EP1929296A2 - Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules - Google Patents

Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules

Info

Publication number
EP1929296A2
EP1929296A2 EP06803284A EP06803284A EP1929296A2 EP 1929296 A2 EP1929296 A2 EP 1929296A2 EP 06803284 A EP06803284 A EP 06803284A EP 06803284 A EP06803284 A EP 06803284A EP 1929296 A2 EP1929296 A2 EP 1929296A2
Authority
EP
European Patent Office
Prior art keywords
rows
corresponding column
phosphorylated
protein
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Withdrawn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
EP06803284A
Other languages
German (de)
English (en)
Inventor
Roberto Polakiewicz
Kimberly Lee
Charles Farnsworth
Ailan Guo
Albrecht Moritz
Eric Spek
Francesco Boccalatte
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 EP1929296A2 publication Critical patent/EP1929296A2/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Withdrawn legal-status Critical Current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
    • G01N33/57407Specifically defined cancers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/008Peptides; Proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase

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 criticahrole in the etiology of many pathological conditions and diseases, including cancer, developmental disorders, autoimmune diseases, and diabetes.
  • protein modification it is not yet well understood at the molecular level. The reasons for this lack of understanding are, first, that the cellular modification system is extraordinarily complex, and second, that the technology necessary to unravel its complexity has not yet been fully developed.
  • the complexity of protein modification, including phosphorylation, on a proteome-wide scale derives from 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 encodes, for example, over 520 different protein kinases, making them the most abundant class of enzymes known. See Hunter, Nature 411: 355-65 (2001).
  • Each of these kinases phosphorylates specific serine, threonine, or tyrosine residues located within distinct amino acid sequences, or motifs, contained within different protein substrates.
  • Most kinases phosphorylate many different proteins: 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. See Graves et a/., Pharmacol. Ther. 82: 111-21 (1999).
  • motif-specific, context-independent antibodies which are capable of specifically binding short, recurring signaling motifs comprising one or more modified (e.g. phosphorylated) amino acids in many different proteins in which the motif recurs.
  • modified (e.g. phosphorylated) amino acids in many different proteins in which the motif recurs.
  • ALCL Anaplastic Large-Cell Lymphoma
  • NHL non-Hodgkin's lymphomas
  • ALCL a form of T-cell lymphoma (CD30+), is most prevalent among young children, representing about 15% of all pediatric non-Hodgkin's lymphomas (source: UMDNJ Hematopathology (2004)). It is an aggressive disease that can be either systemic or primary cutaneous, with median survival rates of about 5 years from diagnosis. - A -
  • NPM-ALK A number of downstream signaling protein targets of NPM-ALK have identified as potentially involved in mediating cellular transformation in NPM-ALK positive ALCL, including She, IRS-1 , Grb2, phospholipase C- ⁇ , PI3-kinase, and Stat3/5. See Ouyang et al. supra; Zamo et ai, Oncogene 21: 1038-1047 (2002). NPM-ALK activates the AKT/PI3K anti- apoptotic signaling pathway. See Bai et ai, Blood 96: 4319-4327 (2000). Transgenic mice experiments have established that Stat3 and Jak3 are constitutively activated in NPM-ALK positive transgenic mice that develop ALCL.
  • PLCgamma phospholipase gamma
  • SHC and IRS-1 are known to be phosphorylated in the context of other signaling cascades (such as the Ras/ERk pathway) and many of their phosphorylation sites have been identified.
  • diagnosis of ALCL is made by tissue biopsy and detection of T-cell markers, such as CD30 and/or CD4.
  • T-cell markers such as CD30 and/or CD4.
  • misdiagnosis can occur since some ALCL can be negative for certain markers and/or can be positive for keratin, a marker for carcinoma.
  • the NPM-ALK genetic translocation itself can be detected, it is clear that other downstream effectors of ALCL, having diagnostic, predictive, or therapeutic value, remain to be elucidated. Accordingly, identification of downstream signaling molecules and phospho-sites involved in NPM-ALK positive ALCL 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 nearly 219 novel phosphorylation sites identified in signal transduction proteins and pathways underlying Anaplastic Large Cell Lymphoma (ALCL) involving the NPM-ALK translocation/fusion, 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 quantification and profiling of the disclosed phosphorylation sites.
  • ACL Anaplastic Large Cell Lymphoma
  • FIG. 1 - I is a diagram broadly depicting the immunoaffinity isolation and mass-spectrometric characterization methodology (IAP) employed to identify the novel phosphorylation sites disclosed herein.
  • IAP immunoaffinity isolation and mass-spectrometric characterization methodology
  • FIGURE 3 - is an exemplary mass spectrograph depicting the detection of the tyrosine 1284 phosphorylation site in ALK (see Row 109 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).
  • FIGURE 4 - is an exemplary mass spectrograph depicting the detection of the tyrosine 406 phosphorylation site in ARGHEF2 (see Row 79 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).
  • FIGURE 5 - is an exemplary mass spectrograph depicting the detection of the tyrosine 111 phosphorylation site in IRS4 (see Row 12 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.
  • FIGURE 6 - is an exemplary mass spectrograph depicting the detection of the tyrosine 466 phosphorylation site in PKM2 (see Row 65 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).
  • FIGURE 7 - is an exemplary mass spectrograph depicting the detection of the tyrosine 284 phosphorylation site in PPP2CB (see Row 127 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).
  • FIGURE 8 - is an exemplary mass spectrograph depicting the detection of the tyrosine 588 phosphorylation site in ACLY (see Row 76 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).
  • FIGURE 9 - is an exemplary mass spectrograph depicting the detection of the tyrosine 3914 phosphorylation site in MLL (see Row 170 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).
  • the invention provides novel reagents - phospho-specific antibodies and AQUA peptides - for the specific detection and/or quantification of an ALCL- related signaling protein/polypeptide only when phosphorylated (or only when not phosphorylated) at a particular phosphorylation site disclosed herein.
  • the invention also provides methods of detecting and/or quantifying one or more phosphorylated ALCL-related signaling proteins using the phosphorylation-site specific antibodies and AQUA peptides of the invention, and methods of obtaining a phosphorylation profile of such proteins (e.g. Kinases).
  • the invention provides an isolated phosphorylation site- specific antibody that specifically binds a given ALCL-related signaling protein only when phosphorylated (or not phosphorylated, respectively) at a particular amino acid 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 quantification of a given ALCL-related signaling protein, the labeled peptide comprising a particular phosphorylatable peptide site/sequence enumerated in Column E of Table 1/ Figure 2 herein.
  • reagents provided by the invention is an isolated phosphorylation site-specific antibody that specifically binds the MAPK6 protein only when phosphorylated (or only when not phosphorylated) at tyrosine 628 (see Row 100 (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 MAPK6 protein, the AQUA peptide comprising the phosphorylatable peptide sequence listed in Column E, Row 100, of Table 1/ Figure 2.
  • the invention provides an isolated phosphorylation site-specific antibody that specifically binds an Anaplastic Large Cell Lymphoma (ALCL)-related signaling protein selected from Column A of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D of Table 1 , comprised within the peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 1-15, 17-39, 41-48, 50-64, 66-107, 109-148, 151-191 , 193-215, 217-219), wherein said antibody does not bind said signaling protein when not phosphorylated at said tyrosine.
  • ACL Anaplastic Large Cell Lymphoma
  • the invention provides an isolated phosphorylation site-specific antibody that specifically binds an ALCL-related signaling protein selected from Column A of Table 1 only when not phosphorylated at the tyrosine 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- 15, 17-39, 41-48, 50-64, 66-107, 109-148, 151-191 , 193-215, 217-219), 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 an ALCL-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: 1-15, 17-39, 41-48, 50-64, 66-107, 109-148, 151-191 , 193-215, 217-219), 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 tyrosine within the labeled peptide is not phosphorylated.
  • Reagents (antibodies and AQUA peptides) provided by the invention may conveniently be grouped by the type of ALCL-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: Acetyltransferases, Actin Binding Proteins, Adaptor/Scaffold Proteins, Adhesion Proteins, Cell Cycle Regulation Proteins, Cell Surface Proteins, Channel Proteins, Chaperone Proteins, Chemokine Proteins, Cytokines, Cytoskeletal Proteins, DNA Binding Proteins, DNA Repair Proteins, Cellular Metabolism and Miscellaneous Enzymes, GTPase Activating Proteins, Guanine Nucleotide Exchange Factors, Helicases, Hydrolases, Inhibitor Proteins, Kinase Proteins, Ligase Proteins, Lipid Binding Proteins, Mit
  • Each of these distinct protein groups is considered a preferred subset of ALCL-related signal transduction protein phosphorylation sites disclosed herein, and reagents for their detection/quantification may be considered a preferred subset of reagents provided by the invention.
  • 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: Protein Kinase Proteins, Adaptor/Scaffold Protein(s), Phosphatase Proteins, Transcription Factor/Transcription Initiation Complex Proteins, Transferase Proteins, Ubiquitin Conjugating System Proteins, Oxidoreductase Proteins, Receptor Proteins, RNA binding Proteins, and Enzymes.
  • 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.
  • a heavy-isotope labeled peptide for the quantification of a Protein Kinase selected from Column A, Rows 89-109, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 89-109, of Table 1 (SEQ ID NOs: 89-107, 109), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 89-109, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Kinase Protein phosphorylation sites are particularly preferred: CSNK2B (Y108), PCTK3 (Y155), BMX (Y197), and ACS (Y524), (see SEQ ID NOs: 97, 101 , 109 and 110).
  • antibodies and AQUA peptides for the detection/quantification of the following Adaptor/Scaffold Protein phosphorylation sites are particularly preferred: CBL (Y114), GAB3 (Y542) and IRS4 (Y112), (see SEQ ID NOs: 8, 10, and 13).
  • a heavy-isotope labeled peptide for the quantification of a Phosphatase Protein selected from Column A, Rows 125-129, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 125-129, of Table 1 (SEQ ID NOs: 125-129), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 125-129, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Phosphatase Protein phosphorylation sites are particularly preferred: PPP2CB (Y284), and PTPN11 (Y66), (see SEQ ID NOs: 127 and 128).
  • a heavy-isotope labeled peptide (AQUA peptide) for the quantification of a Transcription Factor/Transcription Initiation Complex Protein selected from Column A, Rows 167-189, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 167-189, of Table 1 (SEQ ID NOs: 167-189), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 167-189, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Transcription Factor/Transcription Initiation Complex Protein phosphorylation sites are particularly preferred: GATA6 (Y417), MLL (Y3914), POLR2A (Y1881)and TP53BP2 (Y487), (see SEQ ID NOs: 168, 170, 180, and 188).
  • antibodies and AQUA peptides for the detection/quantification of the following Transferase Protein phosphorylation sites are particularly preferred: ATIC (Y192), (see SEQ ID NO: 191).
  • antibodies and AQUA peptides for the detection/quantification of the following Ubiquitin Conjugating System Protein phosphorylation sites are particularly preferred: DTX3L (Y235) and USP11 (Y870), (see SEQ ID NOs: 212 and 214).
  • a heavy-isotope labeled peptide for the quantification of an Oxidoreductase Protein selected from Column A, Rows 117-124, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 117-124, of Table 1 (SEQ ID NOs: 117-124), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 117-124, of Table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Oxidoreductase Protein phosphorylation site is particularly preferred: GSR (Y67) (see SEQ ID NO: 117).
  • ADRA2B (Y120) (see SEQ ID NO: 142).
  • antibodies and AQUA peptides for the detection/quantification of the following RNA Binding Protein phosphorylation site is particularly preferred: NUP160 (Y355) (see SEQ ID NO: 156).
  • a heavy-isotope labeled peptide for the quantification of an Enzyme selected from Column A, Rows 62-76, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 62-76, of Table 1 (SEQ ID NOs: 62-64, 66-76), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 62-76, of table 1.
  • antibodies and AQUA peptides for the detection/quantification of the following Enzyme phosphorylation sites are particularly preferred: ACLY (Y588) (see SEQ ID NO: 76).
  • an isolated phosphorylation site-specific antibody that specifically binds a protein selected from Column A, Rows 32, 78, 79 and 211 , of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 32, 78, 79 and 211 , of Table 1 , comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 32, 78, 79 and 211 , of Table 1 (SEQ ID NOs: 32, 78, 79 and 211), 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.
  • the reagents comprise a subset of preferred reagents as described above.
  • Also provided by the invention is a method for obtaining a phosphorylation profile of protein kinases that are phosphorylated in Carcinoma signaling pathways, said method comprising the step of utilizing one or more isolated antibody that specifically binds a protein kinase selected from Column A, Rows 138-165, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows138-165, of Table 1 , comprised within the phosphorylation site sequence listed in corresponding Column E, Rows 138-165, of Table 1 (SEQ ID NOs: 137-154, and 156-164), to detect the phosphorylation of one or more of said protein kinases, thereby obtaining a phosphorylation profile for said kinases.
  • 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 antibodies.
  • ALCL-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 Anaplastic Large Cell Lymphoma (ALCL) cell line(s).
  • An ALCL-related signaling protein may also be phosphorylated in other non-ALCL cell lines.
  • 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 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). Subsequently, (e) a search program (e.g.
  • 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.
  • 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 ALCL cell extracts.
  • lysates were prepared from both cell lines and digested with trypsin after treatment with DTT and iodoacetamide to alkylate cysteine residues.
  • peptides were prefractionated 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 MOPS buffer and treated with phosphotyrosine 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 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 revefsed-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 an ALCL-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 Column D of Table 1 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.
  • a new MAPK6 phosphorylation sites tyrosine 628) (see Row 100 of Table 1) are presently disclosed.
  • antibodies that specifically bind this novel MAPK6 site can now be produced by using (all or part of) the amino acid sequence encompassing the respective phosphorylated residue as a peptide antigen used to immunize an animal (e.g.
  • a peptide antigen comprising the sequence set forth in Row 100, Column E, of Table 1 (which encompasses the phosphorylated tyrosine as position 628 in MAPK6) may be employed to produce an antibody that only binds MAPK6 when phosphorylated at tyr628).
  • 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 ALCL-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 E 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 ALCL-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 E of Table 1
  • immune serum i.e. a phosphorylation site enumerated in Column E of Table 1 , which comprises the corresponding phospho
  • a peptide comprising any 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 consist of the full sequence disclosed in Column E of Table 1 , 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”).
  • 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. 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. Sci., 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 CBLB tyrosine 276 phosphorylation site sequence disclosed in Row 23, Column E of Table 1), and antibodies of the invention thus specifically bind a target ALCL 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. Nat'l. 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 ef a/.)
  • the invention also provides immortalized cell lines that produce an antibody of the invention.
  • immortalized cell lines that produce an antibody of the invention.
  • hybridoma clones, constructed as described above, that produce monoclonal antibodies to the ALCL-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.
  • 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). For example, 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.
  • 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 ALCL-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.
  • Phosphorylation-site specific antibodies of the invention may exhibit some limited cross-reactivity 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 ALCL-related signaling protein epitope for which the antibody of the invention is specific.
  • polyclonal antisera may be exhibit some undesirable general cross-reactivity to phosphotyrosine, which may be removed by further purification of antisera, e.g. over a phosphotyramine column.
  • Antibodies of the invention specifically bind their target protein (i.e. 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 Column D, 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) staining using normal and diseased tissues to examine ALCL- related phosphorylation and activation status in diseased tissue.
  • IHC immunohistochemical
  • 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 etal., 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 an ALCL-relat ⁇ d 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 ALCL-related signal transduction protein only when phosphorylated at a disclosed site, but are not limited only to binding the human species, per se.
  • the invention includes antibodies that also bind conserved and highly-homologous phosphorylation sites in respective ALCL-related proteins from other species (e.g. mouse, rat, monkey, yeast), in addition to binding the human phosphorylation site. Highly- homologous sites conserved in other species can readily be identified by standard sequence comparisons, such as using BLAST, with the human ALCL-signal transduction protein phosphorylation sites disclosed herein.
  • the novel ALCL-signaling protein phosphorylation sites disclosed herein now enable the production of corresponding heavy-isotope labeled peptides for the absolute quantification of such signaling proteins (both phosphorylated and not phosphorylated at a disclosed site) in biological samples.
  • the production and use of AQUA peptides for the absolute quantification of proteins (AQUA) in complex mixtures has been described. See WO/03016861 , "Absolute Quantification of Proteins and Modified Forms Thereof by Multistage Mass Spectrometry," Gygi et al. and also Gerber et al. Proc. Natl. Acad. ScL U.S.A. 100: 6940-5 (2003) (the teachings of which are hereby incorporated herein by reference, in their entirety).
  • 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 mass shift.
  • the 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
  • 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 in 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- phosphorylated form of the residue developed.
  • the two standards may be used to detect and quantify both the phosphorylated 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), metallo 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 fragments 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 MS and not undergo unfavorable fragmentation. 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 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 n ) to obtain a fragment ion spectrum, to obtain a peptide fragmentation signature.
  • MS n 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 is 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.
  • Microcapillary 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 n 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 al., and Gerber et al. supra.
  • AQUA internal peptide standards may now be produced, as described above, for any of the 219 novel ALCL-related signaling protein phosphorylation sites disclosed herein (see Table 1/ Figure 2).
  • Peptide standards for a given phosphorylation site e.g. the tyrosine 184 site in CAMK1 - see Row 96 of Table 1
  • Peptide standards for a given phosphorylation site may be produced for both the phosphorylated and non-phosphorylated forms of the site (e.g. see CAMK1 site sequence in Column E, Row 96 of Table 1) and such standards employed in the AQUA methodology to detect and quantify both forms of such phosphorylation site in a biological sample.
  • 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 ALCL-related phosphorylation sites disclosed in Table 1 (see Column E) and/or their corresponding parent proteins (see Column A).
  • Each such phosphorylation sequence may be considered a preferred AQUA peptide of the invention.
  • an AQUA peptide of the invention consists of a phosphorylation site sequence enumerated in Table 1.
  • tyrosine 111 being the phosphorylatable residue within the site.
  • a larger AQUA peptide comprising the disclosed phosphorylation site sequence (and additional residues downstream or upstream of it) may also be constructed.
  • AQUA peptide comprising less than all of the residues of a disclosed phosphorylation site sequence (but still comprising the phosphorylatable residue enumerated in Column D) may alternatively be constructed.
  • Such larger or shorter 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 al. supra.).
  • AQUA peptides provided by the invention are described above (corresponding to particular protein types/groups in Table 1 , for example, Kinase Proteins 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.
  • AQUA peptides corresponding to the both the phosphorylated and non-phosphorylated forms of the disclosed IRS4 tyrosine 111 phosphorylation site (LETADAPARLEyYENAR (SEQ ID NO: 12) - see Row 12 of Table 1/ Figure 2) may be used to quantify the amount of phosphorylated IRS4 (tyr111) in biological sample, e.g. an ALCL 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 an ALCL-related signal transduction protein disclosed in Table 1), and, optionally, a second detecting reagent conjugated to a detectable group.
  • a kit may include AQUA peptides for both the phosphorylation 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 ALCL, and in identifying diagnostic/bio-markers of this disease, new potential drug targets, and/or in monitoring the effects of test compounds on ALCL- 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 et al., "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.
  • ALCL-related 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 assay to determine the activation/phosphorylation status of a target ALCL-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.
  • a flow cytometry assay to determine the activation/phosphorylation status of a target ALCL-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.
  • bone marrow cells or peripheral blood cells from patients may be analyzed by flow cytometry for target ALCL-related 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 % 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 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. Such an analysis would identify the presence of activated ALCL-related signal transduction protein(s)elated in the malignant cells and reveal the drug response on the targeted protein.
  • a flow cytometer e.g. a Beckman Coulter EPICS-XL
  • antibodies of the invention may be employed in immunohistochemical (IHC) staining to detect differences in signal transduction or protein activity using normal and diseased ALCL 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)).
  • bead-based multiplex- type assays such as IGEN, LuminexTM and/or BioplexTM assay formats
  • antibody arrays formats such as reversed- phase array applications
  • the invention provides a method for the multiplex detection of ALCL-related protein phosphorylation in a biological sample, the method comprising utilizing at two or more antibodies or AQUA peptides of the invention to detect the presence of two or more phosphorylated ALCL-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 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 an ALCL-related signal transduction protein disclosed in Table 1), 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.
  • ALCL anaplastic large cell lymphomas
  • Tryptic phosphotyrosine peptides were purified and analyzed from extracts of the two ALCL cell lines as follows. Cells were grown in a 5% CO 2 incubator at 37 0 C. Cells were cultured to a density of 0.5-1.4 x 10 6 cells/ml in RPMI 1640 medium containing 10% calf serum. Cells were washed with PBS at 4° C, resuspended at 1.25 x 10 8 cells/ml in lysis buffer (20 mM HEPES pH 8.0, 9 M urea, 1 mM sodium vanadate) and sonicated. In some experiments, the PBS wash step was omitted.
  • 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 immobilized TLCK-trypsin (Pierce) was added at 1-2.5 ml beads (200 TAME units trypsin/ml) per 10 9 cells.
  • lysates were diluted in 20 mM HEPES pH 8.0 to a final concentration of 1 M urea, and GIuC (Worthington Biochemicals) or elastase (Roche) was added at 0.5 mg per 10 9 cells.
  • GIuC Wood Biochemicals
  • elastase Roche
  • Chymotrypsin was added at 10 mg per 10 9 cells. 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% MeCN in 0.1% TFA and combining the eluates. Fractions Il and III were a combination of eluates after eluting columns with 18, 22, 25% MeCN in 0.1% TFA and with 30, 35, 40% MeCN in 0.1% TFA, respectively. All peptide fractions were lyophilized.
  • Peptides from each fraction corresponding to 2 x 10 8 cells were dissolved in 1 ml of IAP buffer (20 mM Tris/HCI or 50 mM MOPS pH 7.2, 10 mM sodium phosphate, 50 mM NaCI) and insoluble matter (mainly in peptide fractions III) was removed by centrifugation. IAP was performed on each peptide fraction separately.
  • the phosphotyrosine monoclonal antibody P-Tyr-100 (Cell Signaling Technology, Inc., catalog number 9411) was coupled at 4 mg/ml beads to protein G agarose (Roche).
  • 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 min.
  • a thin layer of ⁇ -cyano-4-hydroxy-cinnamic acid (ACHA) matrix was applied to a Bruker 384-spot MALDI target by spreading 5 ⁇ l of a saturated solution in MeCN/water (2/1 , v/v) over an entire row of spots on the target; drying occurred in 2-5 sec.
  • the IAP eluate (10 ⁇ l) was loaded onto an 0.2 ⁇ l C-18 ZipTip (Millipore), which then was washed with 5% formic acid.
  • Peptide was eluted with 1 ⁇ l of 10 mg/ml ACHA in 60% methanol, 5% formic acid onto the MALDI target containing the thin layer of matrix. Samples were analyzed on a Bruker BiFlex III MALDI-TOF instrument in positive ion mode.
  • 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.
  • NCBI RefSeq protein release #11 8 May 2005; 1 ,826,611 proteins, including 47,859 human proteins.
  • Peptides that did not match RefSeq were compared to NCBI GenPept release #148; 15 June 2005 release date; 2,479,172 proteins, including 196,054 human proteins.
  • Cysteine carboxamidomethylation 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.
  • certain peptides were originally isolated in mouse and later normalized to human sequences as shown by Table1/ Figure2.
  • Sequest scoring thresholds were used to select phosphopeptide assignments that are likely to be correct: RSp ⁇ 6, XCorr > 2.2, and DeltaCN > 0.099. Further, the assigned sequences could be accepted or rejected with respect to accuracy by using the following conservative, two-step process.
  • a subset of high-scoring sequence assignments should be 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 should be rejected if any of the following criteria were satisfied: (i) the spectrum contains at least one major peak (at least 10% as intense as the most intense ion in the spectrum) that can 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 does not contain a series of b or y ions equivalent to at least six uninterrupted residues; or (iii) the sequence is not observed at least five times in all the studies conducted (except for overlapping sequences due to incomplete proteolysis or use of proteases other than trypsin).
  • Polyclonal antibodies that specifically bind an ALCL-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, as further described below. Production of exemplary polyclonal antibodies is provided below.
  • MAPK6 (tyrosine 628).
  • 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, are eluted and kept in v antibody storage buffer.
  • the isolated antibody is then tested for phospho-specificity using Western blot assay using an appropriate cell line the expresses (or overexpresses) target phospho-protein (i.e. phosphorylated MAPK6, GAB3 or PPP2CB).
  • Cells are cultured in DMEM supplemented with 10% FCS and 5U/ml IL-3. Before stimulation, the cells are starved in serum- free DMEM medium for 4 hours. The cells are then stimulated ligand (e.g. 50 ng/ml) for 5 minutes. Cell are collected, washed with PBS and directly lysed in cell lysis buffer. The protein concentration of cell lysates are 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.
  • phosphorylated MAPK6, GAB3 or PPP2CB phosphorylated MAPK6, GAB3 or PPP2
  • a standard Western blot may be performed according to the lmmunoblotting Protocol set out in the CELL SIGNALING TECHNOLOGY, ING. 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 recognize the target protein when not phosphorylated at the appropriate phosphorylation site in the non-stimulated cells (e.g., MAPK6 is not bound when not phosphorylated at tyrosine 628).
  • Monoclonal antibodies that specifically bind an ALCL-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.
  • A. CAMK1 (tyrosine 184).
  • 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 CAMK4(tyr184) 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 IRS4(tyr111) 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 PTPN11 (tyr66) 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 intradermal ⁇ (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.
  • ID intradermal ⁇
  • complete Freunds adjuvant e.g. 50 ⁇ g antigen per mouse
  • incomplete Freund adjuvant e.g. 25 ⁇ g antigen per mouse
  • 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 CAMK1 , IRS4, or PTPN11 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. CAMK1 phosphorylated at tyrosine 184).
  • Heavy-isotope labeled peptides for the detection and quantification of an ALCL-related signal transduction protein only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1) are produced according to the standard AQUA methodology (see Gygi et a/., Gerber et al., supra.) methods by first constructing a synthetic peptide standard corresponding to the phosphorylation site sequence and incorporating a heavy-isotope label. Subsequently, the MS" and LC-SRM signature of the peptide standard is validated, and the AQUA peptide is used to quantify native peptide in a biological sample, such as a digested cell extract. Production and use of exemplary AQUA peptides is provided below.
  • An AQUA peptide having a sequence corresponding to the tyrosine 334 phosphorylation site in human BMX 1 ILPQYDSySKKSCGS (y* phosphotyrosine) (see Row 107 in Table 1 (SEQ ID NO: 107)) but incorporating 14 C/ 15 N-labeled leucine (indicated by bold L) is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer (see Merrifield, supra.) as further described below in Synthesis & MS/MS Signature.
  • the BMX(tyr194) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated BMX (tyr194) in the sample, as further described below in Analysis & Quantification.
  • An AQUA peptide having a sequence corresponding to the tyrosine 858 phosphorylation site in human CBLB, ARLQKySTKPGSYIFR (y* phosphotyrosine) (see Row 23 in Table 1 (SEQ ID NO: 23)) but incorporating 14 C/ 15 N-labeled proline (indicated by bold P) is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer (see Merrifield, supra.) as further described below in Synthesis & MS/MS Signature.
  • the CBLB(tyr276) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated CBLB (tyr276) in the sample, as further described below in Analysis & Quantification.
  • An AQUA peptide having a sequence corresponding to the tyrosine 654 phosphorylation site in human Enolase alpha, DGTGHYLCNACGLySK (y* phosphotyrosine) (see Row 168 in Table 1 (SEQ ID NO: 168)) but incorporating 14 C/ 15 N-labeled leucine (indicated by bold L) is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer (see Merrifield, supra.) as further described below in Synthesis '& MS/MS Signature.
  • the GATA6 (tyr417) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated GATA6(tyr417)in the sample, as further described below in Analysis & Quantification.
  • An AQUA peptide having a sequence corresponding to the tyrosine 56 phosphorylation site in human USP11 , DLDFSEFVIQPQNESNPELy*K (y* phosphotyrosine) (see Row 214 in Table 1 (SEQ ID NO: 214)) but incorporating 14 C/ 15 N-labeled leucine (indicated by bold L) is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer (see Merrifield, supra.) as further described below in Synthesis & MS/MS Signature.
  • the USP11(tyr870) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated USP11(tyr870) 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(dimethyiamino) 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

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

Abstract

L'invention concerne presque 219 nouveaux sites de phosphorylation identifiés dans des protéines de transduction de signaux et des voies sous-jacentes à un lymphome anaplasique à grandes cellules impliquant la translocation/fusion NPM-ALK, ainsi que des anticorps spécifiques de sites de phosphorylation et des peptides étiquetés avec des isotopes lourds (peptides AQUA) dans la détection sélective et la quantification de ces sites/protéines phosphorylés. Ladite invention a aussi trait à des méthodes d'utilisation desdits réactifs à de telles fins.
EP06803284A 2005-09-09 2006-09-08 Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules Withdrawn EP1929296A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US71582305P 2005-09-09 2005-09-09
PCT/US2006/035203 WO2007030792A2 (fr) 2005-09-09 2006-09-08 Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules

Publications (1)

Publication Number Publication Date
EP1929296A2 true EP1929296A2 (fr) 2008-06-11

Family

ID=37836560

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06803284A Withdrawn EP1929296A2 (fr) 2005-09-09 2006-09-08 Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules

Country Status (2)

Country Link
EP (1) EP1929296A2 (fr)
WO (1) WO2007030792A2 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015114633A1 (fr) * 2014-01-30 2015-08-06 Yissum Research And Development Company Of The Hebrew University Of Jerusalem Ltd. Peptides se liant à l'actine et compositions les contenant pour inhiber l'angiogenèse et traiter des affections médicales associées à celle-ci

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007030792A3 *

Also Published As

Publication number Publication date
WO2007030792A3 (fr) 2009-04-30
WO2007030792A2 (fr) 2007-03-15

Similar Documents

Publication Publication Date Title
EP1718976B1 (fr) Phosphorylation proteique dans le lymphome cellulaire large anaplasique
US7888480B2 (en) Reagents for the detection of protein phosphorylation in leukemia signaling pathways
WO2007027957A9 (fr) Reactifs pour la detection de la phosphorylation des proteines dans des processus de signalisation de la leucemie
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
US20110130547A1 (en) Reagents For The Detection Of Protein Phosphorylation In EGFR Signaling Pathways
WO2006086111A2 (fr) Reactifs permettant de detecter une phosphorylation de proteines dans des voies de signalisation de leucemie
US20090298093A1 (en) Reagents for the Detection of Protein Phosphorylation in ATM & ATR Kinase Signaling Pathways
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
US7973134B2 (en) Reagents for the detection of protein phosphorylation in anaplastic large cell lymphoma signaling pathways
WO2007133689A2 (fr) Réactifs pour la détection des chemins de signalisation de l'acétylation des protéines
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
WO2007133688A2 (fr) Réactifs pour la détection de la phosphorylation throsiniques dans els chemins de signalisation de l'ischémie cervicale
US7935790B2 (en) Reagents for the detection of protein phosphorylation in T-cell receptor signaling pathways
WO2006113050A2 (fr) Reactifs de detection de phosphorylation de proteines dans la voie de signalisation de carcinome
WO2007030792A2 (fr) Reactifs pour la detection de la phosphorylation proteique dans des voies de signalisation de lymphome anaplasique a grandes cellules
US7939636B2 (en) Reagents for the detection of protein phosphorylation in c-Src signaling pathways
EP1934614A2 (fr) Réactifs de détection de la phosphorylation protéinique dans la voie de signalisation de carcinome
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

17P Request for examination filed

Effective date: 20080320

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 RS

RTI1 Title (correction)

Free format text: REAGENT FOR THE DETECTION OF PROTEIN PHOSPHORYLATION IN ANAPLASTIC LARGE CELL LYMPHOMA SIGNALING PATHWAYS

RIN1 Information on inventor provided before grant (corrected)

Inventor name: BOCCALATTE, FRANCESCO

Inventor name: SPEK, ERIC

Inventor name: MORITZ, ALBRECHT

Inventor name: GUO, AILAN

Inventor name: FARNSWORTH, CHARLES

Inventor name: LEE, KIMBERLY

Inventor name: POLAKIEWICZ, ROBERTO

R17D Deferred search report published (corrected)

Effective date: 20090430

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: 20090401