EP1872134A2 - Réactifs de détection de phosphorylation de protéines dans la voie de signalisation de carcinome - Google Patents

Réactifs de détection de phosphorylation de protéines dans la voie de signalisation de carcinome

Info

Publication number
EP1872134A2
EP1872134A2 EP06739581A EP06739581A EP1872134A2 EP 1872134 A2 EP1872134 A2 EP 1872134A2 EP 06739581 A EP06739581 A EP 06739581A EP 06739581 A EP06739581 A EP 06739581A EP 1872134 A2 EP1872134 A2 EP 1872134A2
Authority
EP
European Patent Office
Prior art keywords
rows
corresponding column
phosphorylated
peptide
column
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.)
Withdrawn
Application number
EP06739581A
Other languages
German (de)
English (en)
Other versions
EP1872134A4 (fr
Inventor
Ailan Guo
Klarisa Rikova
Albrecht Moritz
Yu Li
Charles Farnsworth
Kimberly Lee
Roberto Polakiewicz
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.)
Cell Signaling Technology Inc
Original Assignee
Cell Signaling Technology 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 Cell Signaling Technology Inc filed Critical Cell Signaling Technology Inc
Publication of EP1872134A2 publication Critical patent/EP1872134A2/fr
Publication of EP1872134A4 publication Critical patent/EP1872134A4/fr
Withdrawn legal-status Critical Current

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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids

Definitions

  • the invention relates generally to antibodies and peptide reagents for the detection of protein phosphorylation, and to protein phosphorylation in cancer.
  • Protein phosphorylation plays a critical role in the etiology of many pathological conditions and diseases, including cancer, developmental disorders, autoimmune diseases, and diabetes. Yet, in spite of the importance of protein modification, it is not yet well understood at the molecular level, due to the extraordinary complexity of signaling pathways, and the slow development of technology necessary to unravel it.
  • Protein phosphorylation on a proteome-wide scale is extremely complex as a result of three factors: the large number of modifying proteins, e.g. kinases, encoded in the genome, the much larger number of sites on substrate proteins that are modified by these enzymes, and the dynamic nature of protein expression during growth, development, disease states, and aging.
  • the human genome for example, encodes over 520 different protein kinases, making them the most abundant class of enzymes known. See Hunter, Nature 411: 355-65 (2001). Most kinases phosphorylate many different substrate proteins, at distinct tyrosine, serine, and/or threonine residues. Indeed, it is estimated that one-third of all proteins encoded by the human genome are phosphorylated, and many are phosphorylated at multiple sites by different kinases.
  • Carcinoma is one of the two main categories of cancer, and is generally characterized by the formation of malignant tumors or cells of epithelial tissue original, such as skin, digestive tract, glands, etc.
  • Carcinomas are malignant by definition, and tend to metastasize to other areas of the body.
  • the most common forms of carcinoma are skin cancer, lung cancer, breast cancer, and colon cancer, as well as other numerous but less prevalent carcinomas.
  • Current estimates show that, collectively, various carcinomas will account for approximately 1.65 million cancer diagnoses in the United States alone, and more than 300,000 people will die from some type of carcinoma during 2005. (Source: American Cancer Society (2005)). The worldwide incidence of carcinoma is much higher.
  • RTKs receptor tyrosine kinases
  • Constitutively active RTKs can contribute not only to unrestricted cell proliferation, but also to other important features of malignant tumors, such as evading apoptosis, the ability to promote blood vessel growth, the ability to invade other tissues and build metastases at distant sites (see Blume-Jensen et al., Nature 411: 355-365 (2001)). These effects are mediated not only through aberrant activity of RTKs themselves, but, in turn, by aberrant activity of their downstream signaling molecules and substrates.
  • non-small cell lung carcinoma patients carrying activating mutations in the epidermal growth factor receptor (EGFR), an RTK appear to respond better to specific EGFR inhibitors than do patients without such mutations (Lynch et al., supra.; Paez et al., Science 304: 1497-1500 (2004)).
  • EGFR epidermal growth factor receptor
  • carcinoma is made by tissue biopsy and detection of different cell surface markers.
  • misdiagnosis can occur since some carcinoma cases can be negative for certain markers and because these markers may not indicate which genes or protein kinases may be deregulated.
  • the genetic translocations and/or mutations characteristic of a particular form of carcinoma can be sometimes detected, it is clear that other downstream effectors of constitutively active kinases having potential diagnostic, predictive, or therapeutic value, remain to be elucidated. Accordingly, identification of downstream signaling molecules and phosphorylation sites involved in different types of carcinoma and development of new reagents to detect and quantify these sites and proteins may lead to improved diagnostic/prognostic markers, as well as novel drug targets, for the detection and treatment of this disease.
  • the invention discloses 214 novel phosphorylation sites identified in signal transduction proteins and pathways underlying human carcinomas and provides new reagents, including phosphorylation-site specific antibodies and AQUA peptides, for the selective detection and quantification of these phosphorylated sites/proteins. Also provided are methods of using the reagents of the invention for the detection and quantification of the disclosed phosphorylation sites.
  • FIG. 3 - is an exemplary mass spectrograph depicting the detection of the tyrosine 224 phosphorylation site in Etk (see Row 119 in Figure 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • FIG. 4 - is an exemplary mass spectrograph depicting the detection of the tyrosine 1159 phosphorylation site in HER3 (see Row 133 in Figure 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • FIG. 5 - is an exemplary mass spectrograph depicting the detection of the tyrosine 542 phosphorylation site in IRS-2 (see Row 15 in Figure 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase “y” in Figure 2) and M# (and lowercase “m”) indicates an oxidized methionine also detected.
  • FIG. 6 - is an exemplary mass spectrograph depicting the detection of the tyrosine 849 phosphorylation site in PDGFR ⁇ (see Row 139 in Figure 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y * (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • FIG. 7 - is an exemplary mass spectrograph depicting the detection of the tyrosine 66 phosphorylation site in RasGAP 3 (see Row 87 in Figure 2/ Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • FIG. 8 - is an exemplary mass spectrograph depicting the detection of the tyrosine 172 phosphorylation site in Requiem (see Row 197 in Figure 2/ Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • FIG. 9 - is an exemplary mass spectrograph depicting the detection of the tyrosine 516 phosphorylation site in WNK1 (see Row 117 in Figure 2/ Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (shown as lowercase "y" in Figure 2).
  • phosphorylation sites correspond to numerous different parent proteins (the full sequences of which (human) are all publicly available in SwissProt database and their Accession numbers listed in Column B of Table 1/Fig. 2), each of which fall into discrete protein type groups, for example Protein Kinases (Serine/Threonine nonreceptor, Tyrosine receptor, Tyrosine nonreceptor, dual specificity and other), Adaptor/Scaffold proteins, Cytoskeletal proteins, and Cellular Metabolism enzymes, etc. (see Column C of Table 1), the phosphorylation of which is relevant to signal transduction activity underlying carcinomas (e.g., skin, lung, breast and colon cancer), as disclosed herein.
  • carcinomas e.g., skin, lung, breast and colon cancer
  • the discovery of the 214 novel protein phosphorylation sites described herein enables the production, by standard methods, of new reagents, such as phosphorylation site-specific antibodies and AQUA peptides (heavy-isotope labeled peptides), capable of specifically detecting and/or quantifying these phosphorylated sites/proteins.
  • new reagents such as phosphorylation site-specific antibodies and AQUA peptides (heavy-isotope labeled peptides), capable of specifically detecting and/or quantifying these phosphorylated sites/proteins.
  • Such reagents are highly useful, inter alia, for studying signal transduction events underlying the progression of carcinoma.
  • the invention provides novel reagents -- phospho-specific antibodies and AQUA peptides ⁇ for the specific detection and/or quantification of a carcinoma-related signaling protein/polypeptide only when phosphorylated (or only when not phosphorylated) at a particular phosphorylation site disclosed herein.
  • the invention provides an isolated phosphorylation site- specific antibody that specifically binds a given carcinoma-related signaling protein only when phosphorylated (or not phosphorylated, respectively) at a particular tyrosine enumerated in Column D of Table 1/ Figure 2 comprised within the phosphorylatable peptide site sequence enumerated in corresponding Column E.
  • the invention provides a heavy-isotope labeled peptide (AQUA peptide) for the detection and quantification of a given carcinoma-related signaling protein, the labeled peptide comprising a particular phosphorylatable peptide site/sequence enumerated in Column E of Table 1/ Figure 2 herein.
  • the reagents provided by the invention is an isolated phosphorylation site-specific antibody that specifically binds the PRK2 kinase (serine/threonine) only when phosphorylated (or only when not phosphorylated) at tyrosine 635 (see Row 115 (and Columns D and E) of Table 1/ Figure 2).
  • the group of reagents provided by the invention is an AQUA peptide for the quantification of phosphorylated PRK2 kinase, the AQUA peptide comprising the phosphorylatable peptide sequence listed in Column E, Row 115, of Table 1/ Figure 2 (which encompasses the phosphorylatable tyrosine at position 635).
  • the invention provides an isolated phosphorylation site-specific antibody that specifically binds a human carcinoma-related signaling protein selected from Column A of Table 1 (Rows 2-215) only when phosphorylated at the tyrosine residue listed in corresponding Column D of Table 1 , comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 1-214), wherein said antibody.does not bind said signaling protein when not phosphorylated at said tyrosine.
  • the invention provides an isolated phosphorylation site- specific antibody that specifically binds a carcinoma-related signaling protein selected from Column A of Table 1 only when not phosphorylated at the tyrosine residue listed in corresponding Column D of Table 1, comprised within the peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 1-214), wherein said antibody does not bind said signaling protein when phosphorylated at said tyrosine.
  • Such reagents enable the specific detection of phosphorylation (or non- phosphorylation) of a novel phosphorylatable site disclosed herein.
  • the invention further provides immortalized cell lines producing such antibodies.
  • the immortalized cell line is a rabbit or mouse hybridoma.
  • the invention provides a heavy-isotope labeled peptide (AQUA peptide) for the quantification of a carcinoma- related signaling protein selected from Column A of Table 1 , said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 1-214), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D of Table 1.
  • the phosphorylatable tyrosine within the labeled peptide is phosphorylated, while in other preferred embodiments, the phosphorylatable residue within the labeled peptide is not phosphorylated.
  • Reagents may conveniently be grouped by the type of carcinoma-related signaling protein in which a given phosphorylation site (for which reagents are provided) occurs.
  • the protein types for each respective protein are provided in Column C of Table 1/ Figure 2, and include: Adaptor/Scaffold proteins, Calcium-binding proteins, Cell Cycle Regulation proteins, Channel proteins, Chaperone proteins, Cholesterol metabolism proteins, Coagulation proteins, Cytoskeleton proteins, Extracellular Matrix proteins, Glycosylation proteins, GTP signaling proteins, lnflammasome proteins, Intracellular transport proteins, Kinases (Serine/ Threonine, dual specificity, Tyrosine etc.), Metabolism proteins, Neurotransmitter pathway proteins, Phosphatases, Phosphodiesterases, Proteases, Receptor proteins and Receptor ligands, RNA processing proteins, Transcription/ Translation proteins, Trans
  • Particularly preferred subsets of the phosphorylation sites (and their corresponding proteins) disclosed herein are those occurring on the following protein types/groups listed in Column C of Table 1/ Figure 2: Adaptor/Scaffold proteins, Cytoskeleton proteins, GTP Signaling proteins, Kinases (including Serine/Threonine dual specificity, and Tyrosine kinases), Metabolism proteins, Phosphatases, Phosphodiesterases/ Proteases, Receptor proteins, RNA Processing proteins, Translation proteins, and Ubitquitin proteins. Accordingly, among preferred subsets of reagents provided by the invention are isolated antibodies and AQUA peptides useful for the detection and/or quantification of the foregoing preferred protein/phosphorylation site subsets.
  • an isolated phosphorylation site-specific antibody that specifically binds an Adaptor/Scaffold protein selected from Column A, Rows 2-35, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 2-35, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 2-35, of Table 1 (SEQ ID NOs: 1-34), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • a heavy-isotope labeled peptide (AQUA peptide) for the quantification of a carcinoma-related signaling protein that is an Adaptor/Scaffold protein selected from Column A, Rows 2-35, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 2-35, of Table 1 (SEQ ID NOs: 1-34), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 2-35, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Adaptor/Scaffold protein phosphorylation sites are particularly preferred: GRB7 (Y107), IRS-2 (Y542, Y766, Y598, Y742), P130Cas (Y287) and SOCS5 (Y519) (see SEQ ID NOs: 12, 14-17, 24 and 29).
  • antibodies and AQUA peptides for the detection/quantification of the following Cytoskeleton protein phosphorylation sites are particularly preferred: MAPI B (Y1062, Y1938, Y1889, Y2042, Y1940, Y1923, Y1887), Plakophilin4 (Y415, Y306, Y1115), Radixin (Y134), Smoothelin (Y897, Y902) and WIRE (Y255) (see SEQ ID NOs: 55-61 , 65-67 and 71-74).
  • an isolated phosphorylation site-specific antibody that specifically binds a GTP signaling protein selected from Column A, Rows 82-87, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 82-87, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 82-87, of Table 1 (SEQ ID NOs: 81-86), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • antibodies and AQUA peptides for the detection/quantification of the following GTP signaling protein phosphorylation sites are particularly preferred: BCAR3 (Y117, Y429) and RasGAP 3 (Y66) (see SEQ ID NOs: 81-82 and 86).
  • MARK4 (Y273), PAK5 (Y146, Y160, Y159), PRK2 (Y635), WNK1 (Y516), Etk (Y224, Y365), AxI (Y696), CSFR (Y923, Y571 , Y556, Y873), EphA5 (Y623), HER3 (Y1159), Kit (Y730, Y578, Y7470), Met (Y830, Y835) and PDGFR ⁇ (Y849) (see SEQ ID NOs: 108-111, 114, 116, 118, 119, 123-128 and 132-138).
  • antibodies and AQUA peptides for the detection/quantification of the following Cellular metabolism enzyme phosphorylation sites are particularly preferred: adolase A (Y363) (see SEQ ID NO: 140).
  • antibodies and AQUA peptides for the detection/quantification of the following Phosphatase/Phosphodiesterase/Protease phosphorylation sites are particularly preferred: Cdc25a (Y463, Y469, Y459), CNP (Y110), and ACE (Y1067) (see SEQ ID NOs: 153-157).
  • antibodies and AQUA peptides for the detection/quantification of the following Receptor protein phosphorylation sites are particularly preferred: IFNGR1 (Y397), IGF2R (Y1592), LDLR (Y847, Y828) and TNF-R1 (Y401) (see SEQ ID NOs: 161 , 163, 165, 166 and 171).
  • antibodies and AQUA peptides for the detection/quantification of the following RNA Processing protein phosphorylation sites are particularly preferred: RBM3 (Y 117, Y127), and SF3A3 (Y479) (see SEQ ID NOs: 186-187 and 189).
  • a heavy-isotope labeled peptide for the quantification of a carcinoma-related signaling protein that is a Transcription protein selected from Column A, Rows 191-203, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 191-203, of Table 1 (SEQ ID NOs: 190- 202), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 191-203, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Transcription protein phosphorylation sites are particularly preferred: CBP (Y659), Requiem (Y172), TBX2 (Y237), and Trap170 (Y746, Y749) (see SEQ ID NOs: 190, 196, and 200-202).
  • An isolated phosphorylation site-specific antibody specifically binds a Translation protein selected from Column A, Rows 204-206, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 204-206, of Table 1 , comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 204-206, of Table 1 (SEQ ID NOs: 203-205), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • an isolated phosphorylation site-specific antibody that specifically binds a Transporter protein selected from Column A, Rows 210-213, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 210-213, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 210-213, of Table 1 (SEQ ID NOs: 209-212), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • the invention also provides, in part, an immortalized cell line producing an antibody of the invention, for example, a cell line producing an antibody within any of the foregoing preferred subsets of antibodies.
  • the immortalized cell line is a rabbit hybridoma or a mouse hybridoma.
  • a heavy-isotope labeled peptide (AQUA peptide) of the invention comprises a disclosed site sequence wherein the phosphorylatable tyrosine is phosphorylated.
  • a heavy-isotope labeled peptide of the invention comprises a disclosed site sequence wherein the phosphorylatable tyrosine is not phosphorylated.
  • methods for detecting or quantifying a carcinoma-related signaling protein that is tyrosine phosphorylated comprising the step of utilizing one or more of the above-described reagents of the invention to detect or quantify one or more carcinoma-related signaling protein(s) selected from Column A of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D of Table 1.
  • the reagents comprise a subset of preferred reagents as described above.
  • the identification of the disclosed 214 novel carcinoma-related signaling protein phosphorylation sites, and the standard production and use of the reagents provided by the invention are described in further detail below and in the Examples that follow.
  • Antibody refers to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including F a b or antigen-recognition fragments thereof, including chimeric, polyclonal, and monoclonal 20 antibodies.
  • Carcinoma-related signaling protein means any protein (or polypeptide derived therefrom) enumerated in Column A of Table 1/ Figure 2, which is disclosed herein as being phosphorylated in one or more human carcinoma cell line(s).
  • Carcinoma-related signaling proteins may be protein kinases, or direct substrates of such kinases, or may be indirect substrates downstream of such kinases in signaling pathways.
  • a carcinoma-related signaling protein may also be phosphorylated in other cell lines (non-carcinomic) harboring activated kinase activity.
  • Heavy-isotope labeled peptide (used interchangeably with AQUA peptide) means a peptide comprising at least one heavy-isotope label, which is suitable for absolute quantification or detection of a protein as described in WO/03016861, “Absolute Quantification of Proteins and Modified Forms Thereof by Multistage Mass Spectrometry” (Gygi et al), further discussed below.
  • Protein is used interchangeably with polypeptide, and includes protein fragments and domains as well as whole protein.
  • Phosphorylatable amino acid means any amino acid that is capable of being modified by addition of a phosphate group, and includes both forms of such amino acid.
  • Phosphorylatable peptide sequence means a peptide sequence comprising a phosphorylatable amino acid.
  • Phosphorylation site-specific antibody means an antibody that specifically binds a phosphorylatable peptide sequence/epitope only when phosphorylated, or only when not phosphorylated, respectively. The term is used interchangeably with "phospho-specific" antibody.
  • the immunoaffinity/mass spectrometric technique described in the '848 Patent Publication (the "IAP" method) ⁇ and employed as described in detail in the Examples — is briefly summarized below.
  • the IAP method employed generally comprises the following steps: (a) a proteinaceous preparation (e.g.
  • a digested cell extract comprising phosphopeptides from two or more different proteins is obtained from an organism; (b) the preparation is contacted with at least one immobilized general phosphotyrosine-specific antibody; (c) at least one phosphopeptide specifically bound by the immobilized antibody in step (b) is isolated; and (d) the modified peptide isolated in step (c) is characterized by mass spectrometry (MS) and/or tandem mass spectrometry (MS-MS).
  • a search program e.g. Sequest
  • Sequest may be utilized to substantially match the spectra obtained for the isolated, modified peptide during the characterization of step (d) with the spectra for a known peptide sequence.
  • a quantification step employing, e.g. SILAC or AQUA, may also be employed to quantify isolated peptides in order to compare peptide levels in a sample to a baseline.
  • SILAC SILAC
  • AQUA a general phosphotyrosine-specific monoclonal antibody (commercially available from Cell Signaling Technology, Inc., Beverly, MA, Cat #9411 (p-Tyr-100)) was used in the immunoaffinity step to isolate the widest possible number of phospho- tyrosine containing peptides from the cell extracts. Extracts from the human carcinoma cell lines described above were employed.
  • lysates were prepared from these cells line and digested with trypsin after treatment with DTT and iodoacetamide to alkylate cysteine residues.
  • peptides were pre-fractionated by reversed-phase solid phase extraction using Sep-Pak Cis columns to separate peptides from other cellular components.
  • the solid phase extraction cartridges were eluted with varying steps of acetonitrile. Each lyophilized peptide fraction was redissolved in PBS and treated with phosphotyrosine-specific antibody (P-Tyr-100, CST #9411) immobilized on protein G-Sepharose.
  • Immunoaffinity-purified peptides were eluted with 0.1% TFA and a portion of this fraction was concentrated with Stage or Zip tips and analyzed by LC-MS/MS, using a ThermoFinnigan LCQ Deca XP Plus ion trap mass spectrometer. Peptides were eluted from a 10 cm x 75 ⁇ m reversed- phase column with a 45-min linear gradient of acetonitrile. MS/MS spectra were evaluated using the program Sequest with the NCBI human protein database.
  • Isolated phosphorylation site-specific antibodies that specifically bind a carcinoma-related signaling protein disclosed in Column A of Table 1 only when phosphorylated (or only when not phosphorylated) at the corresponding amino acid and phosphorylation site listed in Columns D and E of Table 1/ Figure 2 may now be produced by standard antibody production methods, such as anti-peptide antibody methods, using the phosphorylation site sequence information provided in Column E of Table 1.
  • two previously unknown Etk kinase phosphorylation sites (tyrosine 224 and 365, respectively) (see Rows 119 and 120 of Table 1/Fig. 2) are presently disclosed.
  • Etk kinase phosphorylation sites see Rows 119 and 120 of Table 1/Fig. 2
  • antibodies that specifically bind either of these novel Etk kinase sites can now be produced, e.g.
  • a peptide antigen comprising all or part of the amino acid sequence encompassing the respective phosphorylated residue (e.g. a peptide antigen comprising the sequence set forth in Rows 119 and 120, Column E, of Table 1 (SEQ ID NO: 118 and 119) (which encompasses the phosphorylated tyrosine at positions 224 and 365 in Etk), to produce an antibody that only binds Etk kinase when phosphorylated at those sites.
  • Polyclonal antibodies of the invention may be produced according to standard techniques by immunizing a suitable animal (e.g., rabbit, goat, etc.) with a peptide antigen corresponding to the carcinoma-related phosphorylation site of interest (i.e. a phosphorylation site enumerated in Column E of Table 1, which comprises the corresponding phosphorylatable amino acid listed in Column D of Table 1), collecting immune serum from the animal, and separating the polyclonal antibodies from the immune serum, in accordance with known procedures.
  • a suitable animal e.g., rabbit, goat, etc.
  • a peptide antigen corresponding to the carcinoma-related phosphorylation site of interest i.e. a phosphorylation site enumerated in Column E of Table 1, which comprises the corresponding phosphorylatable amino acid listed in Column D of Table 1
  • a peptide comprising all or part of any one of the phosphorylation site sequences provided in Column E of Table 1 may employed as an antigen to produce an antibody that only binds the corresponding protein listed in Column A of Table 1 when phosphorylated (or when not phosphorylated) at the corresponding residue listed in Column D.
  • the peptide antigen includes the phosphorylated form of the amino acid. Conversely, if an antibody that only binds the protein when not phosphorylated at the disclosed site is desired, the peptide antigen includes the non- phosphorylated form of the amino acid.
  • Peptide antigens suitable for producing antibodies of the invention may be designed, constructed and employed in accordance with well- known techniques. See, e.g., ANTIBODIES: A LABORATORY MANUAL, Chapter 5, p. 75-76, Harlow & Lane Eds., Cold Spring Harbor Laboratory (1988); Czernik, Methods In Enzymology, 201: 264-283 (1991); Merrifield, J. Am. Chem. Soc. 85: 21-49 (1962)).
  • a peptide antigen may comprise the full sequence disclosed in Column E of Table 1/ Figure 2, or it may comprise additional amino acids flanking such disclosed sequence, or may comprise of only a portion of the disclosed sequence immediately flanking the phosphorylatable amino acid (indicated in Column E by lowercase "y").
  • a desirable peptide antigen will comprise four or more amino acids flanking each side of the phosphorylatable amino acid and encompassing it.
  • Polyclonal antibodies produced as described herein may be screened as further described below.
  • Monoclonal antibodies of the invention may be produced in a hybridoma cell line according to the well-known technique of Kohler and Milstein. See Nature 265: 495-97 (1975); Kohler and Milstein, Eur. J. Immunol. 6: 511 (1976); see also, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Ausubel et al. Eds. (1989). Monoclonal antibodies so produced are highly specific, and improve the selectivity and specificity of diagnostic assay methods provided by the invention. For example, a solution containing the appropriate antigen may be injected into a mouse or other species and, after a sufficient time (in keeping with conventional techniques), the animal is sacrificed and spleen cells obtained.
  • the spleen cells are then immortalized by fusing them with myeloma cells, typically in the presence of polyethylene glycol, to produce hybridoma cells.
  • Rabbit fusion hybridomas may be produced as described in U. S Patent No. 5,675,063, C. Knight, Issued October 7, 1997.
  • the hybridoma cells are then grown in a suitable selection media, such as hypoxanthine-aminopterin-thymidine (HAT), and the supernatant screened for monoclonal antibodies having the desired specificity, as described below.
  • the secreted antibody may be recovered from tissue culture supernatant by conventional methods such as precipitation, ion exchange or affinity chromatography, or the like.
  • Monoclonal Fab fragments may also be produced in Escherichia coli by recombinant techniques known to those skilled in the art, See, e.g., W. Huse, Science 246: 1275-81 (1989); Mullinax et al., Proc. Nat'l Acad. Sci. 87: 8095 (1990). If monoclonal antibodies of one isotype are preferred for a particular application, particular isotypes can be prepared directly, by selecting from the initial fusion, or prepared secondarily, from a parental hybridoma secreting a monoclonal antibody of different isotype by using the sib selection technique to isolate class-switch variants (Steplewski, et al., Proc. Nat'l. Acad. ScL, 82: 8653 (1985); Spira et al., J. Immunol. Methods, 74: 307 (1984)).
  • the preferred epitope of a phosphorylation-site specific antibody of the invention is a peptide fragment consisting essentially of about 8 to 17 amino acids including the phosphorylatable tyrosine, wherein about 3 to 8 amino acids are positioned on each side of the phosphorylatable tyrosine (for example, the BCAR3 tyrosine 429 phosphorylation site sequence disclosed in Row 83, Column E of Table 1), and antibodies of the invention thus specifically bind a target carcinoma-related signaling polypeptide comprising such epitopic sequence.
  • Particularly preferred epitopes bound by the antibodies of the invention comprise all or part of a phosphorylatable site sequence listed in Column E of Table 1 , including the phosphorylatable amino acid.
  • non-antibody molecules such as protein binding domains or nucleic acid aptamers, which bind, in a phospho-specific manner, to essentially the same phosphorylatable epitope to which the phospho-specific antibodies of the invention bind. See, e.g., Neuberger et al., Nature 312: 604 (1984).
  • Such equivalent non-antibody reagents may be suitably employed in the methods of the invention further described below.
  • Antibodies provided by the invention may be any type of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including F a b or antigen-recognition fragments thereof.
  • the antibodies may be monoclonal or polyclonal and may be of any species of origin, including (for example) mouse, rat, rabbit, horse, or human, or may be chimeric antibodies. See, e.g., M. Walker et a/., Molec. Immunol. 26: 403-11 (1989); Morrision et al., Proc. Natl Acad. Sci. 81: 6851 (1984); Neuberger et al., Nature 312: 604 (1984)).
  • the antibodies may be recombinant monoclonal antibodies produced according to the methods disclosed in U.S. Pat. No. 4,474,893 (Reading) or U.S. Pat. No. 4,816,567 (Cabilly et al.)
  • the antibodies may also be chemically constructed by specific antibodies made according to the method disclosed in U.S. Pat. No. 4,676,980 (Segel et al.)
  • the invention also provides immortalized cell lines that produce an antibody of the invention.
  • hybridoma clones constructed as described above, that produce monoclonal antibodies to the carcinoma- related signaling protein phosphorylation sitess disclosed herein are also provided.
  • the invention includes recombinant cells producing an antibody of the invention, which cells may be constructed by well known techniques; for example the antigen combining site of the monoclonal antibody can be cloned by PCR and single-chain antibodies produced as phage-displayed recombinant antibodies or soluble antibodies in E. coli (see, e.g., ANTIBODY ENGINEERING PROTOCOLS, 1995, Humana Press, Sudhir Paul editor.)
  • Phosphorylation site-specific antibodies of the invention may be screened for epitope and phospho- specificity according to standard techniques. See, e.g. Czernik et al., Methods in Enzymology, 201: 264-283 (1991).
  • the antibodies may be screened against the phospho and non-phospho peptide library by ELISA to ensure specificity for both the desired antigen (i.e. that epitope including a phosphorylation site sequence enumerated in Column E of Table 1) and for reactivity only with the phosphorylated (or non-phosphorylated) form of the antigen.
  • Peptide competition assays may be carried out to confirm lack of reactivity with other phospho- epitopes on the given carcinoma-related signaling protein.
  • the antibodies may also be tested by Western blotting against cell preparations containing the signaling protein, e.g. cell lines over-expressing the target protein, to confirm reactivity with the desired phosphorylated epitope/target.
  • Specificity against the desired phosphorylated epitope may also be examined by constructing mutants lacking phosphorylatable residues at positions outside the desired epitope that are known to be phosphorylated, or by mutating the desired phospho-epitope and confirming lack of reactivity.
  • Phosphorylation-site specific antibodies of the invention may exhibit some limited cross-reactivity to related epitopes in non-target proteins. This is not unexpected as most antibodies exhibit some degree of cross-reactivity, and anti-peptide antibodies will often cross-react with epitopes having high homology to the immunizing peptide. See, e.g., Czernik, supra. Cross-reactivity with non-target proteins is readily characterized by Western blotting alongside markers of known molecular weight. Amino acid sequences of cross-reacting proteins may be examined to identify sites highly homologous to the carcinoma-related signaling protein epitope for which the antibody of the invention is specific.
  • polyclonal antisera may exhibit some undesirable general cross-reactivity to phosphotyrosine itself, which may be removed by further purification of antisera, e.g. over a phosphotyramine column.
  • Antibodies of the invention specifically bind their target protein (Ae. a protein listed in Column A of Table 1) only when phosphorylated (or only when not phosphorylated, as the case may be) at the site disclosed in corresponding Columns D/E, and do not (substantially) bind to the other form (as compared to the form for which the antibody is specific).
  • Antibodies may be further characterized via immunohistochemical
  • IHC immunohistoneum X-(IHC) staining using normal and diseased tissues to examine carcinoma- related phosphorylation and activation status in diseased tissue.
  • IHC may be carried out according to well-known techniques. See, e.g., ANTIBODIES: A LABORATORY MANUAL, Chapter 10, Harlow & Lane Eds., Cold Spring Harbor Laboratory (1988). Briefly, paraffin-embedded tissue (e.g.
  • tumor tissue is prepared for immunohistochemical staining by deparaffinizing tissue sections with xylene followed by ethanol; hydrating in water then PBS; unmasking antigen by heating slide in sodium citrate buffer; incubating sections in hydrogen peroxide; blocking in blocking solution; incubating slide in primary antibody and secondary antibody; and finally detecting using ABC avidin/biotin method according to manufacturer's instructions.
  • Antibodies may be further characterized by flow cytometry carried out according to standard methods. See Chow et ai, Cytometry (Communications in Clinical Cytometry) 46: 72-78 (2001). Briefly and by way of example, the following protocol for cytometric analysis may be employed: samples may be centrifuged on Ficoll gradients to remove erythrocytes, and cells may then be fixed with 2% paraformaldehyde for 10 minutes at 37 0 C followed by permeabilization in 90% methanol for 30 minutes on ice.
  • Cells may then be stained with the primary phosphorylation-site specific antibody of the invention (which detects a carcinoma-related signal transduction protein enumerated in Table 1), washed and labeled with a fluorescent-labeled secondary antibody. Additional fluorochrome-conjugated marker antibodies (e.g. CD45, CD34) may also be added at this time to aid in the subsequent identification of specific hematopoietic cell types. The cells would then be analyzed on a flow cytometer (e.g. a Beckman Coulter FC500) according to the specific protocols of the instrument used.
  • a flow cytometer e.g. a Beckman Coulter FC500
  • Antibodies of the invention may also be advantageously conjugated to fluorescent dyes (e.g. Alexa488, PE) for use in multi- parametric analyses along with other signal transduction (phospho-CrkL, phospho-Erk 1/2) and/or cell marker (CD34) antibodies.
  • fluorescent dyes e.g. Alexa488, PE
  • CD34 cell marker
  • Phosphorylation-site specific antibodies of the invention specifically bind to a human carcinoma-related signal transduction protein or polypeptide only when phosphorylated at a disclosed site, but are not limited only to binding the human species, perse.
  • the invention includes antibodies that also bind conserved and highly homologous or identical phosphorylation sites in respective carcinoma-related proteins from other species (e.g. mouse, rat, monkey, yeast), in addition to binding the human phosphorylation site. Highly homologous or identical sites conserved in other species can readily be identified by standard sequence comparisons, such as using BLAST, with the human carcinoma-related signal transduction protein phosphorylation sites disclosed herein.
  • the AQUA methodology employs the introduction of a known quantity of at least one heavy-isotope labeled peptide standard (which has a unique signature detectable by LC-SRM chromatography) into a digested biological sample in order to determine, by comparison to the peptide standard, the absolute quantity of a peptide with the same sequence and protein modification in the biological sample.
  • the AQUA methodology has two stages: peptide internal standard selection and validation and method development; and implementation using validated peptide internal standards to detect and quantify a target protein in sample.
  • the method is a powerful technique for detecting and quantifying a given peptide/protein within a complex biological mixture, such as a cell lysate, and may be employed, e.g., to quantify change in protein phosphorylation as a result of drug treatment, or to quantify differences in the level of a protein in different biological states.
  • a particular peptide (or modified peptide) within a target protein sequence is chosen based on its amino acid sequence and the particular protease to be used to digest.
  • the peptide is then generated by solid-phase peptide synthesis such that one residue is replaced with that same residue containing stable isotopes ( 13 C, 15 N).
  • the result is a peptide that is chemically identical to its native counterpart formed by proteolysis, but is easily distinguishable by MS via a 7-Da mass shift.
  • a newly synthesized AQUA internal standard peptide is then evaluated by LC-MS/MS.
  • This process provides qualitative information about peptide retention by reverse-phase chromatography, ionization efficiency, and fragmentation via collision- induced dissociation. Informative and abundant fragment ions for sets of native and internal standard peptides are chosen and then specifically monitored in rapid succession as a function of chromatographic retention to form a selected reaction monitoring (LC-SRM) method based on the unique profile of the peptide standard.
  • LC-SRM reaction monitoring
  • the second stage of the AQUA strategy is its implementation to measure the amount of a protein or modified protein from complex mixtures.
  • Whole cell lysates are typically fractionated by SDS-PAGE gel electrophoresis, and regions of the gel consistent with protein migration are excised. This process is followed by in-gel proteolysis in the presence of the AQUA peptides and LC-SRM analysis.
  • AQUA peptides are spiked in to the complex peptide mixture obtained by digestion of the whole cell lysate with a proteolytic enzyme and subjected to immunoaffinity purification as described above.
  • the retention time and fragmentation pattern of the native peptide formed by digestion e.g.
  • trypsinization is identical to that of the AQUA internal standard peptide determined previously; thus, LC-MS/MS analysis using an SRM experiment results in the highly specific and sensitive measurement of both internal standard and analyte directly from extremely complex peptide mixtures. Because an absolute amount of the AQUA peptide is added (e.g. 250 fmol), the ratio of the areas under the curve can be used to determine the precise expression levels of a protein or phosphorylated form of a protein in the original cell lysate.
  • the internal standard is present during in-gel digestion as native peptides are formed, such that peptide extraction efficiency from gel pieces, absolute losses during sample handling (including vacuum centrifugation), and variability during introduction into the LC-MS system do not affect the determined ratio of native and AQUA peptide abundances.
  • An AQUA peptide standard is developed for a known phosphorylation site sequence previously identified by the IAP-LC-MS/MS method within a target protein.
  • One AQUA peptide incorporating the phosphorylated form of the particular residue within the site may be developed, and a second AQUA peptide incorporating the non- phosphoryiated form of the residue developed.
  • the two standards may be used to detect and quantify both the phosphoryiated and non-phosphorylated forms of the site in a biological sample.
  • Peptide internal standards may also be generated by examining the primary amino acid sequence of a protein and determining the boundaries of peptides produced by protease cleavage. Alternatively, a protein may actually be digested with a protease and a particular peptide fragment produced can then sequenced. Suitable proteases include, but are not limited to, serine proteases (e.g. trypsin, hepsin), metalio proteases (e.g. PUMP1), chymotrypsin, cathepsin, pepsin, thermolysin, carboxypeptidases, etc.
  • a peptide sequence within a target protein is selected according to one or more criteria to optimize the use of the peptide as an internal standard.
  • the size of the peptide is selected to minimize the chances that the peptide sequence will be repeated elsewhere in other non-target proteins.
  • a peptide is preferably at least about 6 amino acids.
  • the size of the peptide is also optimized to maximize ionization frequency.
  • peptides longer than about 20 amino acids are not preferred.
  • the preferred ranged is about 7 to 15 amino acids.
  • a peptide sequence is also selected that is not likely to be chemically reactive during mass spectrometry, thus sequences comprising cysteine, tryptophan, or methionine are avoided.
  • a peptide sequence that does not include a modified region of the target region may be selected so that the peptide internal standard can be used to determine the quantity of all forms of the protein.
  • a peptide internal standard encompassing a modified amino acid may be desirable to detect and quantify only the modified form of the target protein.
  • Peptide standards for both modified and unmodified regions can be used together, to determine the extent of a modification in a particular sample (i.e. to determine what fraction of the total amount of protein is represented by the modified form).
  • peptide standards for both the phosphorylated and unphosphorylated form of a protein known to be phosphorylated at a particular site can be used to quantify the amount of phosphorylated form in a sample.
  • the peptide is labeled using one or more labeled amino acids (i.e. the label is an actual part of the peptide) or less preferably, labels may be attached after synthesis according to standard methods.
  • the label is a mass-altering label selected based on the following considerations: The mass should be unique to shift fragment masses produced by MS analysis to regions of the spectrum with low background; the ion mass signature component is the portion of the labeling moiety that preferably exhibits a unique ion mass signature in MS analysis; the sum of the masses of the constituent atoms of the label is preferably uniquely different than the fragments of all the possible amino acids.
  • the labeled amino acids and peptides are readily distinguished from unlabeled ones by the ion/mass pattern in the resulting mass spectrum.
  • the ion mass signature component imparts a mass to a protein fragment that does not match the residue mass for any of the 20 natural amino acids.
  • the label should be robust under the fragmentation conditions of
  • Labeling chemistry should be efficient under a range of conditions, particularly denaturing conditions, and the labeled tag preferably remains soluble in the MS buffer system of choice.
  • the label preferably does not suppress the ionization efficiency of the protein and is not chemically reactive.
  • the label may contain a mixture of two or more isotopically distinct species to generate a unique mass spectrometric pattern at each labeled fragment position. Stable isotopes, such as 2 H, 13 C, 15 N, 17 0, 18 O, or 34 S, are among preferred labels. Pairs of peptide internal standards that incorporate a different isotope label may also be prepared. Preferred amino acid residues into which a heavy isotope label may be incorporated include leucine, proline, valine, and phenylalanine.
  • Peptide internal standards are characterized according to their mass-to-charge (m/z) ratio, and preferably, also according to their retention time on a chromatographic column (e.g. an HPLC column). Internal standards that co-elute with unlabeled peptides of identical sequence are selected as optimal internal standards.
  • the internal standard is then analyzed by fragmenting the peptide by any suitable means, for example by collision-induced dissociation (CID) using, e.g., argon or helium as a collision gas.
  • CID collision-induced dissociation
  • the fragments are then analyzed, for example by multi-stage mass spectrometry (MS”) to obtain a fragment ion spectrum, to obtain a peptide fragmentation signature.
  • MS multi-stage mass spectrometry
  • peptide fragments have significant differences in m/z ratios to enable peaks corresponding to each fragment to be well separated, and a signature that is unique for the target peptide is obtained. If a suitable fragment signature is not obtained at the first stage, additional stages of MS are performed until a unique signature is obtained.
  • Fragment ions in the MS/MS and MS 3 spectra are typically highly specific for the peptide of interest, and, in conjunction with LC methods, allow a highly selective means of detecting and quantifying a target peptide/protein in a complex protein mixture, such as a cell lysate, containing many thousands or tens of thousands of proteins.
  • a complex protein mixture such as a cell lysate, containing many thousands or tens of thousands of proteins.
  • Any biological sample potentially containing a target protein/peptide of interest may be assayed. Crude or partially purified cell extracts are preferably employed.
  • the sample has at least 0.01 mg of protein, typically a concentration of 0.1-10 mg/mL, and may be adjusted to a desired buffer concentration and pH.
  • a known amount of a labeled peptide internal standard, preferably about 10 femtomoles, corresponding to a target protein to be detected/quantified is then added to a biological sample, such as a cell lysate.
  • the spiked sample is then digested with one or more protease(s) for a suitable time period to allow digestion.
  • a separation is then performed (e.g. by HPLC, reverse-phase HPLC, capillary electrophoresis, ion exchange chromatography, etc.) to isolate the labeled internal standard and its corresponding target peptide from other peptides in the sample.
  • Microcapillar ⁇ LC is a preferred method.
  • Each isolated peptide is then examined by monitoring of a selected reaction in the MS. This involves using the prior knowledge gained by the characterization of the peptide internal standard and then requiring the MS to continuously monitor a specific ion in the MS/MS or MS" spectrum for both the peptide of interest and the internal standard. After elution, the area under the curve (AUC) for both peptide standard and target peptide peaks are calculated. The ratio of the two areas provides the absolute quantification that can be normalized for the number of cells used in the analysis and the protein's molecular weight, to provide the precise number of copies of the protein per cell. Further details of the AQUA methodology are described in Gygi et a/., and Gerber et a/, supra.
  • AQUA internal peptide standards may now be produced, as described above, for any of the 214 novel carcinoma-related signaling protein phosphorylation sites disclosed herein (see Table 1/ Figure 2).
  • Peptide standards for a given phosphorylation site e.g. the tyrosine 160 site in PAK5 kinase- see Row 111 of Table 1
  • PAK5 site sequence in Column E, Row 111 of Table 1 SEQ ID NO: 110
  • AQUA peptides of the invention may comprise all, or part of, a phosphorylation site peptide sequence disclosed herein (see Column E of Table 1/ Figure 2).
  • an AQUA peptide of the invention consists of, or comprises, a phosphorylation site sequence disclosed herein in Table 1/ Figure 2.
  • Heavy-isotope labeled equivalents of the peptides enumerated in Table 1/ Figure 2 can be readily synthesized and their unique MS and LC-SRM signature determined, so that the peptides are validated as AQUA peptides and ready for use in quantification experiments.
  • the phosphorylation site peptide sequences disclosed herein are particularly well suited for development of corresponding AQUA peptides, since the IAP method by which they were identified (see Part A above and Example 1) inherently confirmed that such peptides are in fact produced by enzymatic digestion (trypsinization) and are in fact suitably fractionated/ionized in MS/MS.
  • heavy-isotope labeled equivalents of these peptides can be readily synthesized and their unique MS and LC-SRM signature determined, so that the peptides are validated as AQUA peptides and ready for use in quantification experiments.
  • the invention provides heavy-isotope labeled peptides (AQUA peptides) for the detection and/or quantification of any of the carcinoma-related phosphorylation sites disclosed in Table 1/ Figure 2 (see Column E) and/or their corresponding parent proteins/polypeptides (see Column A).
  • a phosphopeptide sequence consisting of, or comprising, any of the phosphorylation sequences listed in Table 1 may be considered a preferred AQUA peptide of the invention.
  • AQUA peptides are within the scope of the present invention, and the selection and production of preferred AQUA peptides may be carried out as described above (see Gygi et al., Gerber et a/, supra.). Certain particularly preferred subsets of AQUA peptides provided by the invention are described above (corresponding to particular protein types/groups in Table 1 , for example, Kinases or Adaptor/Scaffold proteins).
  • Example 4 is provided to further illustrate the construction and use, by standard methods described above, of exemplary AQUA peptides provided by the invention.
  • the above-described AQUA peptides corresponding to the both the phosphorylated and non- phosphorylated forms of the disclosed Met kinase tyrosine 835 phosphorylation site may be used to quantify the amount of phosphorylated Met (Tyr 835) in a biological sample, e.g. a tumor cell sample (or a sample before or after treatment with a test drug).
  • AQUA peptides of the invention may also be employed within a kit that comprises one or multiple AQUA peptide(s) provided herein (for the quantification of a carcinoma-related signal transduction protein disclosed in Table 1/ Figure 2), and, optionally, a second detecting reagent conjugated to a detectable group.
  • a kit may include AQUA peptides for both the phosphorylated and non-phosphorylated form of a phosphorylation site disclosed herein.
  • the reagents may also include ancillary agents such as buffering agents and protein stabilizing agents, e.g., polysaccharides and the like.
  • the kit may further include, where necessary, other members of the signal-producing system of which system the detectable group is a member (e.g., enzyme substrates), agents for reducing background interference in a test, control reagents, apparatus for conducting a test, and the like.
  • the test kit may be packaged in any suitable manner, typically with all elements in a single container along with a sheet of printed instructions for carrying out the test.
  • AQUA peptides provided by the invention will be highly useful in the further study of signal transduction anomalies underlying cancer, including carcinomas, and in identifying diagnostic/bio-markers of these diseases, new potential drug targets, and/or in monitoring the effects of test compounds on carcinoma-related signal transduction proteins and pathways.
  • Antibodies provided by the invention may be advantageously employed in a variety of standard immunological assays (the use of AQUA peptides provided by the invention is described separately above). Assays may be homogeneous assays or heterogeneous assays. In a homogeneous assay the immunological reaction usually involves a phosphorylation-site specific antibody of the invention), a labeled analyte, and the sample of interest. The signal arising from the label is modified, directly or indirectly, upon the binding of the antibody to the labeled analyte. Both the immunological reaction and detection of the extent thereof are carried out in a homogeneous solution. Immunochemical labels that may be employed include free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, coenzymes, and so forth.
  • the reagents are usually the specimen, a phosphorylation-site specific antibody of the invention, and suitable means for producing a detectable signal. Similar specimens as described above may be used.
  • the antibody is generally immobilized on a support, such as a bead, plate or slide, and contacted with the specimen suspected of containing the antigen in a liquid phase.
  • the support is then separated from the liquid phase and either the support phase or the liquid phase is examined for a detectable signal employing means for producing such signal.
  • the signal is related to the presence of the analyte in the specimen.
  • Means for producing a detectable signal include the use of radioactive labels, fluorescent labels, enzyme labels, and so forth.
  • an antibody which binds to that site can be conjugated to a detectable group and added to the liquid phase reaction solution before the separation step.
  • the presence of the detectable group on the solid support indicates the presence of the antigen in the test sample.
  • suitable immunoassays are the radioimmunoassay, immunofluorescence methods, enzyme-linked immunoassays, and the like.
  • Immunoassay formats and variations thereof that may be useful for carrying out the methods disclosed herein are well known in the art. See generally E. Maggio, Enzyme-lmmunoassay, (1980) (CRC Press, Inc., Boca Raton, FIa.); see also, e.g., U.S. Pat. No. 4,727,022 (Skold etal., “Methods for Modulating Ligand-Receptor Interactions and their Application”); U.S. Pat. No. 4,659,678 (Forrest et al., "Immunoassay of Antigens"); U.S. Pat. No.
  • Phosphorylation site-specific antibodies disclosed herein may be conjugated to a solid support suitable for a diagnostic assay (e.g., beads, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as precipitation.
  • Antibodies, or other target protein or target site-binding reagents may likewise be conjugated to detectable groups such as radiolabels (e.g., 35 S, 125 1, 131 I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein) in accordance with known techniques.
  • radiolabels e.g., 35 S, 125 1, 131 I
  • enzyme labels e.g., horseradish peroxidase, alkaline phosphatase
  • fluorescent labels e.g., fluorescein
  • Antibodies of the invention may also be optimized for use in a flow cytometry (FC) assay to determine the activation/phosphorylation status of a target carcinoma-related signal transduction protein in patients before, during, and after treatment with a drug targeted at inhibiting phosphorylation at such a protein at the phosphorylation site disclosed herein.
  • FC flow cytometry
  • bone marrow cells or peripheral blood cells from patients may be analyzed by flow cytometry for target carcinoma-related signal transduction protein phosphorylation, as well as for markers identifying various hematopoietic cell types. In this manner, activation status of the malignant cells may be specifically characterized.
  • Flow cytometry may be carried out according to standard methods. See, e.g.
  • cytometric analysis may be employed: fixation of the cells with 1 % para- formaldehyde for 10 minutes at 37 0 C followed by permeabilization in 90% methanol for 30 minutes on ice. Cells may then be stained with the primary antibody (a phospho-specific antibody of the invention), washed and labeled with a fluorescent-labeled secondary antibody. Alternatively, the cells may be stained with a fluorescent-labeled primary antibody. The cells would then be analyzed on a flow cytometer (e.g. a Beckman Coulter EPICS-XL) according to the specific protocols of the instrument used.
  • a flow cytometer e.g. a Beckman Coulter EPICS-XL
  • IHC immunohistochemical staining to detect differences in signal transduction or protein activity using normal and diseased tissues.
  • IHC may be carried out according to well-known techniques. See, e.g., ANTIBODIES: A LABORATORY MANUAL, supra. Briefly, paraffin-embedded tissue (e.g.
  • tumor tissue is prepared for immunohistochemical staining by deparaffinizing tissue sections with xylene followed by ethanol; hydrating in water then PBS; unmasking antigen by heating slide in sodium citrate buffer; incubating sections in hydrogen peroxide; blocking in blocking solution; incubating slide in primary antibody and secondary antibody; and finally detecting using ABC avidin/biotin method according to manufacturer's instructions.
  • Antibodies of the invention may be also be optimized for use in other clinically-suitable applications, for example bead-based multiplex- type assays, such as IGEN, LuminexTM and/or BioplexTM assay formats, or otherwise optimized for antibody arrays formats, such as reversed- phase array applications (see, e.g. Paweletz et al., Oncogene 20(16): 1981-89 (2001)).
  • the invention provides a method for the multiplex detection of carcinoma-related protein phosphorylation in a biological sample, the method comprising utilizing two or more antibodies or AQUA peptides of the invention to detect the presence of two or more phosphorylated carcinoma-related signaling proteins enumerated in Column A of Table 1/ Figure 2.
  • two to five antibodies or AQUA peptides of the invention are employed in the method.
  • six to ten antibodies or AQUA peptides of the invention are employed, while in another preferred embodiment eleven to twenty such reagents are employed.
  • Antibodies and/or AQUA peptides of the invention may also be employed within a kit that comprises at least one phosphorylation site- specific antibody or AQUA peptide of the invention (which binds to or detects a carcinoma-related signal transduction protein disclosed in Table 1/ Figure 2), and, optionally, a second antibody conjugated to a detectable group.
  • the kit is suitable for multiplex assays and comprises two or more antibodies or AQUA peptides of the invention, and in some embodiments, comprises two to five, six to ten, or eleven to twenty reagents of the invention.
  • the kit may also include ancillary agents such as buffering agents and protein stabilizing agents, e.g., polysaccharides and the like.
  • the kit may further include, where necessary, other members of the signal-producing system of which system the detectable group is a member (e.g., enzyme substrates), agents for reducing background interference in a test, control reagents, apparatus for conducting a test, and the like.
  • the test kit may be packaged in any suitable manner, typically with all elements in a single container along with a sheet of printed instructions for carrying out the test.
  • IAP isolation techniques were employed to identify phosphotyrosine-containing peptides in cell extracts from the following human carcinoma cell lines and patient cell lines: H69 LS, A431, DMS153 NS, SW620, HT116, MDA_MB_468, MCF10, HPAC, HT29, H460 NS, HCT166, H526, H526, BxPC-3, Hs766T, Su.86.86,
  • H345, H209, H441 , H209, A549, MIAPACA2, LNCaP, H226, H69, A431, H460, H23, H1703, Hs766T, DU145, H345, HCT 116, and PANC-1 DU 145 (see Figure 2, Column G). Tryptic phosphotyrosine-containing peptides were purified and analyzed from extracts of each of the cell lines mentioned above, as follows. Cells were cultured in DMEM medium or RPMI 1640 medium supplemented with 10% fetal bovine serum and penicillin/streptomycin. Cells were harvested by low speed centrifugation.
  • cells were resuspended in 1 mL lysis buffer per 1.25 x 10 8 cells (20 mM HEPES pH 8.0, 9 M urea, 1 mM sodium vanadate, supplemented or not with 2.5 mM sodium pyrophosphate, 1 mM ⁇ -glycerol-phosphate) and sonicated.
  • Sonicated cell lysates were cleared by centrifugation at 20,000 x g, and proteins were reduced with DTT at a final concentration of 4.1 mM and alkylated with iodoacetamide at 8.3 mM.
  • protein extracts were diluted in 20 mM HEPES pH 8.0 to a final concentration of 2 M urea and soluble TLCK-trypsin (Worthington) was added at 10-20 ⁇ g/mL. Digestion was performed for 1-2 days at room temperature.
  • Trifluoroacetic acid was added to protein digests to a final concentration of 1%, precipitate was removed by centrifugation, and digests were loaded onto Sep-Pak Ci 8 columns (Waters) equilibrated with 0.1% TFA. A column volume of 0.7-1.0 ml was used per 2 x 10 8 cells. Columns were washed with 15 volumes of 0.1% TFA, followed by 4 volumes of 5% acetonitrile (MeCN) in 0.1% TFA. Peptide fraction I was obtained by eluting columns with 2 volumes each of 8, 12, and 15%
  • the phosphotyrosine monoclonal antibody P-Tyr-100 (Cell Signaling Technology, Inc., catalog number 9411) was coupled at 4 mg/ml beads to protein G (Roche), respectively.
  • Immobilized antibody (15 ⁇ l, 60 ⁇ g) was added as 1:1 slurry in IAP buffer to 1 ml of each peptide fraction, and the mixture was incubated overnight at 4° C with gentle rotation.
  • the immobilized antibody beads were washed three times with 1 ml IAP buffer and twice with 1 ml water, all at 4° C.
  • Peptides were eluted from beads by incubation with 75 ⁇ l of 0.1% TFA at room temperature for 10 minutes.
  • one single peptide fraction was obtained from Sep-Pak C18 columns by elution with 2 volumes each of 10%, 15%, 20 %, 25 %, 30 %, 35 % and 40 % acetonitirile in 0.1% TFA and combination of all eluates.
  • IAP on this peptide fraction was performed as follows: After lyophilization, peptide was dissolved in 1.4 ml IAP buffer (MOPS pH 7.2, 10 mM sodium phosphate, 50 mM NaCI) and insoluble matter was removed by centrifugation. Immobilized antibody (40 ⁇ l, 160 ⁇ g) was added as 1:1 slurry in IAP buffer, and the mixture was incubated overnight at 4° C with gentle shaking.
  • the immobilized antibody beads were washed three times with 1 ml IAP buffer and twice with 1 ml water, all at 4° C. Peptides were eluted from beads by incubation with 55 ⁇ l of 0.15% TFA at room temperature for 10 min (eluate 1), followed by a wash of the beads (eluate 2) with 45 ⁇ l of 0.15% TFA. Both eluates were combined.
  • IAP eluate 40 ⁇ l or more of IAP eluate were purified by 0.2 ⁇ l StageTips or ZipTips. Peptides were eluted from the microcolumns with 1 ⁇ l of 40% MeCN, 0.1% TFA (fractions I and II) or 1 ⁇ l of 60% MeCN 1 0.1% TFA
  • fraction III fraction III into 7.6 ⁇ l of 0.4% acetic acid/0.005% heptafluorobutyric acid.
  • 1 ⁇ l of 60% MeCN, 0.1% TFA was used for elution from the microcolumns. This sample was loaded onto a 10 cm x 75 ⁇ m PicoFrit capillary column (New Objective) packed with Magic C18 AQ reversed-phase resin (Michrom Bioresources) using a Famos autosampler with an inert sample injection valve (Dionex).
  • MS/MS spectra were evaluated using TurboSequest in the Sequest Browser package (v. 27, rev. 12) supplied as part of BioWorks 3.0 (ThermoFinnigan). Individual MS/MS spectra were extracted from the raw data file using the Sequest Browser program CreateDta, with the following settings: bottom MW, 700; top MW, 4,500; minimum number of ions, 20; minimum TIC, 4 x 10 5 ; and precursor charge state, unspecified. Spectra were extracted from the beginning of the raw data file before sample injection to the end of the eluting gradient. The IonQuest and VuDta programs were not used to further select MS/MS spectra for Sequest analysis.
  • MS/MS spectra were evaluated with the following TurboSequest parameters: peptide mass tolerance, 2.5; fragment ion tolerance, 0.0; maximum number of differential amino acids per modification, 4; mass type parent, average; mass type fragment, average; maximum number of internal cleavage sites, 10; neutral losses of water and ammonia from b and y ions were considered in the correlation analysis.
  • Proteolytic enzyme was specified except for spectra collected from elastase digests. Searches were performed against the NCBI human protein database (either as released on April 29, 2003 and containing 37,490 protein sequences or as released on February 23, 2004 and containing 27,175 protein sequences).
  • Cysteine carboxamidomethyiation was specified as a static modification, and phosphorylation was allowed as a variable modification on serine, threonine, and tyrosine residues or on tyrosine residues alone. It was determined that restricting phosphorylation to tyrosine residues had little effect on the number of phosphorylation sites assigned.
  • the last criterion is routinely employed to confirm novel site assignments of particular interest. All spectra and all sequence assignments made by Sequest were imported into a relational database. Assigned sequences were accepted or rejected following a conservative, two-step process. In the first step, a subset of high-scoring sequence assignments was selected by filtering for XCorr values of at least 1.5 for a charge state of +1 , 2.2 for +2, and 3.3 for +3, allowing a maximum RSp value of 10.
  • Assignments in this subset were rejected if any of the following criteria were satisfied: (i) the spectrum contained at least one major peak (at least 10% as intense as the most intense ion in the spectrum) that could not be mapped to the assigned sequence as an a, b, or y ion, as an ion arising from neutral-loss of water or ammonia from a b or y ion, or as a multiply protonated ion; (ii) the spectrum did not contain a series of /? or y ions equivalent to at least six uninterrupted residues; or (iii) the sequence was not observed at least five times in all the studies we have conducted (except for overlapping sequences due to incomplete proteolysis or use of proteases other than trypsin).
  • Polyclonal antibodies that specifically bind a carcinoma-related signal transduction protein only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1/ Figure 2) are produced according to standard methods by first constructing a synthetic peptide antigen comprising the phosphorylation site sequence and then immunizing an animal to raise antibodies against the antigen, as further described below. Production of exemplary polyclonal antibodies is provided below.
  • This peptide is then coupled to KLH and used to immunize animals to produce (and subsequently screen) phospho- specific HER3 (tyr 1159) polyclonal antibodies as described in Immunization/Screening below.
  • a synthetic phospho-peptide antigen as described in A-C above is coupled to KLH, and rabbits are injected intradermal ⁇ (ID) on the back with antigen in complete Freunds adjuvant (500 ⁇ g antigen per rabbit). The rabbits are boosted with same antigen in incomplete Freund adjuvant (250 ⁇ g antigen per rabbit) every three weeks. After the fifth boost, bleeds are collected. The sera are purified by Protein A-affinity chromatography by standard methods (see ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor, supra.).
  • the eluted immunoglobulins are further loaded onto a non-phosphorylated synthetic peptide antigen-resin Knotes column to pull out antibodies that bind the non-phosphorylated form of the phosphorylation site.
  • the flow through fraction is collected and applied onto a phospho-synthetic peptide antigen-resin column to isolate antibodies that bind the phosphorylated form of the site.
  • the bound antibodies i.e. antibodies that bind a phosphorylated peptide described in A-C above, but do not bind the non-phosphorylated form of the peptide
  • the bound antibodies i.e. antibodies that bind a phosphorylated peptide described in A-C above, but do not bind the non-phosphorylated form of the peptide
  • the isolated antibody is then tested for phospho-specificity using Western blot assay using an appropriate cell line that expresses (or overexpresses) target phospho-protein (i.e. phosphorylated HER3, GRB7 or Smoothelin), for example, A431 , and A549, respectively.
  • Cells are cultured in DMEM or RPMI supplemented with 10% FCS. Cell are collected, washed with PBS and directly lysed in cell lysis buffer. The protein concentration of cell lysates is then measured. The loading buffer is added into cell lysate and the mixture is boiled at 100 0 C for 5 minutes. 20 ⁇ l (10 ⁇ g protein) of sample is then added onto 7.5% SDS-PAGE gel.
  • a standard Western blot may be performed according to the lmmunoblotting Protocol set out in the CELL SIGNALING TECHNOLOGY, INC. 2003-04 Catalogue, p. 390.
  • the isolated phospho-specific antibody is used at dilution 1:1000. Phosphorylation-site specificity of the antibody will be shown by binding of only the phosphorylated form of the target protein.
  • Isolated phospho-specific polyclonal antibody does not (substantially) recognize the target protein when not phosphorylated at the appropriate phosphorylation site in the non-stimulated cells (e.g. HER3 is not bound when not phosphorylated at tyrosine 1159).
  • Monoclonal antibodies that specifically bind a carcinoma-related signal transduction protein only when phosphorylated at the respective phosphorylation site disclosed herein are produced according to standard methods by first constructing a synthetic peptide antigen comprising the phosphorylation site sequence and then immunizing an animal to raise antibodies against the antigen, and harvesting spleen cells from such animals to produce fusion hybridomas, as further described below. Production of exemplary monoclonal antibodies is provided below.
  • This peptide is then coupled to KLH and used to immunize animals and harvest spleen cells for generation (and subsequent screening) of phospho-specific monoclonal Cdc25A (tyr463) antibodies as described in Immunization/Fusion/Screening below.
  • TNF-R1 (tyrosine 401).
  • This peptide is then coupled to KLH and used to immunize animals and harvest spleen cells for generation (and subsequent screening) of phospho-specific monoclonal TNF-R1 (ty r 401 ) antibodies as described in Immunization/Fusion/Screening below.
  • This peptide is then coupled to KLH and used to immunize animals and harvest spleen cells for generation (and subsequent screening) of phospho-specific monoclonal Requiem (tyr 172) antibodies as described in Immunization/Fusion/Screening below.
  • a synthetic phospho-peptide antigen as described in A-C above is coupled to KLH, and BALB/C mice are injected intradermally (ID) on the back with antigen in complete Freunds adjuvant (e.g. 50 ⁇ g antigen per mouse). The mice are boosted with same antigen in incomplete Freund adjuvant (e.g. 25 ⁇ g antigen per mouse) every three weeks. After the fifth boost, the animals are sacrificed and spleens are harvested. Harvested spleen cells are fused to SP2/0 mouse myeloma fusion partner cells according to the standard protocol of Kohler and Milstein (1975).
  • Colonies originating from the fusion are screened by ELISA for reactivity to the phospho-peptide and non-phospho-peptide forms of the antigen and by Western blot analysis (as described in Example 1 above). Colonies found to be positive by ELISA to the phospho-peptide while negative to the non-phospho-peptide are further characterized by Western blot analysis. Colonies found to be positive by Western blot analysis are subcloned by limited dilution.
  • Mouse ascites are produced from a single clone obtained from subcloning, and tested for phospho- specificity (against the Cdc25A, TNF-R1 , or Requiem) phospho-peptide antigen, as the case may be) on ELISA.
  • Clones identified as positive on Western blot analysis using cell culture supernatant as having phospho- specificity, as indicated by a strong band in the induced lane and a weak band in the uninduced lane of the blot, are isolated and subcloned as clones producing monoclonal antibodies with the desired specificity.
  • Ascites fluid from isolated clones may be further tested by Western blot analysis.
  • the ascites fluid should produce similar results on Western blot analysis as observed previously with the cell culture supernatant, indicating phospho-specificity against the phosphorylated target (e.g. Requiem phosphorylated at tyrosine 172).
  • AQUA Peptides for the Quantification of carcinoma-related Signaling Protein Phosphorylation Heavy-isotope labeled peptides (AQUA peptides (internal standards)) for the detection and quantification of a carcinoma-related signal transduction protein only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1 / Figure 2) are produced according to the standard AQUA methodology (see Gygi et al., Gerber et a/., supra.) methods by first constructing a synthetic peptide standard corresponding to the phosphorylation site sequence and incorporating a heavy-isotope label.
  • the Met (tyr 835) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated Met (tyr 835) in the sample, as further described below in Analysis & Quantification.
  • the P130Cas (tyr 287) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated P130Cas (tyr 287) in the sample, as further described below in Analysis & Quantification.
  • the MAPI B (tyr 1062) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated MAPI B (tyr 1062) in the sample, as further described below in Analysis & Quantification.
  • Adolase A (tyrosine 363).
  • An AQUA peptide comprising the sequence
  • the Adolase A (tyr 363) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated Adolase A (tyr 363) in the sample, as further described below in Analysis & Quantification.
  • Fluorenylmethoxycarbonyl (Fmoc)-derivatized amino acid monomers may be obtained from AnaSpec (San Jose, CA). Fmoc-derivatized stable- isotope monomers containing one 15 N and five to nine 13 C atoms may be obtained from Cambridge Isotope Laboratories (Andover, MA). Preloaded Wang resins may be obtained from Applied Biosystems. Synthesis scales may vary from 5 to 25 ⁇ mol.
  • Amino acids are activated in situ with 1-H- benzotriazolium, i-bis(dimethylamino) methylene]-hexafluorophosphate (1-),3-oxide:1-hydroxybenzotriazole hydrate and coupled at a 5-fold molar excess over peptide. Each coupling cycle is followed by capping with acetic anhydride to avoid accumulation of one-residue deletion peptide byproducts. After synthesis peptide-resins are treated with a standard scavenger-containing trifluoroacetic acid (TFA)-water cleavage solution, and the peptides are precipitated by addition to cold ether.
  • Peptides i.e.
  • a desired AQUA peptide described in A-D above are purified by reversed- phase C18 HPLC using standard TFA/acetonitrile gradients and characterized by matrix-assisted laser desorption ionization-time of flight (Biflex III, Bruker Daltonics, Billerica, MA) and ion-trap (ThermoFinnigan, LCQ DecaXP) MS.
  • MS/MS spectra for each AQUA peptide should exhibit a strong y-type ion peak as the most intense fragment ion that is suitable for use in an SRM monitoring/analysis.
  • Reverse-phase microcapillary columns (0.1 A- 150-220 mm) are prepared according to standard methods.
  • An Agilent 1100 liquid chromatograph may be used to develop and deliver a solvent gradient [0.4% acetic acid/0.005% heptafluorobutyric acid (HFBA)/7% methanol and 0.4% acetic acid/0.005% HFBA/65% methanol/35% acetonitrile] to the microcapillary column by means of a flow splitter.
  • HFBA heptafluorobutyric acid
  • Samples are then directly loaded onto the microcapillary column by using a FAMOS inert capillary autosampler (LC Packings, San Francisco) after the flow split. Peptides are reconstituted in 6% acetic acid/0.01% TFA before injection.
  • Target protein e.g. a phosphorylated protein of A-D above
  • AQUA peptide as described above
  • the IAP method is then applied to the complex mixture of peptides derived from proteolytic cleavage of crude cell extracts to which the AQUA peptides have been spiked in.
  • LC-SRM of the entire sample is then carried out.
  • MS/MS may be performed by using a ThermoFinnigan (San Jose, CA) mass spectrometer (LCQ DecaXP ion trap or TSQ Quantum triple quadrupole).
  • parent ions are isolated at 1.6 m/z width, the ion injection time being limited to 150 ms per microscan, with two microscans per peptide averaged, and with an AGC setting of 1 x 10 8 ; on the Quantum, Q1 is kept at 0.4 and Q3 at 0.8 m/z with a scan time of 200 ms per peptide.
  • analyte and internal standard are analyzed in alternation within a previously known reverse-phase retention window; well-resolved pairs of internal standard and analyte are analyzed in separate retention segments to improve duty cycle.
  • Data are processed by integrating the appropriate peaks in an extracted ion chromatogram (60.15 m/z from the fragment monitored) for the native and internal standard, followed by calculation of the ratio of peak areas multiplied by the absolute amount of internal standard (e.g., 500 fmol).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne de nouveaux sites de phosphorylation identifiés dans des protéines de transduction de signal et dans des voies de tendance de carcinome humain et, fournit des anticorps spécifiques de sites de phosphorylation et des peptides étiquetés par des isotopes lourds (peptides AQUA) pour la détection et la quantification sélective de ces sites/protéines phosphorylés, ainsi que des techniques d'utilisation de ces réactifs à cet effet. Parmi les sites de phosphorylation identifiés, on distingue les sites survenant dans les types de protéine suivants : des protéines d'échafaudage/adaptateur, des protéines de cytosquelette, des protéine de signalisation GTP, des kinases, des protéines de métabolisme, des phosphatases/phospho-diesterases/protéases, des protéines réceptrices, des protéine de traitement ARN, des protéines de transcription, des protéines de traduction, des protéine transporteuses et des protéines d'ubiquitine, ainsi que d'autres types de protéines.
EP06739581A 2005-04-12 2006-03-24 Réactifs de détection de phosphorylation de protéines dans la voie de signalisation de carcinome Withdrawn EP1872134A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US67044705P 2005-04-12 2005-04-12
PCT/US2006/010868 WO2006113050A2 (fr) 2005-04-12 2006-03-24 Reactifs de detection de phosphorylation de proteines dans la voie de signalisation de carcinome

Publications (2)

Publication Number Publication Date
EP1872134A2 true EP1872134A2 (fr) 2008-01-02
EP1872134A4 EP1872134A4 (fr) 2010-09-01

Family

ID=37115632

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06739581A Withdrawn EP1872134A4 (fr) 2005-04-12 2006-03-24 Réactifs de détection de phosphorylation de protéines dans la voie de signalisation de carcinome

Country Status (2)

Country Link
EP (1) EP1872134A4 (fr)
WO (1) WO2006113050A2 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010008838A2 (fr) * 2008-06-23 2010-01-21 Perkinelmer Health Sciences, Inc. Substrats de kinases
CN111116734A (zh) * 2019-12-23 2020-05-08 维塔恩(广州)医药有限公司 肿瘤相关基因c-kit突变相关抗原短肽及其应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021230A1 (fr) * 1992-04-10 1993-10-28 Dana-Farber Cancer Institute, Inc. Immunodetection de phosphoproteine specifique a l'etat d'activation
WO2002081638A2 (fr) * 2001-04-06 2002-10-17 Origene Technologies, Inc Profils d'expression de cancer de la prostate
US20030108888A1 (en) * 2001-05-15 2003-06-12 Ludwig Institute For Cancer Research Breast cancer antigens

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021230A1 (fr) * 1992-04-10 1993-10-28 Dana-Farber Cancer Institute, Inc. Immunodetection de phosphoproteine specifique a l'etat d'activation
WO2002081638A2 (fr) * 2001-04-06 2002-10-17 Origene Technologies, Inc Profils d'expression de cancer de la prostate
US20030108888A1 (en) * 2001-05-15 2003-06-12 Ludwig Institute For Cancer Research Breast cancer antigens

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
AMAR L S ET AL: "Involvement of desmoplakin phosphorylation in the regulation of desmosomes by protein kinase C, in HeLa cells." CELL ADHESION AND COMMUNICATION 1999 LNKD- PUBMED:10427965, vol. 7, no. 2, 1999, pages 125-138, XP008124497 ISSN: 1061-5385 *
CELL SIGNALING TECHNOLOGY: "PHospho-Tyrosine MOuse mAb (P-Tyr-100)" 1 January 2005 (2005-01-01), XP002592671 Retrieved from the Internet: URL:http://www.cellsignal.com/pdf/9411.pdf [retrieved on 2010-07-19] *
KANNER S B: "MONOCLONAL ANTIBODIES TO INDIVIDUAL TYROSINE-PHOSPHORYLATED PROTEIN SUBSTRATES OF ONCOGENE-ENCODED TYROSINE KINASES" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES (PNAS), NATIONAL ACADEMY OF SCIENCE, US LNKD- DOI:10.1073/PNAS.87.9.3328, vol. 87, no. 9, 1 May 1990 (1990-05-01), pages 3328-3332, XP002002168 ISSN: 0027-8424 *
MANDELL J W: "Phosphorylation state-specific antibodies: applications in investigative and diagnostic pathology" AMERICAN JOURNAL OF PATHOLOGY, AMERICAN SOCIETY FOR INVESTIGATIVE PATHOLOGY, US, vol. 163, no. 5, 1 November 2003 (2003-11-01), pages 1687-1698, XP003017035 ISSN: 0002-9440 *
RUSH J ET AL: "Immunoaffinity profiling of tyrosine phosphorylation in cancer cells" NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US LNKD- DOI:10.1038/NBT1046, vol. 23, no. 1, 1 January 2005 (2005-01-01), pages 94-101, XP002322741 ISSN: 1087-0156 *
See also references of WO2006113050A2 *

Also Published As

Publication number Publication date
EP1872134A4 (fr) 2010-09-01
WO2006113050A3 (fr) 2008-12-24
WO2006113050A8 (fr) 2007-12-27
WO2006113050A2 (fr) 2006-10-26
WO2006113050A4 (fr) 2009-03-26

Similar Documents

Publication Publication Date Title
EP1718760B1 (fr) Phosphorylation des proteines dans les voies de signalisation c-src
EP1929003A2 (fr) Reactifs de detection de phosporylation proteinique dans des voies de signalisation de carcinome
WO2007133702A2 (fr) Réactifs pour la détection des chemins de signalisation de l'acétylation des protéines
US7807789B2 (en) Reagents for the detection of protein phosphorylation in EGFR-signaling pathways
WO2007027906A2 (fr) Reactifs de detection de phosphorylation proteinique dans des voies de signalisation de leucemie
WO2007127335A2 (fr) Réactifs pour la détection de la phosphorylation de protéines dans les voies de signalisation des kinases atm et atr
EP2126580A2 (fr) Reactifs permettant de detecter une phosphorylation de proteines dans des voies de signalisation de leucemie
US20090258442A1 (en) Reagents for the detection of protein phosphorylation in carcinoma signaling pathways
WO2008009000A2 (fr) Réactifs pour la détection de phosphorylation de protéines dans les chemins de signalisation
US20100151495A9 (en) Reagents for the detection of protein phosphorylation in carcinoma signaling pathways
US20090263832A1 (en) Reagents for the Detection of Protein Phosphorylation in Leukemia Signaling Pathways
US20110105732A1 (en) Reagents for the Detection of Protein Phosphorylation in Carcinoma Signaling Pathways
WO2007133689A2 (fr) Réactifs pour la détection des chemins de signalisation de l'acétylation des protéines
US20090203034A1 (en) Reagents for the detection of tyrosine phosphorylation in brain ischemia signaling pathways
US20100173322A1 (en) Reagents for the detection of protein phosphorylation in anaplastic large cell lymphoma signaling pathways
WO2006068640A1 (fr) Phosphorylation des protéines suivant des voies contrôlées par les egfr
WO2006113050A2 (fr) Reactifs de detection de phosphorylation de proteines dans la voie de signalisation de carcinome
US7935790B2 (en) Reagents for the detection of protein phosphorylation in T-cell receptor signaling pathways
WO2007027916A2 (fr) Reactifs de detection de la phosphorylation proteinique dans la voie de signalisation de carcinome
US7939636B2 (en) Reagents for the detection of protein phosphorylation in c-Src signaling pathways
WO2007030792A2 (fr) Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules
CA2616588A1 (fr) Reactifs pour la detection d'atr kinase phosphorylee (ser 428/ser 2317) et utilisations
US20090142777A1 (en) Reagents for the detection of protein phosphorylation in leukemia signaling pathways

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20071109

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RIN1 Information on inventor provided before grant (corrected)

Inventor name: POLAKIEWICZ, ROBERTO

Inventor name: LEE, KIMBERLY

Inventor name: FARNSWORTH, CHARLES

Inventor name: LI, YU

Inventor name: MORITZ, ALBRECHT

Inventor name: RIKOVA, KLARISA

Inventor name: GUO, AILAN

DAX Request for extension of the european patent (deleted)
R17D Deferred search report published (corrected)

Effective date: 20081224

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/00 20060101ALI20090210BHEP

Ipc: G01N 33/53 20060101AFI20090210BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 7/00 20060101ALI20100722BHEP

Ipc: C07K 16/44 20060101ALI20100722BHEP

Ipc: C07K 16/00 20060101ALI20100722BHEP

Ipc: G01N 33/53 20060101AFI20090210BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20100803

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20101001