharmaceuticals Aktiengesellschaft Method and apparatus for etablishing ADME profiles in the early stage of develloppment of active substances
DE10308894A1 (en) * 2003-02-28 2004-09-09 Graffinity Pharmaceuticals Ag Quality control of binding measurements on microarrays
WO2006132326A1 (en) * 2005-06-09 2006-12-14 Hiroshima University Method for immobilizing vital cell and sensor for measuring vital cell activation function
HUP0600668A2 (en) 2006-08-22 2008-02-28 Avicor Kft Active carrier, process for producing thereof and the use of thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6475808B1 (en) * 1998-07-14 2002-11-05 Zyomyx, Incorporated Arrays of proteins and methods of use thereof

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5156989A (en) * 1988-11-08 1992-10-20 Siliconix, Incorporated Complementary, isolated DMOS IC technology
WO1990005503A1 (en) * 1988-11-23 1990-05-31 California Medical Products, Inc. A head immobilizer
US5387555A (en) * 1992-09-03 1995-02-07 Harris Corporation Bonded wafer processing with metal silicidation
DE59106527D1 (en) * 1990-11-14 1995-10-26 Hoffmann La Roche Immunosensitive transducer and method for its production.
DE4219159A1 (en) * 1992-06-11 1993-12-16 Boehringer Mannheim Gmbh Self-assembling monolayer with short chain linkers
US5763191A (en) * 1990-12-12 1998-06-09 Boehringer Mannheim Gmbh Universal binding film
US5386136A (en) * 1991-05-06 1995-01-31 Siliconix Incorporated Lightly-doped drain MOSFET with improved breakdown characteristics
KR950013435B1 (en) * 1992-05-22 1995-11-08 삼성전자주식회사 Solid image sensor for mirror image
US5439842A (en) * 1992-09-21 1995-08-08 Siliconix Incorporated Low temperature oxide layer over field implant mask
US5648281A (en) * 1992-09-21 1997-07-15 Siliconix Incorporated Method for forming an isolation structure and a bipolar transistor on a semiconductor substrate
US5422508A (en) * 1992-09-21 1995-06-06 Siliconix Incorporated BiCDMOS structure
US5414292A (en) * 1993-05-26 1995-05-09 Siliconix Incorporated Junction-isolated floating diode
US5817546A (en) * 1994-06-23 1998-10-06 Stmicroelectronics S.R.L. Process of making a MOS-technology power device
DE4430023A1 (en) * 1994-08-24 1996-02-29 Boehringer Mannheim Gmbh Electrochemical sensor
US5545909A (en) * 1994-10-19 1996-08-13 Siliconix Incorporated Electrostatic discharge protection device for integrated circuit
US5777362A (en) * 1995-06-07 1998-07-07 Harris Corporation High efficiency quasi-vertical DMOS in CMOS or BICMOS process
US6391754B1 (en) * 1996-09-27 2002-05-21 Texas Instruments Incorporated Method of making an integrated circuit interconnect
US5798295A (en) * 1997-06-09 1998-08-25 Motorola, Inc. Method for forming a buried contact on a semiconductor substrate
DE19751658A1 (en) * 1997-11-21 1999-07-29 Wolfbeis Otto S Prof Dr Process for the formation of laterally organized structures on support surfaces
US6303961B1 (en) * 1998-04-29 2001-10-16 Aqere Systems Guardian Corp. Complementary semiconductor devices
KR100272176B1 (en) * 1998-09-30 2000-12-01 김덕중 Method fabricating bicdmos device
KR100281908B1 (en) * 1998-11-20 2001-02-15 김덕중 Semiconductor device and manufacturing method
DE19924606A1 (en) * 1999-05-28 2000-11-30 Graffinity Pharm Design Gmbh Ligand-anchor conjugates
US6501131B1 (en) * 1999-07-22 2002-12-31 International Business Machines Corporation Transistors having independently adjustable parameters
US6251757B1 (en) * 2000-02-24 2001-06-26 Advanced Micro Devices, Inc. Formation of highly activated shallow abrupt junction by thermal budget engineering
IT1316871B1 (en) * 2000-03-31 2003-05-12 St Microelectronics Srl MONOLITHICALLY INTEGRATED ELECTRONIC DEVICE AND RELATED MANUFACTURING PROCESS
US6630699B1 (en) * 2000-08-31 2003-10-07 Lucent Technologies, Inc. Transistor device having an isolation structure located under a source region, drain region and channel region and a method of manufacture thereof
DE10144268B4 (en) * 2001-09-08 2015-03-05 Robert Bosch Gmbh Device for measuring the strength of a vector component of a magnetic field
US6586297B1 (en) * 2002-06-01 2003-07-01 Newport Fab, Llc Method for integrating a metastable base into a high-performance HBT and related structure

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6475808B1 (en) * 1998-07-14 2002-11-05 Zyomyx, Incorporated Arrays of proteins and methods of use thereof

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020127577A1 (en) * 1999-05-28 2002-09-12 Jutta Eichler Bioactive sensors
US20050153357A1 (en) * 1999-05-28 2005-07-14 Jutta Eichler Bioactive sensors
WO2006002141A2 (en) * 2004-06-18 2006-01-05 Wisconsin Alumni Research Foundation Detection of post-translationally modified peptides with liquid crystals
WO2006002141A3 (en) * 2004-06-18 2006-04-06 Wisconsin Alumni Res Found Detection of post-translationally modified peptides with liquid crystals
US20080054256A1 (en) * 2006-09-05 2008-03-06 Hyoyoung Lee Molecular electronic device including organic dielectric thin film and method of fabricating the same
US7759677B2 (en) * 2006-09-05 2010-07-20 Electronics And Telecommunications Research Institute Molecular electronic device including organic dielectric thin film and method of fabricating the same

Also Published As

Publication number Publication date
WO2001092883A2 (en) 2001-12-06
DE10192267D2 (en) 2003-05-08
DE10027397A1 (en) 2001-12-13
US20040235146A9 (en) 2004-11-25
DE50113061D1 (en) 2007-11-08
ATE374368T1 (en) 2007-10-15
AU7232801A (en) 2001-12-11
JP2003536056A (en) 2003-12-02
US20080167196A1 (en) 2008-07-10
EP1287361B1 (en) 2007-09-26
CA2413855A1 (en) 2001-12-06
US7955638B2 (en) 2011-06-07
AU2001272328B2 (en) 2005-09-01
WO2001092883A3 (en) 2002-08-08
EP1287361A2 (en) 2003-03-05

Similar Documents

Publication Publication Date Title
US7955638B2 (en) Surface for the immobilization of ligands
Neumann et al. SPR-based fragment screening: advantages and applications
CA2370261A1 (en) Process to create biomolecule arrays on metal surfaces
US7951572B2 (en) Construction of gold nanoparticle-based peptide chip, and assaying enzyme activity and inhibitor effect using secondary ion mass spectrometric analysis thereof
US7002004B2 (en) Biomolecule and/or cellular arrays on metal surfaces
JP2005513503A (en) Immobilization of binding substances
CA2818483C (en) Method of and system for printing in-well calibration features
US6221674B1 (en) Process for the application of reagent spots
US20100086920A1 (en) Method to assess cancer susceptibility and differential diagnosis of metastases of unknown primary tumors
WO2002063299A1 (en) Low affinity screening method
US8158342B2 (en) Method for the identification of human immunodeficiency virus related antibodies in blood
Cheran et al. Protein microarray scanning in label-free format by Kelvin nanoprobe
US8158440B2 (en) Method for quantitative measurement of thyroid related antibodies or antigens in a serum sample
Gajos et al. A perspective on ToF-SIMS analysis of biosensor interfaces: Controlling and optimizing multi-molecular composition, immobilization through bioprinting, molecular orientation
Seidel et al. A miniaturized heterogeneous fluorescence immunoassay on gold-coated nano-titer plates
KR100869909B1 (en) Detection method for the reaction between biomolecules and protein using biochip based on the monolayer of gold nanoparticles
US20080058225A1 (en) Artificial receptors, building blocks, and methods
Grunwald A brief introduction to the streptavidin-biotin system and its usage in modern surface based assays
Senaratne et al. Molecular templates for bio-specific recognition by low-energy electron beam lithography
US20100047815A1 (en) Method to detect tumor markers and diagnosis of undifferentiated tumors
US20230417668A1 (en) Surface plasmon resonance signal amplification
US20110152119A1 (en) Quantification Method of Biochemical Substances Using Ion Scattering Spectroscopy and Specific-Binding Efficiency Quantification Method of Biochemical Substances Using Ion Scattering Spectroscopy
Prilutsky et al. Bioreceptor functionalization of gold-coated sensor surfaces
JP2006184004A (en) Cantilever sensor system
Mecklenburg et al. Microarray platform for omics analysis

Legal Events

Date Code Title Description
AS Assignment

Owner name: GRAFFINITY PHARMACEUTICALSD AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DICKOPF, STEFAN;OTTLEBEN, HOLGER;SCHMIDT, KRISTINA;AND OTHERS;REEL/FRAME:014322/0876;SIGNING DATES FROM 20030515 TO 20030606

Owner name: GRAFFINITY PHARMACEUTICALSD AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DICKOPF, STEFAN;OTTLEBEN, HOLGER;SCHMIDT, KRISTINA;AND OTHERS;SIGNING DATES FROM 20030515 TO 20030606;REEL/FRAME:014322/0876

AS Assignment

Owner name: GRAFFINITY PHARMACEUTICALS AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DICKOPF, STEFAN;OTTLEBEN, HOLGER;RAU, HARALD;AND OTHERS;REEL/FRAME:015084/0896;SIGNING DATES FROM 20030515 TO 20030606

Owner name: GRAFFINITY PHARMACEUTICALS AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DICKOPF, STEFAN;OTTLEBEN, HOLGER;RAU, HARALD;AND OTHERS;SIGNING DATES FROM 20030515 TO 20030606;REEL/FRAME:015084/0896

STCB Information on status: application discontinuation

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