WO1996040276A1 - Epreuves de selection de composes - Google Patents

Epreuves de selection de composes Download PDF

Info

Publication number
WO1996040276A1
WO1996040276A1 PCT/US1996/008332 US9608332W WO9640276A1 WO 1996040276 A1 WO1996040276 A1 WO 1996040276A1 US 9608332 W US9608332 W US 9608332W WO 9640276 A1 WO9640276 A1 WO 9640276A1
Authority
WO
WIPO (PCT)
Prior art keywords
assay
phosphotyrosine
antibody
protein substrate
substrate
Prior art date
Application number
PCT/US1996/008332
Other languages
English (en)
Inventor
Axel Ullrich
Harald App
Klaus P. Hirth
Jianming Tsai
Original Assignee
Sugen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sugen, Inc. filed Critical Sugen, Inc.
Priority to AU59658/96A priority Critical patent/AU5965896A/en
Priority to EP96916950A priority patent/EP0873142A4/fr
Publication of WO1996040276A1 publication Critical patent/WO1996040276A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/42Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving phosphatase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6806Determination of free amino acids
    • G01N33/6812Assays for specific amino acids

Definitions

  • the present application is a continuation-in-part of copending application Serial No. 08/279,321, filed July 22, 1994.
  • the present invention relates to rapid, quantitative, specific, high through-put assay systems for screening test compounds for their ability to modulate tyrosine kinase or phosphotyrosine phosphatase activity within the cell or in a cell-free system.
  • Protein phosphorylation is a common regulatory mechanism used by cells to selectively modify proteins carrying regulatory signals from outside the cell to the nucleus.
  • the proteins that execute these biochemical modifications are a group of enzymes known as protein kinases. They may further be defined by the amino acid that they target for phosphorylation.
  • protein kinases One group of protein kinases are the tyrosine kinases (TKs) which selectively phosphorylate a target protein on its tyrosine residues.
  • TKs tyrosine kinases
  • Some tyrosine kinases are transmembrane receptors (RTKs) , and, upon activation by a ligand, can autophosphorylate as well as modify substrates.
  • the initiation of sequential phosphorylation by ligand stimulation is a paradigm that underlies the action of such effectors as, for example, epidermal growth factor (EGF) , insulin, platelet-derived growth factor (PDGF) , and fibroblast growth factor (FGF) .
  • the receptors for these ligands are tyrosine kinases and provide the interface between the binding of a ligand (hormone, growth factor) to a target cell and the transmission of a signal into the cell by the activation of one or more biochemical pathways.
  • Ligand binding to a receptor tyrosine kinase activates its intrinsic enzymatic activity. (Ullrich and Schlessinger, Cell 61:203-212, 1990) .
  • Tyrosine kinases can
  • LE26 also be cytoplasmic, non-receptor-type enzymes and act as a downstream component of a signal transduction pathway.
  • Phosphotyrosine phosphatases specifically remove the phosphates from tyrosine residues of modified proteins, although some may also have enzymatic activity for other phosphoamino acids.
  • these phosphatases may be transmembrane molecules or they may be localized in intracellular compartments (Walton et al . , 1993, Ann. Rev. Biochem. 62:101-120) . These proteins also play an integral role in the signal transduction pathways of the cell.
  • the secondary signal transduction molecules activated by activated receptors result in a signal cascade that regulates cell functions such as cell division, differentiation and survival.
  • These molecules can be cytoplasmic enzymes such as, for example, kinases and phosphatases or can be non-catalytic adapter molecules such as, for example, the Grbs (Growth factor Receptor Bound) (Skolnik, et al . , 1991, Cell 65:83-90) .
  • Adapter proteins have in common one or two copies of an approximately 100 amino acid long motif called an SH2 (Src homology 2) domain due to its similarity to a motif originally identified in the c-Src cytoplasmic TKs.
  • SH2-containing polypeptides may otherwise be structurally and functionally distinct from one another (Koch, CA. et al. 1991, Science 252:66-674) .
  • SH2 domains directly recognize specific phosphorylated tyrosine residues. There is evidence that the amino acid sequences flanking the phosphorylated tyrosine confers a certain specificity such that a particular SH2 domain will bind preferentially to particular sequences (Koch, C.A., et al. 1991, Science 252:668-674; Cantley, L.C., et al. 1991, Cell 64:281-302) .
  • SH3 domains proteins with SH3 domains bind to proteins with proline rich regions, such as, for example, —PPPLPP—.
  • a third domain known as the pleckstrin-homology or PH domain has also been identified in many adapter type proteins (Musacchio et al . , 1993, TIBS 18:342-348) .
  • tyrosine kinases and phosphotyrosine phosphatases have made them attractive targets for the development of new therapeutic molecules. It is known, for example, that the overexpression of tyrosine kinases, such as HER2, can play a decisive role in the development of cancer (Slamon, D.J., et al. , 1987, Science, 235:177-182) and that antibodies capable of blocking the activity of this enzyme can abrogate tumor growth. (Drebin, et al. 1988, Oncogene 2:387-394) .
  • Assays currently used for screening drugs/ligands that act on cells containing TKs involve exposing cells that express the TK to a test substance and either: (a) scoring phenotypic changes in the cell culture as compared to control cells that were not exposed to the test substance; or (b) biochemically analyzing cell lysates to assess the level and/or identity of tyrosine phosphorylated proteins.
  • This latter approach is illustrated by several methodologies.
  • a common technique involves incubating cells with ligand and radiolabeled phosphate, lysing the cells, separating cellular protein components of the lysate using an
  • SHEET ROLE 26 SDS-polyacrylamide gel (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of phosphorylated proteins by exposing X-ray film.
  • the phosphorylated proteins are detected by immunoblotting 5 techniques, in which case the phosphate that is detected is not radiolabeled. Instead, the cellular components separated by SDS-PAGE are transferred to a nitrocellulose membrane, where the presence of phosphorylated tyrosines is detected using an antiphosphotyrosine antibody (anti-PY) .
  • anti-PY antiphosphotyrosine antibody
  • the anti-PY _0 can be detected by labeling it with a radioactive substance, which then requires scanning the labeled nitrocellulose with a piece of specialized equipment designed to detect radioactivity or exposure of X-ray film.
  • the anti-PY can be conjugated with an enzyme, such as horseradish 5 peroxidase, and detected by subsequent addition of a colorimetric substrate for the enzyme.
  • a further alternative involves detecting the anti-PY by reacting with a second antibody which recognizes the anti-PY, this second antibody being labeled with either a radioactive moiety or an enzyme as 0 previously described. Examples of these and similar techniques are described in Hansen et al. , 1993, Electrophoresis 14:112-126; Campbell et al. 1993, J.
  • ELISA-type assays in microtitre plates have been used to test purified substrates. See for example Peraldi et al., 5 1992, J. Biochem. 285: 71-78; Schraag et al. , 1993, Analytical Biochemistry 211:233-239; Cleavland, 1990, Analytical Biochemistry 190:249-253; Farley, 1992, Analytical Biochemistry 203:151-157; and L ⁇ zaro, 1991, Analytical Biochemistry 192:257-261.
  • these techniques involve assaying purified components, and thus are unsuitable for evaluating the effects of a test substance on phosphorylation within the normal cellular context. Purified components may be difficult or expensive to obtain in sufficient quantities or may require significant experimental work to develop a purification process for the desired component.
  • the present invention relates to rapid, quantitative, specific, high through-put assays for screening test compounds such as drugs, ligands (natural or synthetic) , ligand antagonists, peptides, or small organic molecules, for their ability to modulate tyrosine kinase or phosphotyrosine phosphatase activities involved in signal transduction.
  • test compounds such as drugs, ligands (natural or synthetic) , ligand antagonists, peptides, or small organic molecules, for their ability to modulate tyrosine kinase or phosphotyrosine phosphatase activities involved in signal transduction.
  • a target cell that expresses a particular substrate that is phosphorylated or dephosphorylated on tyrosine residues during signal transduction is used.
  • the tyrosine kinase is capable of phosphorylating its own specific tyrosine residues, in which case the kinase enzyme is also the substrate.
  • the target cell is exposed to a test substance, and thereafter lysed to release cellular contents, including the protein substrate.
  • the substrate is isolated by contacting the cell lysate with a substrate-specific antibody immobilized directly or indirectly onto a solid support and subsequently washing away the majority of other cellular components, some of which may also be phosphorylated.
  • An assay is performed on the isolated substrate to detect the presence or absence of phosphotyrosine residues on the
  • BlE26 substrate compared to lysates of control target cells which were not exposed to the test substance.
  • the target cells are exposed to the test substance in the absence of ligand and the results compared to negative controls that were not exposed to either the ligand or the test substance, and to positive controls that were exposed to ligand only.
  • the target cells are exposed to the test compound in the presence of the ligand and results are compared to those of controls where the cells are exposed only to ligand.
  • the effect of test substances on the ability of the tyrosine kinases and phosphotyrosine phosphatases to phosphorylate and dephosphorylate, respectively, tyrosine residues of a particular substrate can be tested in a cell free system.
  • an appropriate reaction mixture is prepared containing the substrate and kinase (these are one and the same in the case when autophosphorylation is measured) or the phosphorylated substrate and phosphatase, which can also be one and the same.
  • the kinase reaction is initiated in the presence of ATP and divalent cations.
  • An immunoassay is performed on the reaction product to detect the presence or absence of phosphotyrosine residues on the substrate, and results are compared to those obtained for controls.
  • Detection of the presence or absence of phosphotyrosine residues on a target substrate is performed in one of several ways.
  • One method utilizes a signal-generating anti-phosphotyrosine antibody to detect phosphotyrosine residues on the immobilized substrate thereby measuring the degree of phosphorylation of the substrate.
  • detection of the presence or absence of phosphotyrosine is accomplished by the binding of a specific SH2 domain or SH2 domain containing protein to the immobilized substrate. Said SH2 domain or SH2 domain containing protein will be relatively specific for a particular phosphorylated
  • SH2 domain containing proteins are, for example but not limited to, Grb molecules such as Grb2 (Margolis, B., et al. 1989, Cell 51:1101-1107; Bjoige, et al. 1990, Proc. Nat. Acad. Sci. USA 87:3816-3820; Pelicci, et al . 1992, Cell 70:93-104; Fu & Zhang, 1993, Cell 74:1135-1145) .
  • Grb molecules such as Grb2 (Margolis, B., et al. 1989, Cell 51:1101-1107; Bjoige, et al. 1990, Proc. Nat. Acad. Sci. USA 87:3816-3820; Pelicci, et al . 1992, Cell 70:93-104; Fu & Zhang, 1993, Cell 74:1135-1145) .
  • the degree of binding of the SH2 domain or SH2 domain containing protein can be accomplished with a signal-generating anti-SH2 domain or anti-SH2 domain containing protein antibody or by, for example, using an SH2 domain or SH2 domain containing protein fused to a signal-generating protein, such as, for example GST (glutathione-s-transferase, Tsang, V.C.W., et al. Methods in Enzymology 92:377, 1983) .
  • a signal-generating protein such as, for example GST (glutathione-s-transferase, Tsang, V.C.W., et al. Methods in Enzymology 92:377, 1983) .
  • the invention is demonstrated by way of working examples using a whole cell assay to screen for inhibitors of kinase activities of EGF-receptor and HER-2, and inhibitors of phosphatase activity that dephosphorylates insulin receptor (Section 6, infra) ; a cell-free assay to screen for inhibitors of the EGF-receptor (Section 7, infra) , and a cell-free assay to screen for inhibitors of phosphatase IB (Section 8, infra) .
  • Target Cell a cell which expresses a tyrosine kinase or phosphotyrosine phosphatase of interest, and which further contains a substrate which can be phosphorylated or dephosphorylated as a result of signal transduction.
  • the tyrosine kinase or phosphatase may be naturally expressed by the target cell or engineered into the target cell using recombinant DNA techniques well known in the art.
  • Test Substance a chemically defined compound or mixture of compounds (as in the case of a natural extract or tissue culture supernatant) whose effect on the phosphorylation of/by the tyrosine kinases or the dephosphorylation by the tyrosine phosphatases of a target cell is determined by the assay of the invention.
  • Substrate a protein which is acted on by tyrosine kinase or tyrosine phosphatases such that it is either phosphorylated or dephosphorylated on tyrosine residues. Tyrosine kinases and phosphatases can act as both an enzyme and a substrate.
  • Anchoring Molecule an antibody or other protein which binds specifically with high affinity to a substrate of a tyrosine kinase or phosphatase and which is immobilized onto a solid phase.
  • the anchoring molecule can be bound directly or indirectly to the solid phase. Indirect binding can be accomplished, for example, by first coating the solid phase with an antibody which binds specifically to the anchoring molecule and subsequently adding the anchoring molecule.
  • Detection Molecule an antibody or other protein or amino acid sequence that specifically binds to phosphorylated tyrosine residues, or to a particular phosphorylated tyrosine residue, and which is used in the assay to detect phosphorylated tyrosines on the substrate bound by the anchoring molecule.
  • the detection molecule may be used as an anchoring molecule, in which case the anchoring molecule as defined above can be used to detect the level of bound substrate.
  • An example of a protein other than an antibody that specifically binds to phosphorylated tyrosines is a SH2 domain or a protein containing an SH2 domain.
  • the present invention relates to methods for screening compounds that modulate tyrosine kinase or phosphotyrosine phosphatase activities involved in signal transduction.
  • the assays of the invention involve monitoring the phosphorylation or dephosphorylation of tyrosine residues on selected substrates involved in signal transduction in a target cell and can be practiced in a whole cell or a cell-free system.
  • Test substances which mimic, enhance or inhibit the signal transduction which is the result of ligand binding to the receptor may be readily identified using the assays of the invention.
  • the test substances include but are not limited to
  • the present invention provides a method for determining the effect of a test substance on specific tyrosine kinases or phosphatases or their substrates in a target cell.
  • target cells that express a protein substrate that is phosphorylated or dephosphorylated on a tyrosine residue during signal transduction are exposed to a test substance and, thereafter, lysed to release cellular contents, including the substrate of interest.
  • mimics of the natural ligand for a signal transducing receptor are to be screened, the target cells are exposed to the test substance and compared to positive controls which are exposed only to the natural ligand and to negative controls which were not exposed to either the test substance or the ligand.
  • inhibitors or enhancers of ligand-induced signal transduction are to be screened, the target cells are exposed to the natural ligand in the presence of the test substance and compared to controls which are not exposed to the test substance.
  • the substrate of interest is isolated by incubating the cell lysate with a substrate- specific anchoring molecule bound to a solid support and thereafter washing away non-bound cellular components.
  • a detection procedure is performed to assess the presence or absence of phosphotyrosine residues on the substrate as compared to lysates of control cells which were not exposed to the test substance.
  • This assay offers several advantages.
  • the exposure of the test substance to a whole cell allows for the evaluation of its activity in the natural context in which inhibitors or enhancers of tyrosine kinases and phosphotyrosine phosphatases may act.
  • this assay is performed in whole cells and is based on the use of a known phosphorylated substrate, it does not require redesign or the use of reagents specific for the particular kinase or phosphatase responsible for
  • the assay can detect the effects of the test compounds on cellular kinase or phosphatase activities even if the direct target of the compound is unknown. Because this assay, and those described below, can readily be performed in a microtiter plate format, the assays described can be performed by an automated robotic system, allowing for testing of large numbers of test samples within a reasonably short time frame.
  • the anchoring molecule should be highly specific for the substrate of interest . It is preferred that the anchoring molecule be an antibody, hereinafter referred to as an anchoring antibody.
  • the anchoring antibody is preferably generated against a unique epitope of the substrate, most preferably an epitope wherein binding by the anchoring antibody will not interfere with binding of the detection antibody or SH2 domain or ligand binding.
  • the anchoring antibody may be a monoclonal antibody but can also be a polyclonal antibody.
  • the detection procedure used to assess the phosphorylation state of the substrate may employ an anti-phosphotyrosine antibody or a peptide that recognizes and binds to phosphorylated tyrosines.
  • Said amino acid sequence may be, for example, an SH2 domain and may furthermore be an isolated SH2 domain or an intact protein containing an SH2 domain.
  • the detection antibody is preferably a polyclonal antibody to maximize the signal but may also be specific monoclonal antibodies which have been optimized for signal generation.
  • An alternate embodiment of the invention relates to methods for determining the effect of test compounds on the ability of tyrosine kinases to autophosphorylate or phosphorylate the substrate of interest in a cell-free system, or the ability of tyrosine phosphatases to dephosphorylate such substrates in a cell-free system.
  • the test substance is added to a reaction
  • kinase reaction is initiated by the addition of ATP.
  • a detection procedure as described above is performed on the substrate to assess the presence or absence of the phosphorylated tyrosine residues, and results are compared to those obtained for controls i.e., reaction mixtures to which the test substance was not added.
  • a similar procedure is used to assess autodephosphorylation by phosphotyrosine phosphatases.
  • a further aspect of this embodiment of the invention allows the user to distinguish test substances that inhibit the interaction between the kinase and its substrate from test substances that inhibit the interaction between the kinase and ATP.
  • Test substances that inhibit the interaction between the kinase and its substrate may be more target specific than those that inhibit the interaction between the kinase and ATP.
  • the assays of the invention can be used as a primary screen to assess the activity of a previously untested compound or extract, in which case a single concentration is tested and compared to controls.
  • This assay can also be used to assess the relative potency of a compound by testing a range of concentrations, in a range of 100 ⁇ M to 1 pM, for example, and computing the concentration at which the amount of phosphorylation is reduced by one-half (IC50) compared to controls in the case of inhibitors or increased by one-half relative to the natural ligand in the case of enhancers.
  • assays can be used to identify compounds which modulate protein kinase or protein phosphatase activity. It is further contemplated that the assays of the invention can be used to diagnose clinical conditions in which the effect of a known compound on protein kinases or protein phosphatases is altered by reference to a suitable control experiment.
  • the identification of compounds that modulate protein kinase or protein phosphatase activity can have utility in the selection of compounds to treat neoplastic disorders in which cell proliferation has been shown to correlate with an increase or decrease in kinase or phosphatase activity.
  • a protein kinase such as HER2
  • the identification of a compound that can inhibit protein kinase activity may restore normal growth patterns and reverse oncogenicity.
  • the identification of a compound that stimulates protein phosphatase activity may restore normal growth and reverse oncogenicity.
  • the compounds identified in the assays of the invention may be used as therapeutic agents against any diseases in which modulation of tyrosine kinase or tyrosine phosphatase activity reverses a pathological process in cells, tissues or organs.
  • the assays described herein can be used to determine if a cell line or tumor in which aberrant kinase or phosphatase activity contributes to a pathological process or disease state will respond to a compound that modulates kinase or phosphatase activity and therefore reverse the pathological process or disease state.
  • therapeutic regimens may be devised using the assays of the invention on cells derived from patients, avoiding the necessity to subject the patient to trial and error.
  • these assays may be used to identify the substrates involved in the signal transduction pathway responsible for the phenotype.
  • the specific kinases or phosphatases operating in the signal pathway that causes a particular phenotype may be identified through an analysis of these substrates.
  • the whole cell assay of the invention described herein can be performed, for example, by utilizing pre-packaged kits comprising any or all of the reagents of the assay, such as a solid phase coated with an anchoring molecule to a substrate
  • the cell-free assays of the invention may be performed, for example, by utilizing pre ⁇ packaged kits comprising any or all of the reagents of the assay, such as an enzyme, substrate, anchoring molecule, a solid phase coated with an anchoring molecule to a substrate of interest, or a detection molecule.
  • the anchoring antibody should have a high binding affinity for and be highly specific for the target substrate.
  • the anchoring antibody may be a monoclonal antibody or a polyclonal antibody (see below) .
  • Monoclonal antibodies or polyclonal antibodies selective for the substrate are selected by techniques well known in the art. I munoblots can be performed using lysates from cells that express the target substrate to determine specificity. The preferred antibody will only detect the substrate, preferably where greater than 100,000 molecules per cell.
  • An alternative method for determining specificity is immunoprecipitation.
  • the binding affinity of the monoclonal antibody or polyclonal antibody for the substrate can be determined by the relative strength of the signal generated in the immunoblot or by other techniques well known in the art.
  • a known number of cells expressing the substrate is lysed and serial dilutions of the lysate are applied to wells in a 96 well microtiter plate that have been precoated with the anchoring antibody. After allowing the substrate to bind to the antibody, the unbound material is washed away and the amount of bound substrate is determined using known immunoassay techniques. In order to have the proper signal to noise ratio one must be able to detect the target molecule in approximately 1 x 10 4 cell equivalents per well. The maximum number of cells allowable per well is generally ⁇ 1 x 10 5 due to space constraints although this number may be somewhat larger or smaller depending on the cell type.
  • the preferred amount of anchoring antibody and the preferred number of cells to be seeded in each well can be determined empirically by varying the amount of anchoring antibody used to coat the well and the amount of cell lysate (whole cell equivalents) added per well. The ideal concentration of these two components will produce a signal which will reflect a specific phosphotyrosine content in a linear part of the curve.
  • the extracellular domain which bears unique epitopes that may be involved in ligand recognition and binding, may advantageously be used as the immunogen.
  • the extracellular domain which bears unique epitopes that may be involved in ligand recognition and binding, may advantageously be used as the immunogen.
  • variable regions of the protein may be used as immunogens.
  • Anti-phosphotyrosine antibodies may be generated using phosphotyramine epitopes as described in the examples infra. (Fendly et al. , 1990, Cancer Research 50:1550-1558)
  • the antibodies used in the immunoassay include but are not limited to polyclonal, monoclonal, chimeric, single chain, Fab fragments and an Fab expression library.
  • various host animals including but not limited to rabbits, mice, rats, etc., may be immunized by injection with the particular antigen in a suitable adjuvant or by injecting the epitope conjugated to an immunogenic carrier.
  • conjugates are prepared so that the most unique epitope of the substrate will be most accessible to antibody.
  • the substrate of interest is a cell-surface receptor, whole cells expressing said receptor may be used as the antigen.
  • adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to Freund' s (complete and incomplete) , mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Corynebacterium parvum bacille Calmette-Guerin
  • Monoclonal antibodies may be prepared by using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Koehler and Milstein, (Nature, 1975,
  • Human antibodies may be used and can be obtained by using human hybridomas (Cote at al., 1983, Proc. Natl. Acad. Sci. 80:2026-2030) or by transforming human B cells with EBV virus in vitro (Cole et al. , 1985, in, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, pp. 77-96) .
  • Antibody fragments which contain binding sites specific for the substrate may be generated by known techniques.
  • fragments include but are not limited to: the F(ab')2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • the antibodies may be stored and purified using methods which are well known to those skilled in the art (e.g., see “Antibodies, A Laboratory Manual, eds. Harlow & Lane, Cold Spring Harbor Laboratory, 1988, Ch. 8) .
  • polyclonal or monoclonal antibody specific for the substrate of interest may be obtained from commercial sources.
  • the phosphotyrosine specific proteins or peptides used for the detection procedures are produced by methods well known to those skilled in the art. They may be purified from cells that natively express the protein, or may be produced by recombinant means and purified from genetically engineered cells. A variety of host-expression vector systems may be
  • microorganisms such as bacteria (e.g., E.coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing PTK or adaptor protein coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing the protein or peptide coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the protein or peptide coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing the protein or peptide coding sequence; or mammalian cell systems (
  • COS COS, CHO, BHK, 293, 3T3 harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter) .
  • promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter) .
  • expression vectors include but are not limited to the E. coli expression vector pUR278 (Ruther et al . , 1983, EMBO J. 2:1791) , in which the protein or peptide coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, 1985, Nucleic Acids Res. 13:3101-3109; Van Heeke & Schuster, 1989, J. Biol. Chem. 264:5503-5509); and the like.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST) .
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free reduced glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned protein or peptide can be released from the GST moiety.
  • the fusion protein can be left intact. In the case where the
  • non-phosphotyrosine binding portion of the fusion protein is an enzyme, said fusion protein can be reacted with a colorimetric substrate as described below.
  • the non-phosphotyrosine binding portion of the fusion protein can remain uncleaved so that it may be detected by an antibody specific to it.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spod ⁇ ptera frugiperda cells.
  • the protein or peptide coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter) .
  • Successful insertion of the coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene) .
  • a number of viral based expression systems may be utilized.
  • the protein or peptide coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing proteins or peptides in infected hosts (e.g., See Logan & Shenk, 1984, Proc.
  • Specific initiation signals may also be required for efficient translation of inserted coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an protein gene, including its own initiation
  • 26 codon and adjacent sequences is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of the coding sequence is inserted, exogenous translational control signals, including the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al. , 1987, Methods in Enzymol. 153:516-544) . The proteins or peptides may be produced synthetically. See, for example, Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman and Co., NY, which is incorporated herein, by reference, in its entirety.
  • Solid phases used for the immobilization of a substrate may be prepared by coating with the anchoring molecule specific for the substrate.
  • the anchoring molecule may be directly immobilized onto the solid phase, or, alternatively, may be indirectly immobilized to the solid phase by an antibody bound to the solid phase which is specific for the anchoring molecule.
  • the solid phase may first be coated with an anti-Ig that binds to the polyclonal antibody and indirectly immobilizes it to the solid phase.
  • the solid phase may comprise a microtiter plate, a stick, tube, disc, fiber or the like, or a microtiter plate.
  • a preferred solid phase is a 96 well microtiter plate such as those available from Corning, Cynatech and Nunc. Particularly preferred 96 well plates are the Corning, Nunc MaxiSop and Dynatech Immulon I and IV. Ideal conditions for maximum coating with the anchoring antibody can vary with pH, ionic
  • the anchoring molecule may be attached to the solid phase by any of a variety of methods known to those skilled in the art, including but not limited to non-covalent and covalent attachments.
  • the anchoring molecule can be applied to the solid phase in buffer for a specified period of time to allow the anchoring molecule to absorb to the solid phases, at which time the buffer is removed and blocking buffer is added.
  • Blocking buffer may be dry milk, gelatin, bovine serum albumin or ethanolamine as a 1 - 5 % solution in a neutral pH Tris-HCl buffer. Following the removal of the blocking buffer, the solid phase is washed and ready for use. In the case of indirect immobilization, the substrate specific antibody is added after blocking.
  • the anchoring molecule and the coated solid phase may be prepared ahead of time and stored until required for use in the assay.
  • the detection molecules are modified by the addition of a signal-generating system to allow for detection of the detection molecules bound to phosphotyrosines in the assay system.
  • the detection molecules may be labeled directly or can be detected using a secondary reagent that will detect the detection molecule.
  • signal generating systems include, but are not limited to, enzyme-linked systems (such as horseradish peroxidase or alkaline phosphatase) , radiolabels, fluorescent labels, light-emitting labels, light-absorbing labels, or dyes (e.g., See "Antibodies, A Laboratory Manual, eds. Harlow & Lane, Cold Spring Harbor Laboratory 1988, Ch. 9) .
  • an appropriate substrate such as a colorimetric substrate
  • Specific substrates used for detection include ABTS (horseradish peroxidase) , DAB, AEC, BCIP/NT (alkaline
  • an anti-rabbit biotinylated IgG peroxidase conjugate is incubated with the substrate ABTS in order to facilitate detection of the bound antibody. After the reaction is stopped, the product of this reaction can be detected by determination of the O.D. at 410 nm. (Background signal measured at 630 nm is subtracted.)
  • the target cells used in the present invention may express a tyrosine kinase that itself is a substrate for phosphorylation, or that phosphorylates other protein substrates. These cells may have a tyrosine kinase that is natively expressed or they may be genetically engineered to express a specific tyrosine kinase.
  • the kinase may be a receptor tyrosine kinase that is responsive to an exogenous ligand, or the tyrosine kinase may be capable of self- phosphorylation even in the absence of ligand binding.
  • the tyrosine kinase may be a cellular kinase that is not membrane-bound, but is activated in a signal transduction pathway.
  • the target cells of the present invention may also natively express a phosphotyrosine phosphatase (PTP) , or they may be genetically engineered to express a PTP.
  • PTP may be a receptor phosphotyrosine phosphatase or a cytoplasmic enzyme.
  • These target cells may have a PTP that is constitutively active in regulating the level of phosphorylation on substrate proteins that are capable of self-phosphorylation or are phosphorylated by a basal kinase activity, in the absence of ligand binding.
  • the test substances can be any of a variety of substances including but not limited to hormones that interact with a membrane-bound tyrosine kinase receptor, or drugs that exert their effect on the target cells through the modulation of intracellular tyrosine kinases or phosphatases.
  • the test compounds may exert their effect through interference with ligand induced activation of a membrane-bound receptor.
  • the test substances may be molecules that are the physiologic ligands for the receptor. They may be molecules that are not the physiologic ligands for the receptor and can be tested for an effect on the enzymatic activity of a specific receptor tyrosine kinase or phosphatase.
  • the compounds may be drugs that modulate a receptor tyrosine kinase or phosphatase in the absence of ligand. They may be drugs that inhibit the ligand-dependent activation of a receptor tyrosine kinase or phosphatase.
  • the present invention provides a method for the determination of the effect of a drug on specific tyrosine kinases or phosphatases by analysis of their substrates obtained from the target cell.
  • the target cells are grown using standard protocols for tissue culture maintenance, in which the cells are fed with appropriate medium, and incubated at the appropriate temperature with C0 2 , if required.
  • the substance to be tested is added to the growth medium, and the conditions of incubation with the cells are dictated by the particular assay. If a test substance is to be tested for its effect on ligand-dependent modulation of a receptor, the ligand which activates the receptor is added to the target cell in the presence of the test compound.
  • a cell lysate is prepared for analysis by immunoassay of the phosphorylated substrate.
  • Cell lysates may be prepared by known techniques in the art in which the cell membrane is solubilized by the addition of a detergent, and the intracellular contents stabilized with the addition of buffers, protease inhibitors and phosphatase inhibitors.
  • Detergents include but are not limited to Triton X-100, Tween 20, NP-40, or SDS.
  • Protease inhibitors include but are not limited to PMSF, leupeptin, EDTA and aprotinin.
  • Phosphatase inhibitors include but are not limited to sodium orthovanadate sodium pyrophosphate and EDTA. 5.7. EXPOSURE OF SUBSTRATE TO TEST
  • the effect of the test substances on the phosphorylation of the substrate of interest can be assessed in a cell-free reaction mixture.
  • the test substance can be incubated with the substrate of interest in a reaction mixture containing the kinase (where autophosphorylation is to be assessed, the substrate and the kinase are one and the same) .
  • the kinase reaction is then initiated by the addition of ATP and appropriate cations.
  • the substrate is then immunoassayed for the presence of phosphotyrosine residues using an anti-phosphotyrosine detection antibody or a phosphotyrosine binding protein or peptide such as one containing an SH2 domain.
  • test compound is first incubated with a phosphorylated substrate of interest, which can be the PTP itself, in a reaction mixture to which the phosphatase is added.
  • substrate is then immunoassayed for the presence of phosphotyrosine residues using an antiphosphotyrosine detection antibody or a phosphotyrosine binding protein or peptide such as one containing an SH2 domain.
  • the effect of the test substance on the phosphatase activity is reflected by the degree of dephosphorylation detected in the samples treated with the test substance as compared to untreated controls.
  • the phosphorylated substrate used in the assay can be obtained from cell lysates of target cells which were activated by their natural ligand. Alternatively, the substrate can be obtained from unactivated cell lysates and phosphorylated in vitro in a reaction mixture containing the
  • reaction product can be detected by any of the immunoassays described herein, a particularly rapid approach is described in the working examples, infra.
  • the substrate of interest can be immobilized using an anchoring antibody prior to conducting the kinase and/or phosphatase reactions in the presence of the test substances, so that all reactions are carried out on the solid phase.
  • EXAMPLE WHOLE CELL SCREENING ASSAYS
  • the whole cell assays assess the potential of an exogenous test substance, applied to a target cell expressing a tyrosine kinase or tyrosine phosphatase, to modulate the activity of that enzyme.
  • conditions are described for assays assessing the ability of test compounds to inhibit the kinase activity of EGFR or HER2, and to inhibit the phosphatase activity that dephosphorylate the phosphotyrosine residues on insulin receptor (IR) .
  • IR insulin receptor
  • PDGFR Platelet-Derived Growth Factor receptor
  • EGFR exogenous ligand is added to stimulate the kinase activity of the receptor.
  • HER2 kinase activity is stimulated without the addition of exogenous ligand.
  • IR no insulin is added because the receptor is capable of self- phosphorylation even in the absence of ligand binding.
  • cell lysates were prepared and added to microtiter plates coated with anti-target antibody. Phosphorylation of the immobilized target was detected using anti-phosphotyrosine antisera.
  • the cell line used for the EGFR assay was NIH3T3 clone 7 (EGFR/c7, Honegger, et al. Cell 51:199-209, 1987) engineered to over-express human EGFR. Growth media for these cells is DMEM (Gibco) containing 10% calf serum 1% L-glutamine and 20 mM Hepes.
  • the cell line used for the HER2 assay was BT 474 (ATCC HTB20) which over-expresses HER2. Growth media for these cells is RPMI (Gibco) plus 10% fetal calf serum plus GMS-G (Gibco supplement) plus glutamine.
  • the anchoring antibody used for the EGFR assay was a monoclonal antibody that recognizes the EGFR extracellular domain purchased from UBI (catalogue No. 05-101) .
  • An antibody recognizing the extracellular domain of HER2 was prepared, using techniques well know in the art, by immunizing mice with NIH3T3 cells engineered to over-express HER2. Hybridoma supernatants were screened for binding to HER2 expressing whole cells, and one clone (SUM02) was selected and grown in DMEM (GIBCO) + 1% calf serum.
  • the antibody was purified by Protein A agarose chromatography using a citric acid elution buffer, after which it was immediately neutralized.
  • EGF was purchase from Toyobo, Co. Ltd., Japan (EGF 201), and kept as a stock solution of 16.5 ⁇ M.
  • Anti-PY Rabbit polyclonal antiphosphotyrosine antibody
  • Anti-PY Rabbit polyclonal antiphosphotyrosine antibody
  • Goat anti-rabbit IgG horseradish peroxidase conjugate (Tago, Burlingame, CA, Cat.No. 4520 or Biosource Int., Camarillo, CA) was used as the detection antibody.
  • TBST buffer 50 mM Tris-HCl (pH 7.2) , 150 mM NaCl, 0.1% Triton X-100. 7.
  • Blocking buffer TBST plus 5% milk (Carnation instant non-fat dry milk) 8.
  • 5X HNTG buffer 100 mM HEPES, 750 mM NaCl, 50% glycerol, 1% Triton X-100, pH 7.2
  • ABTS solution 100 mM citric acid, 250 mM Na2HP04, 0.5 mg/ml ABTS (2, 2' -azinobis (3-ethylbenzthiazlinesulfonic acid) , pH 4.0.
  • Cell lysis buffer For 10 mis - 2 ml 5X HNTG, 0.1 ml of 0.5 M EDTA-HCl (pH 7.0), 0.1 ml of 0.5 M Na3V04 (kept as a 100X stock at 80 C in aliquots) , 1 ml of 0.2 M NaP207, and 7.3 mis distilled water. 11. Hydrogen peroxide: 30% solution.
  • Test substances were prepared according to Ohmichi, et al., 1993, Biochemistry 32 (17) :4650-4658; and Gazit, A., et al., 1991, J. Med. Chem. 34 (6) :1896-1907.
  • Microtiter plates (96 well) were coated with anchoring antibody at 0.5 ⁇ g per well in PBS (GIBCO), 150 ⁇ l final volume/well, covered with parafilm, and stored overnight at 4°C. Coated plates are good for up to 10 days when stored at 4°C. Before using, the coating buffer was removed and replaced with blocking buffer (200 ⁇ l) , then incubated, shaking, at room temperature for 30 minutes. Blocking buffer was removed and the plate washed 4 times with TBST buffer.
  • ASSAY PROCEDURE Media in the wells was replaced by serum free growth medium (DMEM or RPMI), 90 ⁇ l per well.
  • Serial dilutions of test compound stocks (10 mg/ml in DMSO) were diluted 1:10 into growth media (DMEM or RPMI) and 10 ⁇ l added per well for a final concentration range of 100 ⁇ M to 1 nM.
  • DMEM or RPMI growth media
  • 10 mg/ml test compound stock (in DMSO) is diluted 1:10 into serum free growth medium for final concentrations of 1:200 for the test substance and 0.5% for DMSO in the well.
  • Control wells received DMSO and serum free medium only. The cells were incubated for 1 hour at 37 C, 5% C0 2 .
  • EGF was diluted in DMEM such that upon transfer of 10 ⁇ l of dilute EGF (1:12 dilution, a 25 nM concentration is attained in the microtitre well . After the 1 hour incubation with the test drug, 10 ⁇ l of EGF was added per well. Control wells received DMEM alone. The plate was incubated, shaking, at room temperature an additional 5 minutes.
  • Phosphotyrosine was detected by the addition of anti-PY (100 ⁇ l per well, diluted 1:3,000 with TBST), then incubated, shaking, at room temperature for 30 minutes. The anti-PY solution was removed, and the plate washed 4 times with TBST. Detecting antibody was added (100 ⁇ l per well, diluted 1:3,000 with TBST) , and the plate was incubated for 30 minutes at room temperature, shaking. The detecting antibody solution was removed, the plates were washed with TBST (4x) and fresh
  • SUBSnTUTE SHEET (RULE 26) ABTS/H 2 0 2 was added (100 ⁇ l per well) to start color development. (ABTS/H 2 0 2 is prepared with 1.2 ⁇ l H 2 0 2 to 10 ml ABTS.) Color was allowed to develop for 20 minutes at room temperature. Color development may be stopped by the optional step of adding 50 ⁇ l 5N H 2 S0 4 . Optical density is measured at 410 nM (Dynatec MR5000) .
  • the cell line used for the IR assay was NIH3T3 (ATCC# CRL 1658) engineered to over-express the human IR (H25 cells) . Growth media for these cells is DMEM (Gibco) containing 10% fetal bovine serum, 1% L-glutamine and 20 mM
  • the anchoring antibody used was BBE which recognizes the extracellular domain of human IR, and was purified by the Enzymology Laboratory, Sugen Inc.
  • PBS Gibco
  • Anti-pTyr Rabbit polyclonal antiphosphotyrosine antibody
  • Goat anti-rabbit IgG POD conjugate (Tago, Burlingame, CA, Cat.No. 6430) was used as the detection antibody.
  • TBST buffer 50 mM Tris-HCl, 150 mM NaCl, 0.1% Triton X-100, adjusted to pH7.2 with ION HCl.
  • Blocking buffer PBS plus 5% milk (Carnation instant non-fat dry milk) .
  • 5X HNTG buffer 100 mM HEPES, 750 mM NaCl, 50% glycerol, 0.5% Triton X-100, pH 7.5.
  • ABTS solution 100 mM citric acid, 250 mM Na 2 HP0 4 , 0.5 mg/ml ABTS (2,2' -azinobis (3-ethylbenzthiazlinesulfonic acid), adjusted to pH 4.0 with IN HCl.
  • Cell lysis buffer HNTG containing ImM Na 3 V0 4 (0.5M solution kept as a 100X stock at -80°C in aliquots) , 5mM NaP 2 0 7 and 5mM EDTA prepared fresh and keep on ice until ready for use.
  • Hydrogen peroxide 30% solution.
  • SEEDING CELLS Target cells were grown in 15cm culture dish (Corning 25020-100) in DMEM media containing 10% fetal bovine serum (FBS) until 80-90% confluent. The cells were harvested with trypsin-EDTA (0.25%, 0.5ml, Gibco), resuspended in fresh medium containing 10% FBS, 1% L-glutamine and Hepes, and transferred to round bottom 96-well tissue culture plates (Corning 25806-96) at 25,000 cells/well, lOO ⁇ l/well. The cells were incubated at 37°C at 5% C0 2 for 24 hours. The media was changed by inverting the plate, and adding DMEM medium containing 0.5% FBS and Hepes. The cells were further incubated overnight at 37°C, 5% C0 2 .
  • FBS fetal bovine serum
  • ASSAY PROCEDURE The assay was set up in the 96-well tissue culture plate. Before adding test substance to the cells, media in the wells was replaced by serum free DMEM medium, 90 ⁇ l per well. Positive control wells received 80 ⁇ l DMEM. Negative controls received 90 ⁇ l DMEM. Test substances were diluted 1:10 with DMEM and 10 ⁇ l/well of the diluted test substances were transferred to the cells in the wells to achieve a final dilution of 1:100. Positive and negative control wells received 10 ⁇ l/well of dimethyl sulphoxide (DMSO) to achieve a final concentration of 1%.
  • DMSO dimethyl sulphoxide
  • Positive control wells additionally received 10 ⁇ l/well of 0.1 M Na 3 V0 4 so that the final concentration is 10 mM.
  • the microtiter plate was shaken for 1 minute before incubation at 37°C and 5% C0 2 . After 90 minutes of incubation, the media was removed by inversion of the plate, and lOO ⁇ l/well of lysis buffer was added to the cells.
  • the tissue culture plate was shaken for 5 minutes and then placed on ice for 10 minutes.
  • the cells were homogenized by repeated aspirating and dispensing, and the cell lysates were transferred to the corresponding wells of a precoated assay plate.
  • the substrate in the cell lysates was allowed to bind to the anchoring antibody for 1 hour shaking at room temperature. The lysates were then removed, and the assay plate was washed.
  • the detection antibody was then removed, the plate washed, and fresh ABTS/H 2 0 2 (1.2 ⁇ l 30% H 2 0 2 to 10ml 0.5mg/ml 2,2'- azinobis (3-ethylbenzethiazline) sulfonic acid in lOOmM citric acid, 250mM Na 2 HP0 4 , pH4.0) was added to start color development.
  • the reaction was stopped after 10 minutes by adding lOO ⁇ l/well of 0.2M HCl, and shaking for 1 minute.
  • Absorbance values at 410 nm were measured by a ELISA plate reader (Dynatec MR5000) .
  • test substances 6.2.5. RESULTS Results of the assay and formulae of the test substances are presented in Table III. The activity of the test substances are represented by the concentration of the test substance which produces the indicated percentage increase in the content of phosphotyrosine over the negative control.
  • the test substances have the general formula:
  • test substance has been shown to be active in the assay, a range of concentrations of the compound were used in experiments wherein the target cells were incubated with the test substance for varying periods of time. The kinetics of the inhibition of dephosphorylation by the test substance at various dosage may thus be obtained.
  • the assay is capable of identifying and evaluating test substances that inhibit dephosphorylation of phosphorylated tyrosine residues on the insulin receptor.
  • This assay may also be used for assessing any test substances for their ability to inhibit the dephosphorylation of other substrate molecules, such as insulin-like growth factor 1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) .
  • IGF-1R insulin-like growth factor 1 receptor
  • EGFR epidermal growth factor receptor
  • NIH3T3/EGFR cells expressing EGFR grown in media containing 0.5% for 40 hours were seeded in the wells of 96-well tissue culture plates at a density 10,000 cells/well.
  • the wells of ELISA plate were coated with anti-EGFR antibodies.
  • This assay provides a method for measuring the effects of test substances on the kinase activity of specific targets in a cell-free system.
  • the target substrate is isolated from cell lysates by immobilization in microtiter plate wells coated with anti-substrate antibody.
  • ATP and a bivalent cation are added to the wells to initiate the kinase reaction. Concentrations of ATP can be varied to assess whether a test compound competes for
  • the concentration of ATP should be ⁇ 3 ⁇ M for assessing high sensitivity in the kinase reaction.
  • a concentration of 10 - 50 ⁇ M ATP is preferred for assessing inhibition of ATP interaction, and most preferably
  • test compound stocks (10 mg/ml in DMSO) were diluted 1:20 into 5% DMSO in PBS for a final concentration range of 150 ⁇ M to 1 nM, then 135 ⁇ l was added per well. The plate was incubated for 30 minutes, shaking, at room temperature.
  • the kinase reaction was initiated by the addition of 15 ⁇ l of ATP/Mn mix (in 50 mM MnCL 2 , final concentration of ATP 1 ⁇ M, 3 ⁇ M or 10 ⁇ M) for a total volume of 150 ⁇ l.
  • the plate was incubated for 5 minutes at room temperature, shaking, then washed 5 times with TBST.
  • the amount of phosphorylation present was measured as described in Section 6.2.3. 7.3. RESULTS
  • This assay provides a method for measuring the effects of test substances on the phosphatase activity of PTP IB as measured by dephosphorylation of EGFR.
  • the assay protocol used is substantially the same as that described in Section 7.
  • TBS 50 mM Tris, pH 7.2, 150 mM Nacl
  • PTP IB the protein was produced in bacterial cells (strain B121) using a pET plasmid with a T7 promoter, Tonks, et al. Methods in Enzymology 201:427-443, 1991.
  • phosphatase buffer 100 mM Tris, pH 7.2, 5% DMSO, 1.5 mM NaPyrophosphate.
  • the assay procedure used was substantially the same as that described in Section 8.2.3 with the following changes.
  • a kinase reaction was initiated by adding 135 ⁇ l of TBS per well, then 15 ⁇ l of 0.003 mM ATP in 50 mM MnCl2, for a final concentration of 3 ⁇ M ATP and 5mM MnC12.
  • the reaction was allowed to proceed for 5 minutes, shaking, then stopped with the addition of 16.5 ⁇ l of 200 mM EDTA, pH 8.0, shaking continuously during this addition.
  • the plate was left shaking for an additional minute then washed 5 times with distilled water and once with TBST.
  • Test compounds were diluted 1:100 in phosphatase buffer, and aliquots of 140 ⁇ l were added to each well. Control wells received phosphatase buffer alone.
  • the phosphatase reaction was initiated by the addition of PTP IB diluted in 100 mM Tris, pH 7.2 (10 ⁇ l (20 ng) per well) . The plate was incubated for 15 minutes, shaking, then washed 5 times with distilled water and once with TBST. The amount of phosphotyrosine remaining is measured as described in Section 6.2.3.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Hematology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention se rapporte à des systèmes d'épreuves rapides et quantitatives de sélection de composés d'essais en vue d'apprécier leur aptitude à moduler les activités de la tyrosine kinase ou de la phosphotyrosine phosphatase intervenant dans une transduction de signal. Ces épreuves peuvent être utilisées pour identifier des composés en vue de leurs applications thérapeutiques pour des affections dans lesquelles l'activité de la tyrosine kinase ou de la phosphatase dans un processus de transduction de signal contribue à un processus pathologique.
PCT/US1996/008332 1995-06-07 1996-06-03 Epreuves de selection de composes WO1996040276A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU59658/96A AU5965896A (en) 1995-06-07 1996-06-03 Screening assays for compounds
EP96916950A EP0873142A4 (fr) 1995-06-07 1996-06-03 Epreuves de selection de composes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US488,156 1990-03-02
US48815695A 1995-06-07 1995-06-07

Publications (1)

Publication Number Publication Date
WO1996040276A1 true WO1996040276A1 (fr) 1996-12-19

Family

ID=23938536

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/008332 WO1996040276A1 (fr) 1995-06-07 1996-06-03 Epreuves de selection de composes

Country Status (3)

Country Link
EP (1) EP0873142A4 (fr)
AU (1) AU5965896A (fr)
WO (1) WO1996040276A1 (fr)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998011251A1 (fr) * 1996-09-13 1998-03-19 Cambridge University Technical Services Ltd. Detection de sites de reconnaissance de substrats pour les proteine kinases et les proteine phosphatases
WO1999003498A1 (fr) * 1997-07-18 1999-01-28 Novo Nordisk A/S Utilisation de fvii ou fviiai pour le traitement de pathologies liees au processus de transmission des signaux intracellulaires induites par fviia
US6022708A (en) * 1998-02-26 2000-02-08 Genentech, Inc. Fused
WO2000053801A1 (fr) * 1999-03-08 2000-09-14 Merck Frosst Canada & Co. Mesure de cellules entieres pour des proteine tyrosine phosphatases
EP1113780A2 (fr) * 1998-09-17 2001-07-11 Bionexus Techniques permettant de determiner l'activite de melanges complexes
US6355786B1 (en) 1998-10-30 2002-03-12 Vanderbilt University Purified and isolated protein zero related (PZR) and therapeutic and screening methods using same
US6406869B1 (en) * 1999-10-22 2002-06-18 Pharmacopeia, Inc. Fluorescent capture assay for kinase activity employing anti-phosphotyrosine antibodies as capture and detection agents
US6451977B1 (en) 1998-02-26 2002-09-17 Genentech, Inc. Fused polypeptides
US6461610B1 (en) 1997-07-18 2002-10-08 Novo Nordisk A/S Methods for modifying cell motility using factor VIIa or inactivated factor VIIa
US6531579B1 (en) 1998-02-26 2003-03-11 Genentech, Inc. Fused polypeptides
USRE38915E1 (en) 1997-01-15 2005-12-06 Telik, Inc. Nonpeptide insulin receptor agonists
JP2009102342A (ja) * 1996-08-30 2009-05-14 Novogen Research Pty Ltd イソフラボン類を含有する治療方法及び組成物
US7732151B2 (en) * 2003-05-22 2010-06-08 Sanofi-Aventis Deutschland Gmbh Use of IRS-polypeptides for identifying of pharmaceutically active compounds
US8487078B2 (en) 2003-07-29 2013-07-16 Life Technologies Corporation Kinase and phosphatase assays

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990010234A1 (fr) * 1989-03-03 1990-09-07 Euro-Diagnostics B.V. Procede et kit determinant la phosphorylation de tyrosine ainsi que des activites de dephosphorylation

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993003377A1 (fr) * 1991-07-31 1993-02-18 Ziltener Hermann J Procede de detection de restes de phosphotyrosine

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990010234A1 (fr) * 1989-03-03 1990-09-07 Euro-Diagnostics B.V. Procede et kit determinant la phosphorylation de tyrosine ainsi que des activites de dephosphorylation

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CLEAVELAND J S, ET AL.: "A MICROTITER-BASED ASSAY FOR THE DETECTION OF PROTEIN TYROSINE KINASE ACTIVITY", vol. 190, 1 January 1990 (1990-01-01), pages 249 - 253, XP002937575 *
FARLEY K, ET AL.: "DEVELOPMENT OF SOLID-PHASE ENZYME-LINKED IMMUNOSORBENT ASSAYS FOR THE DETERMINATION OF EPIDERMAL GROWTH FACTOR RECEPTOR AND PP60C-SRCTYROSINE PROTEIN KINASE ACTIVITY", ANALYTICAL BIOCHEMISTRY., ACADEMIC PRESS INC., NEW YORK., vol. 203, 1 January 1992 (1992-01-01), NEW YORK., pages 151 - 157, XP002945391, ISSN: 0003-2697, DOI: 10.1016/0003-2697(92)90056-D *
KING I C, FENG M, CATINO J J: "HIGH THROUGHPUT ASSAY FOR INHIBITORS OF THE EPIDERMAL GROWTH FACTORRECEPTOR-ASSOCIATED TYROSINE KINASE", LIFE SCIENCES., PERGAMON PRESS, OXFORD, GB, vol. 53, 1 January 1993 (1993-01-01), GB, pages 1465 - 1472, XP002945390, ISSN: 0024-3205, DOI: 10.1016/0024-3205(93)90619-E *
RIJKSEN G., ET AL.: "A NONRADIOACTIVE DOT-BLOT ASSAY FOR PROTEIN TYROSINE KINASE ACTIVITY.", ANALYTICAL BIOCHEMISTRY., ACADEMIC PRESS INC., NEW YORK., vol. 182., 1 January 1989 (1989-01-01), NEW YORK., pages 98 - 102., XP000945061, ISSN: 0003-2697, DOI: 10.1016/0003-2697(89)90724-0 *
See also references of EP0873142A4 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009102342A (ja) * 1996-08-30 2009-05-14 Novogen Research Pty Ltd イソフラボン類を含有する治療方法及び組成物
WO1998011251A1 (fr) * 1996-09-13 1998-03-19 Cambridge University Technical Services Ltd. Detection de sites de reconnaissance de substrats pour les proteine kinases et les proteine phosphatases
USRE39866E1 (en) 1997-01-15 2007-10-02 Telik, Inc. Nonpeptide insulin receptor agonists
USRE38915E1 (en) 1997-01-15 2005-12-06 Telik, Inc. Nonpeptide insulin receptor agonists
US6268163B1 (en) 1997-07-18 2001-07-31 Novo Nordisk A/S Identification of drug candidates modulating factor VII-mediated intracellular signaling
WO1999003498A1 (fr) * 1997-07-18 1999-01-28 Novo Nordisk A/S Utilisation de fvii ou fviiai pour le traitement de pathologies liees au processus de transmission des signaux intracellulaires induites par fviia
US6461610B1 (en) 1997-07-18 2002-10-08 Novo Nordisk A/S Methods for modifying cell motility using factor VIIa or inactivated factor VIIa
US7259245B2 (en) 1998-02-26 2007-08-21 Genentech, Inc. Anti-fused antibodies
US6022708A (en) * 1998-02-26 2000-02-08 Genentech, Inc. Fused
US6451977B1 (en) 1998-02-26 2002-09-17 Genentech, Inc. Fused polypeptides
US6531579B1 (en) 1998-02-26 2003-03-11 Genentech, Inc. Fused polypeptides
US8394939B2 (en) 1998-02-26 2013-03-12 Genentech, Inc. Nucleic acid encoding fused
EP1113780A4 (fr) * 1998-09-17 2001-11-07 Bionexus Techniques permettant de determiner l'activite de melanges complexes
EP1113780A2 (fr) * 1998-09-17 2001-07-11 Bionexus Techniques permettant de determiner l'activite de melanges complexes
US6355786B1 (en) 1998-10-30 2002-03-12 Vanderbilt University Purified and isolated protein zero related (PZR) and therapeutic and screening methods using same
US6939952B2 (en) 1998-10-30 2005-09-06 Vanderbilt University Purified and isolated protein zero related (PZR) polypeptide
WO2000053801A1 (fr) * 1999-03-08 2000-09-14 Merck Frosst Canada & Co. Mesure de cellules entieres pour des proteine tyrosine phosphatases
US6329137B1 (en) 1999-03-08 2001-12-11 Merck Frosst Canada & Co. Intact cell assay for protein tyrosine phosphatases using recombinant baculoviruses
US6406869B1 (en) * 1999-10-22 2002-06-18 Pharmacopeia, Inc. Fluorescent capture assay for kinase activity employing anti-phosphotyrosine antibodies as capture and detection agents
US6806056B2 (en) * 1999-10-22 2004-10-19 Pharmacopeia Drug Discovery, Inc. Fluorescent capture assay for kinase activity employing an anti-phosphotyrosine antibody as both the capture agent and detecting agent
US7732151B2 (en) * 2003-05-22 2010-06-08 Sanofi-Aventis Deutschland Gmbh Use of IRS-polypeptides for identifying of pharmaceutically active compounds
US8487078B2 (en) 2003-07-29 2013-07-16 Life Technologies Corporation Kinase and phosphatase assays

Also Published As

Publication number Publication date
EP0873142A4 (fr) 2001-08-08
EP0873142A1 (fr) 1998-10-28
AU5965896A (en) 1996-12-30

Similar Documents

Publication Publication Date Title
US5763198A (en) Screening assays for compounds
Han et al. Tyrphostin AG 1478 preferentially inhibits human glioma cells expressing truncated rather than wild-type epidermal growth factor receptors
WO1996040276A1 (fr) Epreuves de selection de composes
US7279290B2 (en) ATM kinase compositions and methods
US7494784B2 (en) Method of treating cell-migration conditions or diseases
JP2008537488A (ja) Gdf−8モジュレート物質の検出
WO1995024205A1 (fr) Procede et compositions de traitement du cancer du sein
MX2007011405A (es) Deteccion de una respuesta inmune para agentes moduladores de crecimiento y la diferenciacion del factor 8 (gdf 8).
US5856111A (en) Methods for identifying modulators of insulin receptor phosphorylation
JP2002512685A (ja) インスリン受容体活性のモジュレーター
EP1212621B1 (fr) Criblage d'agents modulateurs de signalisation cellulaire tgf-beta
US20150185215A1 (en) Cell-Based Assays For Post-Translational Enzyme Activity
US6045797A (en) Treatment or diagnosis of diseases or conditions associated with a BLM domain
US20200340984A1 (en) Methods of quantifying cftr protein expression
Roberts et al. Tyrosine phosphorylation in signal transduction
EP0797662A1 (fr) Methodes de traitement ou de diagnostic de maladies ou d'etats associes a une transduction anormale des signaux
US5889150A (en) Expression-cloning method for identifying target proteins for eukaryotic tyrosine kinases and novel target protiens
US5807989A (en) Methods for treatment or diagnosis of diseases or disorders associated with an APB domain
US8304521B2 (en) Phospho-specific anti-Pax3 antibodies
US6054280A (en) Methods for diagnosis and treatment of PH domain signal transduction disorders
Waraich et al. Development and precise characterization of phospho-site-specific antibody of Ser357 of IRS-1: Elimination of cross reactivity with adjacent Ser358
Chang et al. Identify Novel Targeting Sites of Calcineurin and CaMKII in Human Cav3. 2 T-type Calcium Channel
AU2020389177A2 (en) Method for detecting cancer bone metastasis and detection reagent
Thakkar et al. Mammalian-heart adenylate deaminase: Cross-species immunoanalysis of tissue distribution with a cardiac-directed antibody
Jin Protein Tyrosine Phosphorylation in Haematopoietic Cancers and the Functional Significance of Phospho-Lyn SH2 Domain

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BB BG BR BY CA CN CZ EE FI GE HU IL IS JP KG KP KR KZ LK LR LS LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1996916950

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996916950

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: CA

WWW Wipo information: withdrawn in national office

Ref document number: 1996916950

Country of ref document: EP