Reagents for the detection of protein phosphorylation in signaling pathways

Abstract
The invention discloses novel phosphorylation sites identified in signal transduction proteins and pathways, and provides phosphorylation-site specific antibodies and heavy-isotope labeled peptides (AQUA peptides) for the selective detection and quantification of these phosphorylated sites/proteins, as well as methods of using the reagents for such purpose. Among the phosphorylation sites identified are sites occurring in the following protein types: adaptor/scaffold proteins, adhesion/extracellular matrix protein, apoptosis proteins, calcium binding proteins, cell cycle regulation proteins, chaperone proteins, chromatin, DNA binding/repair/replication proteins, cytoskeletal proteins, endoplasmic reticulum or golgi proteins, enzyme proteins, G/regulator proteins, inhibitor proteins, motor/contractile proteins, phosphatase, protease, Ser/Thr protein kinases, Protein kinase (Tyr)s, receptor/channel/cell suface proteins, RNA binding proteins, transcriptional regulators, tumor suppressor proteins, ubiquitan conjugating system proteins and proteins of unknown function.
Description
TECHNICAL FIELD

The invention relates generally to a variety of moieties and tools for the detection of protein phosphorylation. Moreover, the invention relates to the use of the same for diagnostic and therapeutic purposes.


BACKGROUND

The activation of proteins by post-translational modification is an important cellular mechanism for regulating most aspects of biological organization and control, including growth, development, homeostasis, and cellular communication. Cellular signal transduction pathways involve protein kinases, protein phosphatases, and phosphoprotein-interacting domain (e.g., SH2, PTB, WW, FHA, 14-3-3) containing cellular proteins to provide multidimensional, dynamic and reversible regulation of many biological activities. See e.g., Sawyer et al., Med. Chem. 1(3): 293-319 (2005).


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. See Graves et al., Pharmacol. Ther. 82: 111-21 (1999).


Many of these phosphorylation sites regulate critical biological processes and may prove to be important for diagnostic or therapeutic modalities useful in the treatment and management of many pathological conditions and diseases, including inter alia cancer, developmental disorders, as as inflammatory, immune, metabolic and bone diseases.


For example, of the more than 100 dominant oncogenes identified to date, 46 are protein kinases. See Hunter, supra. Understanding which proteins are modified by these kinases will greatly expand our understanding of the molecular mechanisms underlying oncogenic transformation. Therefore, the identification of, and ability to detect, phosphorylation sites on a wide variety of cellular proteins is crucially important to understanding the key signaling proteins and pathways implicated in the progression of many disease states.


Understanding reversible protein phosphorylation and its role in the operation and interrelationship between cellular components and functions provides the opportunity to gain a finer appreciation of cellular regulation. In spite of the importance of protein modification, phosphorylation is not yet well understood due to the extraordinary complexity of signaling pathways, and the slow development of the technology necessary to unravel it.


In many instances, such knowledge is likely to provide valuable tools useful to evaluate, and possibly to manipulate target pathways, ultimately altering the functional status of a given cell for a variety of purposes.


The importance of protein kinase-regulated signal transduction pathways is underscored by a number of drugs designed to treat various cancer types by the inhibition of target protein kinases at the apex or intermediary levels of pathways implicated in cancer development. See Stern et al., Expert Opin. Ther. Targets 9(4):851-60 (2005).


Leukemia, a disease in which a number of underlying signal transduction events have been elucidated, has become a disease model for phosphoproteomic research and development efforts. As such, it represent a paradigm leading the way for many other programs seeking to address many classes of diseases (See, Harrison's Principles of Internal Medicine, McGraw-Hill, New York, N.Y.)


Depending on the cell type involved and the rate by which the disease progresses leukemia can be defined as acute or chronic myelogenous leukemia (AML or CML), or acute and chronic lymphocytic leukemia (ALL or CLL).


Most varieties of leukemia are generally characterized by genetic alterations e.g., chromosomal translocations, deletions or point mutations resulting in the constitutive activation of protein kinase genes, and their products, particularly tyrosine kinases. The most well known alteration is the oncogenic role of the chimeric BCR-Abl gene. See Nowell, Science 132: 1497 (1960)). The resulting BCR-Abl kinase protein is constitutively active and elicits characteristic signaling pathways that have been shown to drive the proliferation and survival of CML cells (see Daley, Science 247: 824-830 (1990); Raitano et al., Biochim. Biophys. Acta. December 9; 1333(3): F201-16 (1997)).


The recent success of Imanitib (also known as STI571 or Gleevec®), the first molecularly targeted compound designed to specifically inhibit the tyrosine kinase activity of BCR-Abl, provided critical confirmation of the central role of BCR-Abl signaling in the progression of CML (see Schindler et al., Science 289: 1938-1942 (2000); Nardi et al., Curr. Opin. Hematol. 11: 35-43 (2003)).


The success of Gleevec® now serves as a paradigm for the development of targeted drugs designed to block the activity of other tyrosine kinases known to be involved in many diseased including leukemias and other malignancies (see, e.g., Sawyers, Curr. Opin. Genet. Dev. February; 12(1): 111-5 (2002); Druker, Adv. Cancer Res. 91:1-30 (2004)). For example, recent studies have demonstrated that mutations in the FLT3 gene occur in one third of adult patients with AML. FLT3 (Fms-like tyrosine kinase 3) is a member of the class III receptor tyrosine kinase (RTK) family including FMS, platelet-derived growth factor receptor (PDGFR) and c-KIT (see Rosnet et al., Crit. Rev. Oncog. 4: 595-613 (1993). In 20-27% of patients with AML, an internal tandem duplication in the juxta-membrane region of FLT3 can be detected (see Yokota et al., Leukemia 11: 1605-1609 (1997)). Another 7% of patients have mutations within the active loop of the second kinase domain, predominantly substitutions of aspartate residue 835 (D835), while additional mutations have been described (see Yamamoto et al., Blood 97: 2434-2439 (2001); Abu-Duhier et al., Br. J. Haematol. 113: 983-988 (2001)). Expression of mutated FLT3 receptors results in constitutive tyrosine phosphorylation of FLT3, and subsequent phosphorylation and activation of downstream molecules such as STAT5, Akt and MAPK, resulting in factor-independent growth of hematopoietic cell lines.


Altogether, FLT3 is the single most common activated gene in AML known to date. This evidence has triggered an intensive search for FLT3 inhibitors for clinical use leading to at least four compounds in advanced stages of clinical development, including: PKC412 (by Novartis), CEP-701 (by Cephalon), MLN518 (by Millenium Pharmaceuticals), and SU5614 (by Sugen/Pfizer) (see Stone et al., Blood (in press)(2004); Smith et al., Blood 103: 3669-3676 (2004); Clark et al., Blood 104: 2867-2872 (2004); and Spiekerman et al., Blood 101: 1494-1504 (2003)).


There is also evidence indicating that kinases such as FLT3, c-KIT and Abl are implicated in some cases of ALL (see Cools et al., Cancer Res. 64: 6385-6389 (2004); Hu, Nat. Genet. 36: 453-461 (2004); and Graux et al., Nat. Genet. 36: 1084-1089 (2004)). In contrast, very little is know regarding any causative role of protein kinases in CLL, except for a high correlation between high expression of the tyrosine kinase ZAP70 and the more aggressive form of the disease (see Rassenti et al., N. Eng. J. Med. 351: 893-901 (2004)).


Despite the identification of a few key molecules involved in progression of leukemia, the vast majority of signaling protein changes underlying this disease remains unknown. There is, therefore, relatively scarce information about kinase-driven signaling pathways and phosphorylation sites relevant to the different types of leukemia. This has hampered a complete and accurate understanding of how protein activation within signaling pathways is driving these complex cancers. Accordingly, there is a continuing and pressing need to unravel the molecular mechanisms of kinase-driven oncogenesis in leukemia by identifying the downstream signaling proteins mediating cellular transformation in this disease. Identifying particular phosphorylation sites on such signaling proteins and providing new reagents, such as phospho-specific antibodies and AQUA peptides, to detect and quantify them remains particularly important to advancing our understanding of the biology of this disease.


Presently, diagnosis of leukemia is made by tissue biopsy and detection of different cell surface markers. However, misdiagnosis can occur since some leukemia cases can be negative for certain markers, and because these markers may not indicate which genes or protein kinases may be deregulated. Although the genetic translocations and/or mutations characteristic of a particular form of leukemia 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 leukemia 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.


SUMMARY OF THE INVENTION

Several novel protein phosphorylation sites have been identified in a variety of cell lines. Such novel phosphorylation sites (tyrosine), and their corresponding parent proteins are reported (see Table 1). The elucidation of these sites at long last provides the elements necessary to attain those much needed proteomics tools and modalities.


The invention discloses novel phosphorylation sites identified in signal transduction proteins and pathways underlying various disease states including for example human leukemias. The invention thus 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.





BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1—Is a diagram broadly depicting the immunoaffinity isolation and mass-spectrometric characterization methodology (IAP) employed to identify the novel phosphorylation sites disclosed herein.


FIG. 2—Is a table (corresponding to Table 1) enumerating the Leukemia signaling protein phosphorylation sites disclosed herein: Column A=the name of the parent protein; Column B=the SwissProt accession number for the protein (human sequence); Column C=the protein type/classification; Column D=the tyrosine residue (in the parent protein amino acid sequence) at which phosphorylation occurs within the phosphorylation site; Column E=the phosphorylation site sequence encompassing the phosphorylatable residue (residue at which phosphorylation occurs (and corresponding to the respective entry in Column D) appears in lowercase; Column F=the type of leukemia in which the phosphorylation site was discovered; and Column G=the cell type(s), tissue(s) and/or patient(s) in which the phosphorylation site was discovered.


FIG. 3—is an exemplary mass spectrograph depicting the detection of the tyrosine 786 phosphorylation site in TrkC (see Row 139 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated tyrosine (shown as lowercase “y” in FIG. 2).


FIG. 4—is an exemplary mass spectrograph depicting the detection of the tyrosine 192 phosphorylation site in HSP90B (see Row 30 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated tyrosine (shown as lowercase “y” in FIG. 2).


FIG. 5—is an exemplary mass spectrograph depicting the detection of the tyrosine 328 phosphorylation site in TOP2A (see Row 87 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated serine (shown as lowercase “y” in FIG. 2).


FIG. 6—is an exemplary mass spectrograph depicting the detection of the tyrosine 15 phosphorylation site in SNRPN (see Row 157 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated tyrosine (shown as lowercase “y” in FIG. 2)


FIG. 7—is an exemplary mass spectrograph depicting the detection of the tyrosine 507 phosphorylation site in VPS35 (see Row 383 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated tyrosine (shown as lowercase “y” in FIG. 2).


FIG. 8—is an exemplary mass spectrograph depicting the detection of the tyrosine 192 phosphorylation site in TAGLN3 (see Row 66 in FIG. 2/Table 1), as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* indicates the phosphorylated tyrosine (shown as lowercase “y” in FIG. 2).





DETAILED DESCRIPTION

Several novel protein phosphorylation sites have been identified in a variety of cell lines. Such novel phosphorylation sites (tyrosine), and their corresponding parent proteins are reported (see Table 1). The elucidation of these sites at long last provides the elements necessary to attain those much needed proteomics tools and modalities.


The disclosure of the phosphorylation sites provides the key to the production of new moieties, compositions and methods to specifically detect and/or to quantify these phosphorylated sites/proteins. Such moieties include for example reagents, such as phosphorylation site-specific antibodies and AQUA peptides (heavy-isotope labeled peptides). Such reagents are highly useful, inter alia, for studying signal transduction events underlying the progression of many diseases known or suspected to involve protein phosphorylation e.g., leukemia in a mammal. Accordingly, the invention provides novel reagents—phospho-specific antibodies and AQUA peptides—for the specific detection and/or quantification of a target signaling protein/polypeptide (e.g., a signaling protein/polypeptide implicated in leukemia) 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 target signaling protein/polypeptide using the phosphorylation-site specific antibodies and AQUA peptides of the invention.


These phosphorylation sites correspond to numerous different parent proteins (the full sequences (human) of which are all publicly available in SwissProt database and their Accession numbers listed in Column B of Table 1/FIG. 2), each of which are have been linked to specific functions in the literature and thus may be organized into discrete protein type groups, for example adaptor/scaffold proteins, cytoskeletal proteins, protein kinases, and DNA binding proteins, etc. (see Column C of Table 1), the phosphorylation of which is relevant to signal transduction activity (e.g, underlying AML, CML, CLL, and ALL), as disclosed herein.


In part, the invention provides an isolated phosphorylation site-specific antibody that specifically binds a given target signaling protein/polypeptide only when phosphorylated (or not phosphorylated, respectively) at a particular tyrosine enumerated in Column D of Table 1/FIG. 2 comprised within the phosphorylatable peptide site sequence enumerated in corresponding Column E. In further part, the invention provides a heavy-isotope labeled peptide (AQUA peptide) for the detection and quantification of a given target signaling protein/polypeptide, the labeled peptide comprising a particular phosphorylatable peptide site/sequence enumerated in Column E of Table 1/FIG. 2 herein. For example, among the reagents provided by the invention is an isolated phosphorylation site-specific antibody that specifically binds the VAV1 adaptor/scaffold protein only when phosphorylated (or only when not phosphorylated) at tyrosine 791 (see Row 15 (and Columns D and E) of Table 1/FIG. 2). By way of further example, among the group of reagents provided by the invention is an AQUA peptide for the quantification of phosphorylated SLY adaptor/scaffold protein, the AQUA peptide comprising the phosphorylatable peptide sequence listed in Column E, Row 2, of Table 1/FIG. 2 (which encompasses the phosphorylatable tyrosine at position 116).


In one embodiment, the invention provides an isolated phosphorylation site-specific antibody that specifically binds a target signaling protein/polypeptide selected from Column A of Table 1 (Rows 2-384) 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: 1-383), wherein said antibody does not bind said signaling protein when not phosphorylated at said tyrosine. In another embodiment, the invention provides an isolated phosphorylation site-specific antibody that specifically binds a target signaling protein/polypeptide 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: 1-383), 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. In one embodiment, the immortalized cell line is a rabbit or mouse hybridoma.


In another embodiment, the invention provides a heavy-isotope labeled peptide (AQUA peptide) for the quantification of a target signaling protein/polypeptide 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: 1-383), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D of Table 1. In certain embodiments, the phosphorylatable tyrosine within the labeled peptide is phosphorylated, while in other embodiments, the phosphorylatable residue within the labeled peptide is not phosphorylated.


Reagents (antibodies and AQUA peptides) provided by the invention may conveniently be grouped by the type of target signaling protein/polypeptide in which a given phosphorylation site (for which reagents are provided) occurs. The protein types for each respective protein (in which a phosphorylation site has been discovered) are provided in Column C of Table 1/FIG. 2, and include: adaptor/scaffold proteins, adhesion/extracellular matrix protein, apoptosis proteins, calcium binding proteins, cell cycle regulation proteins, chaperone proteins, chromatin, DNA binding/repair/replication proteins, cytoskeletal proteins, endoplasmic reticulum or golgi proteins, enzyme proteins, G/regulator proteins, inhibitor proteins, motor/contractile proteins, phosphatase, protease, Ser/Thr protein kinases, protein kinase (Tyr)s, receptor/channel/cell suface proteins, RNA binding proteins, transcriptional regulators, tumor suppressor proteins, ubiquitan conjugating system proteins and proteins of unknown function. Each of these distinct protein groups is a subset of target signaling protein/polypeptide phosphorylation sites disclosed herein, and reagents for their detection/quantification may be considered a subset of reagents provided by the invention.


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/FIG. 2 adaptor/scaffold proteins, calcium binding proteins, chromatin or DNA binding/repair/replication proteins, cytoskeletal proteins, enzyme proteins, protein kinases (Tyr), protein kinases (Ser/Thr), receptor/channel/transporter/cell suface proteins, transcriptional regulators and translational regulators. Accordingly, among subsets of reagents provided by the invention are isolated antibodies and AQUA peptides useful for the detection and/or quantification of the foregoing protein/phosphorylation site subsets.


The patents, published applications, and scientific literature referred to herein establish the knowledge of those with skill in the art and are hereby incorporated by reference in their entirety to the same extent as if each was specifically and individually indicated to be incorporated by reference. Any conflict between any reference cited herein and the specific teachings of this specification shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification shall be resolved in favor of the latter.


In one subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds an cell cycle regulation protein selected from Column A, Rows 23-29, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 23-29, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 23-29, of Table 1 (SEQ ID NOs: 22-28), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the cell cycle regulation protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a cell cycle regulation protein selected from Column A, Rows 23-29, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 23-29, of Table 1 (SEQ ID NOs: 22-28), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 23-29, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following cell cycle regulation protein phosphorylation sites are: TSG101 (Y32) and VCP (Y644) (see SEQ ID NOs: 25 and 27).


In a second subset of embodiments there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a chaperone protein selected from Column A, Rows 30-37, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 30-37, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 30-37, of Table 1 (SEQ ID NOs: 29-36), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the chaperone protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a target signaling protein/polypeptide that is a chaperone protein selected from Column A, Rows 30-37, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 30-37, of Table 1 (SEQ ID NOs: 29-36), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 30-37, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following chaperone protein phosphorylation sites are: HSP90B (Y 192), STI1 (Y269) and TPR2 (Y317) (see SEQ ID NOs: 29, 30 and 36).


In another subset of embodiments there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a chromatin or DNA binding/repair/replication protein selected from Column A, Rows 38-55, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 38-55, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 38-55, of Table 1 (SEQ ID NOs: 37-54), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the chromatin or DNA binding/repair/replication protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a target signaling protein/polypeptide that is a chromatin or DNA binding/repair/replication protein selected from Column A, Rows 38-55, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 38-55, of Table 1 (SEQ ID NOs: 37-54), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 38-55, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following chromatin or DNA binding/repair/replication protein phosphorylation sites are: TOP2B (Y230), TSN (Y210), TYMS (Y153) and WRN (Y849) (see SEQ ID NO's: 41, 43, 46 and 50).


In still another subset of embodiments there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a cytoskeletal protein selected from Column A, Rows 56-83, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 56-83, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 56-83, of Table 1 (SEQ ID NOs: 55-82), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the cytoskeletal protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that is a cytoskeletal protein selected from Column A, Rows 56-83, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 56-83, of Table 1 (SEQ ID NOs: 55-82), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 56-83, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following cytoskeletal protein phosphorylation sites are:


SPTA1 (Y1538), SFTBN1 (Y1667), TAGLN3 (Y192), tubulin, beta-2 (Y51), VASP (Y16) and VIM (Y291) (see SEQ ID NOs: 56, 60, 65, 74, 78, and 80).


In still another subset of embodiments there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds an enzyme protein selected from Column A, Rows 84-101, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 84-101, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 84-101, of Table 1 (SEQ ID NOs: 83-100), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the enzyme protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that is a enzyme protein selected from Column A, Rows 84-101, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 84-101, of Table 1 (SEQ ID NOs: 83-100), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 84-101, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following enzyme protein phosphorylation sites are: TOP2A (Y328), TPH1 (Y401), TPI1 (Y48) and UAP1 (Y125) (see SEQ ID NOs: 86, 87, 89 and 91).


In still another subset of embodiments there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a protein kinase (Ser/Thr) selected from Column A, Rows 123-131, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 123-131, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 123-131 of Table 1 (SEQ ID NOs: 122-130), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds protein kinase (Ser/Thr) when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that is a protein kinase (Ser/Thr) selected from Column A, Rows 123-131, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 123-131, of Table 1 (SEQ ID NOs: 122-130), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 123-131, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following protein kinase (Ser/Thr) phosphorylation sites are: PKCD (Y374) and TRRAP (Y3497) (see SEQ ID NO: 122 and 128).


In yet another subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a protein kinase (Tyr) selected from Column A, Rows 132-141, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 132-141, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 132-141, of Table 1 (SEQ ID NOs: 131-140), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the protein kinase (Tyr) when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that is a protein kinase (Tyr) selected from Column A, Rows 132-141, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 132-141, of Table 1 (SEQ ID NOs: 131-140), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 132-141, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following protein kinase (Tyr) phosphorylation sites are: Yes (Y146), TrkC (Y786) and Tyro3 (Y685) (see SEQ ID NOs: 131, 138 and 139).


In yet another subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds an RNA binding protein selected from Column A, Rows 156-175, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 156-175, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 156-175, of Table 1 (SEQ ID NOs: 155-174), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the RNA binding protein when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that is a RNA protein selected from Column A, Rows 156-175, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 156-175, of Table 1 (SEQ ID NOs: 155-174), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 156-175, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following RNA protein phosphorylation sites are: SNRPN (Y15), UPF2 (Y974) and UPF3B (Y160) (see SEQ ID NOs: 156, 169 and 170).


In yet another subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a transcriptional regulator selected from Column A, Rows 176-231, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 176-231, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 176-231, of Table 1 (SEQ ID NOs: 175-230), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the transcriptional regulator when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a target signaling protein/polypeptide that is a transcriptional regulator selected from Column A, Rows 176-231, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 176-231, of Table 1 (SEQ ID NOs: 175-230), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 176-231, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following transcriptional regulator phosphorylation sites are:


SPT5 (Y140), SSB (Y23), SSRP1 (Y452), STAT3 (Y674), STAT5B (Y171), TAF172 (Y415), TCF12 (Y82), TEL (Y401) and TFIIF (Y124) (see SEQ ID NO: 178, 185, 186, 190, 192, 194, 201, 211 and 213).

In still another subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds a translational regulator selected from Column A, Rows 234-249, of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, Rows 234-249, of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E, Rows 234-249, of Table 1 (SEQ ID NOs: 233-248), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds the translational regulator when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of a signaling protein that translational regulator selected from Column A, Rows 234-249, said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E, Rows 234-249, of Table 1 (SEQ ID NOs: 233-248), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Rows 234-249, of Table 1.


Among this subset of reagents, antibodies and AQUA peptides for the detection/quantification of the following a translational regulator phosphorylation sites are: USP14 (Y417) and USP20 (Y227) (see SEQ ID NO: 243 and 245).


In yet a further subset of embodiments, there is provided:


(i) An isolated phosphorylation site-specific antibody that specifically binds SPTAN1 (Y976), (Column A, Row 5, of Table 1) only when phosphorylated at the tyrosine listed in corresponding Column D of Table 1), said tyrosine comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NO: 4), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.


(ii) An equivalent antibody to (i) above that only binds SPTAN1 (Y976) (Column A, Row 5 of Table 1) when not phosphorylated at the disclosed site (and does not bind the protein when it is phosphorylated at the site).


(iii) A heavy-isotope labeled peptide (AQUA peptide) for the quantification of SPTAN1 (Y976) (Column A, Row 5 of Table 1), said labeled peptide comprising the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NO: 4), which sequence comprises the phosphorylatable tyrosine listed in corresponding Column D, Row 5 of Table 1.


The invention also provides an immortalized cell line producing an antibody of the invention, for example, a cell line producing an antibody within any of the foregoing subsets of antibodies. In an embodiment, the immortalized cell line is a rabbit hybridoma or a mouse hybridoma.


In other embodiments, a heavy-isotope labeled peptide (AQUA peptide) of the invention (for example, an AQUA peptide within any of the foregoing subsets of AQUA peptides) comprises a disclosed site sequence wherein the phosphorylatable tyrosine is phosphorylated. In yet other embodiments, a heavy-isotope labeled peptide of the invention comprises a disclosed site sequence wherein the phosphorylatable tyrosine is not phosphorylated.


The foregoing subsets of reagents of the invention should not be construed as limiting the scope of the invention, which, as noted above, includes reagents for the detection and/or quantification of disclosed phosphorylation sites on any of the other protein type/group subsets (each a subset) listed in Column C of Table 1/FIG. 2.


Also provided by the invention are methods for detecting or quantifying a target signaling protein/polypeptide that is tyrosine phosphorylated, said method comprising the step of utilizing one or more of the above-described reagents of the invention to detect or quantify one or more target Signaling Protein(s)/Polypeptide(s) selected from Column A of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D of Table 1. In certain embodiments of the methods of the invention, the reagents comprise a subset of reagents as described above. The antibodies according to the invention maybe used in standard (e.g., ELISA or conventional cytometric assays). The invention thus, provides compositions and methods for the detection and/or quantitation of a given target signaling protein or polypeptide in a sample, by contacting the sample and a control sample with one or more antibody of the invention under conditions favoring the binding and thus formation of the complex of the antibody with the protein or peptide. The formation of the complex is then detected according to methods well established and known in the art.


Also provided by the invention is a method for obtaining a phosphorylation profile of a certain protein type or group, for example adaptor/scaffold proteins or cell cycle regulation proteins (Rows 2-20 and Rows 23-29, respectively, of Table 1), that is phosphorylated in a disease signaling pathway, said method comprising the step of utilizing one or more isolated antibody that specifically binds the protein group selected from Column A of Table 1 only when phosphorylated at the tyrosine listed in corresponding Column D, of Table 1, comprised within the phosphorylation site sequence listed in corresponding Column E, to detect the phosphorylation of one or more of said protein group, thereby obtaining a phosphorylation profile for said protein group.


The invention further contemplates compositions, foremost pharmaceutical compositions, containing onr or a more antibody according to the invention formulated together with a pharmaceutically acceptable carrier. One of skill will appreciate that in certain instances the composition of the invention may further comprise other pharmaceutically active moieties. The compounds according to the invention are optionally formulated in a pharmaceutically acceptable vehicle with any of the well-known pharmaceutically acceptable carriers, including diluents and excipients (see Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, Mack Publishing Co., Easton, Pa. 1990 and Remington: The Science and Practice of Pharmacy, Lippincott, Williams & Wilkins, 1995). While the type of pharmaceutically acceptable carrier/vehicle employed in generating the compositions of the invention will vary depending upon the mode of administration of the composition to a mammal, generally pharmaceutically acceptable carriers are physiologically inert and non-toxic. Formulations of compositions according to the invention may contain more than one type of compound of the invention), as well any other pharmacologically active ingredient useful for the treatment of the symptom/condition being treated.


The invention also provides methods of treating a mammal comprising the step of administering such a mammal a therapeutically effective amount of a composition according to the invention.


As used herein, by “treating” is meant reducing, preventing, and/or reversing the symptoms in the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual not being treated according to the invention. A practitioner will appreciate that the compounds, compositions, and methods described herein are to be used in concomitance with continuous clinical evaluations by a skilled practitioner (physician or veterinarian) to determine subsequent therapy. Hence, following treatment the practitioners will evaluate any improvement in the treatment of the pulmonary inflammation according to standard methodologies. Such evaluation will aid and inform in evaluating whether to increase, reduce or continue a particular treatment dose, mode of administration, etc. The term “therapeutic composition” refers to any compounds administered to treat or prevent a disease. It will be understood that the subject to which a compound (e.g., an antibody) of the invention is administered need not suffer from a specific traumatic state. Indeed, the compounds (e.g., antibodies) of the invention may be administered prophylactically, prior to any development of symptoms. The term “therapeutic,” “therapeutically,” and permutations of these terms are used to encompass therapeutic, palliative as well as prophylactic uses. Hence, as used herein, by “treating or alleviating the symptoms” is meant reducing, preventing, and/or reversing the symptoms of the individual to which a compound of the invention has been administered, as compared to the symptoms of an individual receiving no such administration.


The term “therapeutically effective amount” is used to denote treatments at dosages effective to achieve the therapeutic result sought. Furthermore, one of skill will appreciate that the therapeutically effective amount of the compound of the invention may be lowered or increased by fine tuning and/or by administering more than one compound of the invention, or by administering a compound of the invention with another compound. See, for example, Meiner, C. L., “Clinical Trials: Design, Conduct, and Analysis,” Monographs in Epidemiology and Biostatistics, Vol. 8 Oxford University Press, USA (1986). The invention therefore provides a method to tailor the administration/treatment to the particular exigencies specific to a given mammal. As illustrated in the following examples, therapeutically effective amounts may be easily determined for example empirically by starting at relatively low amounts and by step-wise increments with concurrent evaluation of beneficial effect.









TABLE 1







Phosphorylation Sites















A


D






Protein
B
C
Phospho-
E
H


  1
Name
Accession No.
Protein Type
Residue
Phosphorylation Site Sequence
SEQ ID NO





  2
SLY
NP_061863.1
Adaptor/scaffold
Y116
ALSEEMADTLEEGSASPTSPDySLDSPGPEK
SEQ ID NO. 1






  3
SPTAN1
NP_003118.1
Adaptor/scaffold
Y601
TATDEAyKDPSNLQGK
SEQ ID NO. 2





  4
SPTAN1
NP_003118.1
Adaptor/scaffold
Y942
KHEALMSDLSAyGSSIQALR
SEQ ID NO. 3





  5
SPTAN1
NP_003118.1
Adaptor/scaffold
Y976
QQVAPTDDETGKELVLALyDYQEKSPR
SEQ ID NO. 4





  6
SPTAN1
NP_003118.1
Adaptor/scaffold
Y1020
QGFVPAAyVK
SEQ ID NO. 5





  7
SPTAN1
NP_003118.1
Adaptor/scaffold
Y1579
LQTASDESyKDPTNIQSK
SEQ ID NO. 6





  8
SYNE2
NP_055995.4
Adaptor/scaffold
Y3372
KMEEDIyTNLSK
SEQ ID NO. 7





  9
synergin,
NP_009178.3
Adaptor/scaffold
Y514
ALPSMDKyAVFK
SEQ ID NO. 8



gamma





 10
syntenin
NP_005616.2
Adaptor/scaffold
Y56
LYPELSQyMGLSLNEEEIR
SEQ ID NO. 9





 11
TRAF3IP3
NP_079504.1
Adaptor/scaffold
Y165
AEGPTIKNDASQQTNyGVAVLDKEIIQLSDYLK
SEQ ID NO. 10





 12
TRAF3IP3
NP_079504.1
Adaptor/scaffold
Y234
SLENQLyTCTQKYSPWGMK
SEQ ID NO. 11





 13
TRAF3IP3
NP_079504.1
Adaptor/scaffold
Y240
SLENQLYTCTQKySPWGMK
SEQ ID NO. 12





 14
TRAF3IP3
NP_079504.1
Adaptor/scaffold
Y302
SQyEALKEDWR
SEQ ID NO. 13





 15
VAV1
NP_005419.2
Adaptor/scaffold
Y791
ARyDFCAR
SEQ ID NO. 14





 16
VAV3
NP_006104.4
Adaptor/scaffold
Y508
WLEQFEMALSNIRPDyADSNFHDFK
SEQ ID NO. 15





 17
VAV3
NP_006104.4
Adaptor/scaffold
Y797
ARyDFCAR
SEQ ID NO. 16





 18
WAC
NP_057712.2
Adaptor/scaffold
Y25
RGDSQPyQALK
SEQ ID NO. 17





 19
ZO1
NP_003248.3
Adaptor/scaffold
Y830
ADGATSDDLDLHDDRLSYLSAPGSEySMYSTDSR
SEQ ID NO. 18





 20
ZO1
NP_003248.3
Adaptor/scaffold
Y1324
TLyRIPEPQKPQLKPPEDIVR
SEQ ID NO. 19





 21
URP2
NP_113659.3
Adhesion or
Y162
EKEPEEELyDLSKVVLAGGVAPALFR
SEQ ID NO. 20





extracellular





matrix protein





 22
zyxin
NP_003452.1
Adhesion or
Y20
PSPAISVSVSAPAFyAPQKK
SEQ ID NO. 21





extracellular





matrix protein





 23
SUGT1
NP_006695.1
Cell cycle
Y251
NLYPSSSPyTR
SEQ ID NO. 22





regulation





 24
TPX2
NP_036244.2
Cell cycle
Y81
ANLQQAIVTPLKPVDNTyYK
SEQ ID NO. 23





regulation





 25
TSG101
NP_006283.1
Cell cycle
Y15
yKYRDLTVRETVNVITLYK
SEQ ID NO. 24





regulation





 26
TSG101
NP_006283.1
Cell cycle
Y32
YKYRDLTVRETVNVITLyK
SEQ ID NO. 25





regulation





 27
VCP
NP_009057.1
Cell cycle
Y244
GILLyGPPGTGK
SEQ ID NO. 26





regulation





 28
VCP
NP_009057.1
Cell cycle
Y644
LDQLIyIPLPDEK
SEQ ID NO. 27





regulation





 29
ZW10
NP_004715.1
Cell cycle
Y349
NCLVySIPTNSSK
SEQ ID NO. 28





regulation





 30
HSP90B
NP_031381.2
Chaperone
Y192
VILHLKEDQTEyLEER
SEQ ID NO. 29





 31
STI1
NP_006810.1
Chaperone
Y269
ELDPTNMTYITNQAAVyFEKGDYNK
SEQ ID NO. 30





 32
STI1
NP_006810.1
Chaperone
Y275
ELDPTNMTYITNQAAVYFEKGDyNK
SEQ ID NO. 31





 33
STI1
NP_006810.1
Chaperone
Y444
AAALEAMKDyTK
SEQ ID NO. 32





 34
TBCA
NP_004598.1
Chaperone
Y48
MRAEDGENyDIKK
SEQ ID NO. 33





 35
TBCA
NP_004598.1
Chaperone
Y94
ILENEKDLEEAEEyKEAR
SEQ ID NO. 34





 36
TPR2
NP_003306.1
Chaperone
Y41
KDYNEAYNYyTK
SEQ ID NO. 35





 37
TPR2
NP_003306.1
Chaperone
Y317
KLDDAIEDCTNAVKLDDTyIK
SEQ ID NO. 36





 38
Smc5
NP_055925.1
Chromatin, DNA-
Y282
RPWVEYENVRQEyEEVK
SEQ ID NO. 37





binding, DNA





repair or DNA





replication





protein





 39
SON
NP_115571.1
Chromatin, DNA-
Y1037
SMMSAyER
SEQ ID NO. 38





binding, DNA





repair or DNA





replication





protein





 40
SON
NP_115571.1
Chromatin, DNA-
Y1096
SMMSSySAADR
SEQ ID NO. 39





binding, DNA





repair or DNA





replication





protein





 41
SON
NP_115571.1
Chromatin, DNA-
Y2192
EADSVyGEWVPVEK
SEQ ID NO. 40





binding, DNA





repair or DNA





replication





protein





 42
TOP2B
NP_001059.2
Chromatin, DNA-
Y230
IKHFDGEDyTCITFQPDLSK
SEQ ID NO. 41





binding, DNA





repair or DNA





replication





protein





 43
TOP2B
NP_001059.2
Chromatin, DNA-
Y344
HVDyVVDQVVGK
SEQ ID NO. 42





binding, DNA





repair or DNA





replication





protein





 44
TSN
NP_004613.1
Chromatin, DNA-
Y210
KVEEVVyDLSIR
SEQ ID NO. 43





binding, DNA





repair or DNA





replication





protein





 45
TSNAX
NP_005990.1
Chromatin, DNA-
Y237
QVYDGFSFIGNTGPyEVSKK
SEQ ID NO. 44





binding, DNA





repair or DNA





replication





protein





 46
TTF2
NP_003585.3
Chromatin, DNA-
Y285
GGPLNKEyTNWEAK
SEQ ID NO. 45





binding, DNA





repair or DNA





replication





protein





 47
TYMS
NP_001062.1
Chromatin, DNA-
Y153
DMESDySGQGVDQLQR
SEQ ID NO. 46





binding, DNA





repair or DNA





replication





protein





 48
UKp68
NP_079100.2
Chromatin, DNA-
Y321
KFNHDGEEEEEDDDyGSR
SEQ ID NO. 47





binding, DNA





repair or DNA





replication





protein





 49
WBP11
NP_057396.1
Chromatin, DNA-
Y236
RRDEDMLySPELAQR
SEQ ID NO. 48





binding, DNA





repair or DNA





replication





protein





 50
WBP11
NP_057396.1
Chromatin, DNA-
Y630
DDVyEAFMK
SEQ ID NO. 49





binding, DNA





repair or DNA





replication





protein





 51
WRN
NP_000544.2
Chromatin, DNA-
Y849
DMESYyQEIGR
SEQ ID NO. 50





binding, DNA





repair or DNA





replication





protein





 52
XPG
NP_000114.2
Chromatin, DNA-
Y289
RVVSEDTSHyILIK
SEQ ID NO. 51





binding, DNA





repair or DNA





replication





protein





 53
ZAP
NP_064504.2
Chromatin, DNA-
Y299
KFTyLGSQDR
SEQ ID NO. 52





binding, DNA





repair or DNA





replication





protein





 54
ZAP
NP_064504.2
Chromatin, DNA-
Y348
FLENGSQEDLLHGNPGSTyLASNSTSAPNWK
SEQ ID NO. 53





binding, DNA





repair or DNA





replication





protein





 55
ZAP
NP_064504.2
Chromatin, DNA-
Y826
DAIySHKNCPYDAK
SEQ ID NO. 54





binding, DNA





repair or DNA





replication





protein





 56
SPIRE1
NP_064533.2
Cytoskeletal
Y405
QVLVKAELEKyQQYK
SEQ ID NO. 55





protein





 57
SPTA1
NP_003117.2
Cytoskeletal
Y1538
DATNIQRKyLK
SEQ ID NO. 56





protein





 58
SPTA1
NP_003117.2
Cytoskeletal
Y2333
KGyVSLEDYTAFLIDKESENIK
SEQ ID NO. 57





protein





 59
SPTBN1
NP_842565.2
Cytoskeletal
Y66
ITDLyTDLR
SEQ ID NO. 58





protein





 60
SPTBN1
NP_842565.2
Cytoskeletal
Y1290
MLTAQDMSyDEAR
SEQ ID NO. 59





protein





 61
SPTBN1
NP_842565.2
Cytoskeletal
Y1667
VDKLyAGLKDLAEER
SEQ ID NO. 60





protein





 62
SPTBN1
NP_842565.2
Cytoskeletal
Y17
TSSISGPLSPAyTGQVPYNYNQLEGR
SEQ ID NO. 61



iso 2

protein





 63
SPTBN1
NP_8425652
Cytoskeletal
Y23
TSSISGPLSPAYTGQVPyNYNQLEGR
SEQ ID NO. 62



iso 2

protein





 64
SPTBN2
NP_008877.1
Cytoskeletal
Y1726
EVVAASHELGQDyEHVTMLR
SEQ ID NO. 63





protein





 65
SSFA2
NP_006742.2
Cytoskeletal
Y930
NSLQNLSQyPMMR
SEQ ID NO. 64





protein





 66
TAGLN3
NP_037391.2
Cytoskeletal
Y192
GASQAGMTGyGMPR
SEQ ID NO. 65





protein





 67
talin 1
NP_006280.2
Cytoskeletal
Y71
KGIWLEAGKALDYyMLR
SEQ ID NO. 66





protein





 68
tau
NP_005901.2
Cytoskeletal
Y197
SGySSPGSPGTPGSR
SEQ ID NO. 67





protein





 69
tau
NP_005901.2
Cytoskeletal
Y310
VQIVyKPVDLSK
SEQ ID NO. 68





protein





 70
tensin 2
NP_056134.2
Cytoskeletal
Y255
LGVIVSAyMHYSK
SEQ ID NO. 69





protein





 71
tensin 2
NP_056134.2
Cytoskeletal
Y258
LGVIVSAYMHySK
SEQ ID NO. 70





protein





 72
TES
NP_056456.1
Cytoskeletal
Y237
RTQYSCyCCK
SEQ ID NO. 71





protein





 73
TMOD3
NP_055362.1
Cytoskeletal
Y86
DREDyVPYTGEK
SEQ ID NO. 72





protein





 74
TPM4
NP_003281.1
Cytoskeletal
Y126
HIAEEADRKyEEVAR
SEQ ID NO. 73





protein





 75
tubulin,
NP_006079.1
Cytoskeletal
Y51
INVYyNEATGGK
SEQ ID NO. 74



beta-2

protein





 76
utrophin
NP_009055.2
Cytoskeletal
Y1757
KLLIAQEPLyQC
SEQ ID NO. 75





protein





 77
utrophin
NP_009055.2
Cytoskeletal
Y3111
LHyPMVEYCIPTTSGEDVRDFTK
SEQ ID NO. 76





protein





 78
utrophin
NP_009055.2
Cytoskeletal
Y3116
LHYPMVEyCIPTTSGEDVR
SEQ ID NO. 77





protein





 79
VASP
NP_003361.1
Cytoskeletal
Y16
ATVMLyDDGNKR
SEQ ID NO. 78





protein





 80
VASP
NP_003361.1
Cytoskeletal
Y341
SSSSVTTSETQPCTPSSSDySDLQR
SEQ ID NO. 79





protein





 81
VIM
NP_003371.2
Cytoskeletal
Y291
NLQEAEEWyK
SEQ ID NO. 80





protein





 82
WAVE1
NP_003922.1
Cytoskeletal
Y156
FYTNPSyFFDLWK
SEQ ID NO. 81





protein





 83
WDR1
NP_059830.1
Cytoskeletal
Y98
YEyQPFAGK
SEQ ID NO. 82





protein





 84
SLC27A2
NP_003636.1
Enzyme, misc.
Y602
MyVPMTEDIYNAISAK
SEQ ID NO. 83





 85
SLC27A2
NP_003636.1
Enzyme, misc.
Y610
MYVPMTEDIyNAISAK
SEQ ID NO. 84





 86
TKT
NP_001055.1
Enzyme, misc.
Y202
LGQSDPAPLQHQMDIyQK
SEQ ID NO. 85





 87
TOP2A
NP_001058.2
Enzyme, misc.
Y328
HVDyVADQIVTK
SEQ ID NO. 86





 88
TPH1
NP_004170.1
Enzyme, misc.
Y401
yNPYTRSIQILK
SEQ ID NO. 87





 89
TPH1
NP_004170.1
Enzyme, misc.
Y404
YNPyTRSIQILK
SEQ ID NO. 88





 90
TPI1
NP_000356.1
Enzyme, misc.
Y48
VPADTEVVCAPPTAyIDFAR
SEQ ID NO. 89





 91
TPI1
NP_000356.1
Enzyme, misc.
Y165
VVLAyEPVWAIGTGK
SEQ ID NO. 90





 92
UAP1
NP_003106.2
Enzyme, misc.
Y125
GMyDVGLPSR
SEQ ID NO. 91





 93
UAP1
NP_003106.2
Enzyme, misc.
Y299
TNPTEPVGVVCRVDGVyQVVEYSEISLATAQK
SEQ ID NO. 92





 94
UGP1
NP_006750.3
Enzyme, misc.
Y298
GGTLTQyEGKLR
SEQ ID NO. 93





 95
UMPS
NP_000364.1
Enzyme, misc.
Y16
AALGPLVTGLyDVQAFK
SEQ ID NO. 94





 96
UROD
NP_000365.3
Enzyme, misc.
Y342
YIANLGHGLyPDMDPEHVGAFVDAVHK
SEQ ID NO. 95





 97
UROD
NP_000365.3
Enzyme, misc.
Y30
AAWGEETDyTPVWCMR
SEQ ID NO. 96





 98
VARS2
NP_006286.1
Enzyme, misc.
Y878
SLGNVIDPLDVIyGISLQGLHNQLLNSNLDPSEV
SEQ ID NO. 97







EKAK





 99
WRNIP1
NP_064520.2
Enzyme, misc.
Y500
SHILYDRAGEEHyNCISALHK
SEQ ID NO. 98





100
ZDHHC3
NP_057682.1
Enzyme, misc.
Y127
EFIESLQLKPGQVVyK
SEQ ID NO. 99





101
ZDHHC3
NP_057682.1
Enzyme, misc.
Y323
AVFGHPFSLGWASPFATPDQGKADPyQYVV
SEQ ID NO. 100





102
SRGAP2
NP_056141.2
G protein or
Y161
VLNELySVMK
SEQ ID NO. 101





regulator





103
SWAP-70
NP_055870.2
G protein or
Y141
YPLIIVSEEIEyLLK
SEQ ID NO. 102





regulator





104
TD-60
NP_061185.1
G protein or
Y276
GNLySFGCPEYGQLGHNSDGK
SEQ ID NO. 103





regulator





105
TD-60
NP_061185.1
G protein or
Y359
VFSWGFGGyGR
SEQ ID NO. 104





regulator





106
VAV1
NP_005419.2
G protein or
Y841
VGWFPANYVEEDySEYC
SEQ ID NO. 105





regulator





107
SOCS7
NP_055413.1
Inhibitor protein
Y507
LyPVSRF
SEQ ID NO. 106





108
SPRED1
NP_689807.1
Inhibitor protein
Y82
MVVLECMLKKDLIyNK
SEQ ID NO. 107





109
THBS1
NP_003237.2
Inhibitor protein
Y817
DNCQYVyNVDQR
SEQ ID NO. 108





110
TIMM44
NP_006342.2
Mitochondrial
Y435
DQDELNPyAAWR
SEQ ID NO. 109





protein





111
TOP1MT
NP_443195.1
Mitochondrial
Y144
yFVDKAAARKVLSR
SEQ ID NO. 110





protein





112
TBP7
NP_006494.1
Protease
Y41
AQDEIPALSVSRPQTGLSFLGPEPEDLEDLySR
SEQ ID NO. 111





113
TBP7
NP_006494.1
Protease
Y205
GVLMyGPPGCGK
SEQ ID NO. 112





114
TBP7
NP_006494.1
Protease
Y417
KDEQEHEFyK
SEQ ID NO. 113





115
TPP2
NP_003282.1
Protease
Y645
VNESSHyDLAFTDVHFKPGQIR
SEQ ID NO. 114





116
TPP2
NP_003282.1
Protease
Y1052
LDSSCIYNELKETYPNyLPLYVAR
SEQ ID NO. 115





117
USP24
XP_371254.5
Protease
Y1697
GEVLEGSNAyYCEK
SEQ ID NO. 116





118
USP24
XP_371254.5
Protease
Y1858
VyDQTNPYTDVR
SEQ ID NO. 117





119
XPNPEP1
NP_065116.2
Protease
Y312
CCMPyTPICIAK
SEQ ID NO. 118





120
TIF1-
NP_005753.1
Protein kinase
Y517
VFPGSTTEDyNLIVIER
SEQ ID NO. 119



beta





121
TTK
NP_003309.2
Protein kinase,
Y833
TLyEHYSGGESHNSSSSK
SEQ ID NO. 120





dual-specificity





122
TTK
NP_003309.2
Protein kinase,
Y836
TLYEHySGGESHNSSSSK
SEQ ID NO. 121





dual-specificity





123
PKCD
NP_006245.2
Protein kinase,
Y374
GRGEyFAIK
SEQ ID NO. 122





Ser/Thr (non-





receptor)





124
TAO1
NP_065842.1
Protein kinase,
Y43
EIGHGSFGAVyFAR
SEQ ID NO. 123





Ser/Thr (non-





receptor)





125
TAO2
NP_004774.1
Protein kinase,
Y61
KMSySGKQSNEK
SEQ ID NO. 124





Ser/Thr (non-





receptor)





126
Titin
NP_003310.3
Protein kinase,
Y4912
PIEDVTIyEK
SEQ ID NO. 125





Ser/Thr (non-





receptor)





127
Titin
NP_003310.3
Protein kinase,
Y15976
ITGyIVEK
SEQ ID NO. 126





Ser/Thr (non-





receptor)





128
TRRAP
NP_003487.1
Protein kinase,
Y862
TLELCVDNLQPDFLyDHIQPVR
SEQ ID NO. 127





Ser/Thr (non-





receptor)





129
TRRAP
NP_003487.1
Protein kinase,
Y3497
GHNGKIyPYLVMNDACLTESR
SEQ ID NO. 128





Ser/Thr (non-





receptor)





130
WNK1
NP_061852.1
Protein kinase,
Y1215
DDyGFSGSQK
SEQ ID NO. 129





Ser/Thr(non-





receptor)





131
YSK1
NP_006365.2
Protein kinase,
Y35
IGKGSFGEVyK
SEQ ID NO. 130





Ser/Thr (non-





receptor)





132
Yes
NP_005424.1
Protein kinase,
Y146
NGYIPSNyVAPADSIQAEEWYFGK
SEQ ID NO. 131





Tyr (non-





receptor)





133
ZAP70
NP_001070.2
Protein kinase,
Y178
KLySGAQTDGK
SEQ ID NO. 132





Tyr (non-





receptor)





134
ZAP70
NP_001070.2
Protein kinase,
Y204
KEQGTYALSLIyGK
SEQ ID NO. 133





Tyr (non-





receptor)





135
ZAP70
NP_001070.2
Protein kinase,
Y597
MRACyYSLASK
SEQ ID NO. 134





Tyr (non-





receptor)





136
ZAP70
NP_001070.2
Protein kinase,
Y598
MRACYySLASK
SEQ ID NO. 135





Tyr (non-





receptor)





137
TIE1
NP_005415.1
Protein kinase,
Y1083
NCDDEVyELMR
SEQ ID NO. 136





Tyr (receptor)





138
TrkC
NP_002521.2
Protein kinase,
Y558
VFLAECyNLSPTKDK
SEQ ID NO. 137





Tyr (receptor)





139
TrkC
NP_002521.2
Protein kinase,
Y786
EVyDVMLGCWQR
SEQ ID NO. 138





Tyr (receptor)





140
Tyro3
NP_006284.2
Protein kinase,
Y685
KIYSGDyYRQGCASKLPVK
SEQ ID NO. 139





Tyr (receptor)





141
Tyro3
NP_006284.2
Protein kinase,
Y686
KIYSGDYyRQGCASKLPVK
SEQ ID NO. 140





Tyr (receptor)





142
SLC20A2
NP_006740.1
Receptor,
Y423
LVGDTVSySK
SEQ ID NO. 141





channel,





transporter or





cell surface





protein





143
SLC20A2
NP_006740.1
Receptor,
Y646
NIFVAWFVTVPVAGLFSAAVMALLMyGILPYV
SEQ ID NO. 142





channel,





transporter or





cell surface





protein





144
SLC20A2
NP_006740.1
Receptor,
Y651
NIFVAWFVTVPVAGLFSAAVMALLMYGILPyV
SEQ ID NO. 143





channel,





transporter or





cell surface





protein





145
SLC25A12
NP_003696.2
Receptor,
Y369
GSGSVVGELMyK
SEQ ID NO. 144





channel,





transporter or





cell surface





protein





146
SLC38A2
NP_061849.2
Receptor,
Y30
FSISPDEDSSSYSSNSDFNYSyPTK
SEQ ID NO. 145





channel,





transporter or





cell surface





protein





147
SLC4A7
NP_003606.3
Receptor,
Y44
AVyIGVHVPFSK
SEQ ID NO. 146





channel,





transporter or





cell surface





protein





148
SNX6
NP_067072.3
Receptor,
Y137
IGSSLyALGTQDSTDICK
SEQ ID NO. 147





channel,





transporter or





cell surface





protein





149
SORL1
NP_003096.1
Receptor,
Y136
SNVIVALARDSLALARPKSSDVyVSYDYGKSFK
SEQ ID NO. 148





channel,





transporter or





cell surface





protein





150
SORL1
NP_003096.1
Receptor,
Y139
SNVIVALARDSLALARPKSSDVYVSyDYGKSFK
SEQ ID NO. 149





channel,





transporter or





cell surface





protein





151
TNF-R1
NP_001056.1
Receptor,
Y318
REVAPPyQGADPILATALASDPIPNPLQK
SEQ ID NO. 150





channel,





transporter or





cell surface





protein





152
TNF-R1
NP_001056.1
Receptor,
Y360
WEDSAHKPQSLDTDDPATLyAVVENVPPLR
SEQ ID NO. 151





channel,





transporter or





cell surface





protein





153
TRPM3
NP_066003.2
Receptor,
Y250
INESLRDQLLVTIQKTFTy
SEQ ID NO. 152





channel,





transporter or





cell surface





protein





154
TSPAN15
NP_036471.1
Receptor,
Y97
ASLRDNLYLLQAFMyILGICL
SEQ ID NO. 153





channel,





transporter or





cell surface





protein





155
XG
NP_780778.1
Receptor,
Y52
PPyYPQPENPDSGGNIYPRPKPR
SEQ ID NO. 154





channel,





transporter or





cell surface





protein





156
snRNP116
NP_004238.2
RNA binding
Y167
KRYDQDLCyTDILFTEQER
SEQ ID NO. 155





protein





157
SNRPN
NP_003088.1
RNA binding
Y15
MLQHIDyR
SEQ ID NO. 156





protein





158
SRm300
NP_057417.3
RNA binding
Y145
AAFGISDSyVDGSSFDPQR
SEQ ID NO. 157





protein





159
SRm300
NP_057417.3
RNA binding
Y1145
FQSDSSSyPTVDSNSLLGQSR
SEQ ID NO. 158





protein





160
SRm300
NP_057417.3
RNA binding
Y2390
VPLSAyER
SEQ ID NO. 159





protein





161
SRp46
NP_115285.1
RNA binding
Y175
SPySRSRYRESRYGGSHYSSSGYSNSR
SEQ ID NO. 160





protein





162
SRp46
NP_115285.1
RNA binding
Y180
SPYSRSRyRESRYGGSHYSSSGYSNSR
SEQ ID NO. 161





protein





163
SRp46
NP_115285.1
RNA binding
Y190
SPYSRSRYRESRYGGSHySSSGYSNSR
SEQ ID NO. 162





protein





164
STAU2
NP_055208.1
RNA binding
Y388
GILHLSPDVyQEMEASR
SEQ ID NO. 163





protein





165
TAF15
NP_003478.1
RNA binding
Y67
SQSYGGyENQKQSSY
SEQ ID NO. 164





protein





166
TAF15
NP_003478.1
RNA binding
Y491
GGYGGDRGGGyGGDRGGYGGDRGGYGGDR
SEQ ID NO. 165





protein





167
TARDBP
NP_031401.1
RNA binding
Y155
FTEyETQVK
SEQ ID NO. 166





protein





168
U5-200kD
NP_054733.2
RNA binding
Y1682
IHAYVDYPIyDVLQMVGHANRPLQDDEGR
SEQ ID NO. 167





protein





169
U5-200kD
NP_054733.2
RNA binding
Y1770
RMTQNPNyYNLQGISHR
SEQ ID NO. 168





protein





170
UPF2
NP_056357.1
RNA binding
Y974
KSLEVWTKDHPFPIDIDyMISDTLELLR
SEQ ID NO. 169





protein





171
UPF3B
NP_075386.1
RNA binding
Y160
VGTIDDDPEyR
SEQ ID NO. 170





protein





172
UPF3B
NP_075386.1
RNA binding
Y429
NKDRPAMQLyQPGAR
SEQ ID NO. 171





protein





173
ZNF265
NP_005446.2
RNA binding
Y102
TGyGGGFNER
SEQ ID NO. 172





protein





174
ZNF265
NP_005446.2
RNA binding
Y114
ENVEyIEREESDGEYDEFGRK
SEQ ID NO. 173





protein





175
ZNF265
NP_005446.2
RNA binding
Y124
ENVEYIEREESDGEyDEFGRK
SEQ ID NO. 174





protein





176
SMARCE1
NP_003070.3
Transcriptional
Y142
AYHNSPAYLAyINAK
SEQ ID NO. 175





regulator





177
SMRT
NP_006303.3
Transcriptional
Y2249
SAVyPLLYR
SEQ ID NO. 176





regulator





178
SND1
NP_055205.2
Transcriptional
Y113
TPQGREYGMIyLGKDTNGENIAESLVAEGLATR
SEQ ID NO. 177





regulator





179
SPT5
NP_003160.2
Transcriptional
Y140
DQREEELGEyYMK
SEQ ID NO. 178





regulator





180
SPT5
NP_003160.2
Transcriptional
Y295
GIYKDDIAQVDyVEPSQNTISLK
SEQ ID NO. 179





regulator





181
SPT5
NP_003160.2
Transcriptional
Y778
TPMYGSQTPMyGSGSR
SEQ ID NO. 180





regulator





182
SPT5
NP_003160.2
Transcriptional
Y787
TPMyGSQTPLQDGSR
SEQ ID NO. 181





regulator





183
SS18L1
NP_945173.1
Transcriptional
Y272
yPDGHGDYAYQQSSYTEQSYDRSF
SEQ ID NO. 182





regulator





184
SS18L1
NP_945173.1
Transcriptional
Y281
YPDGHGDYAyQQSSYTEQSYDRSF
SEQ ID NO. 183





regulator





185
SS18L1
NP_945173.1
Transcriptional
Y286
YPDGHGDYAYQQSSyTEQSYDRSF
SEQ ID NO. 184





regulator





186
SSB
NP_003133.1
Transcriptional
Y23
ICHQIEyYFGDFNLPR
SEQ ID NO. 185





regulator





187
SSRP1
NP_003137.1
Transcriptional
Y452
EGMNPSYDEYADSDEDQHDAyLER
SEQ ID NO. 186





regulator





188
STAT1
NP_009330.1
Transcriptional
Y106
RNLQDNFQEDPIQMSMIIySCLKEER
SEQ ID NO. 187





regulator





189
STAT1
NP_009330.1
Transcriptional
Y668
YLyPNIDKDHAFGK
SEQ ID NO. 188





regulator





190
STAT3
NP_644805.1
Transcriptional
Y45
QFLAPWIESQDWAyAASK
SEQ ID NO. 189





regulator





191
STAT3
NP_644805.1
Transcriptional
Y674
IMDATNILVSPLVYLyPDIPKEEAFGK
SEQ ID NO. 190





regulator





192
STAT3
NP_644805.1
Transcriptional
Y686
yCRPESQEHPEADPGSMPYLK
SEQ ID NO. 191





regulator





193
STAT5B
NP_036580.2
Transcriptional
Y171
KLQQTQEyFIIQYQESLR
SEQ ID NO. 192





regulator





194
STAT5B
NP_036580.2
Transcriptional
Y683
YyTPVPCESATAK
SEQ ID NO. 193





regulator





195
TAF172
NP_003963.1
Transcriptional
Y415
yALAVRQDVINTLLPK
SEQ ID NO. 194





regulator





196
TAF6
NP_005632.1
Transcriptional
Y253
RAEALQSIATDPGLyQMLPR
SEQ ID NO. 195





regulator





197
TAT-SF1
NP_055315.2
Transcriptional
Y50
DGDTQTDAGGEPDSLGQQPTDTPyEWDLDKK
SEQ ID NO. 196





regulator





198
TAT-SF1
NP_055315.2
Transcriptional
Y634
EFDEDSDEKEEEEDTyEK
SEQ ID NO. 197





regulator





199
TAT-SF1
NP_055315.2
Transcriptional
Y650
VFDDESDEKEDEEyADEK
SEQ ID NO. 198





regulator





200
TCERG1
NP_006697.2
Transcriptional
Y149
TPDGKVYyYNARTR
SEQ ID NO. 199





regulator





201
TCERG1
NP_006697.2
Transcriptional
Y150
TPDGKVYYyNARTR
SEQ ID NO. 200





regulator





202
TCF12
NP_003196.1
Transcriptional
Y82
GFTDSPHySDHLNDSR
SEQ ID NO. 201





regulator





203
TCF12
NP_003196.1
Transcriptional
Y156
QDLGLGSPAQLSSSGKPGTAyYSFSATSSR
SEQ ID NO. 202





regulator





204
TCF12
NP_003196.1
Transcriptional
Y157
QDLGLGSPAQLSSSGKPGTAYySFSATSSR
SEQ ID NO. 203





regulator





205
TCF20
NP_005641.1
Transcriptional
Y839
QINLTDyPIPR
SEQ ID NO. 204





regulator





206
TEF-3
NP_003204.2
Transcriptional
Y349
QVVEKVETEyARYENGHYSYRIHR
SEQ ID NO. 205





regulator





207
TEF-3
NP_003204.2
Transcriptional
Y352
QVVEKVETEYARyENGHYSYRIHR
SEQ ID NO. 206





regulator





208
TEF-3
NP_003204.2
Transcriptional
Y357
QVVEKVETEYARYENGHySYRIHR
SEQ ID NO. 207





regulator





209
Tel
NP_001978.1
Transcriptional
Y60
LQPIyWSRDDVAQWLK
SEQ ID NO. 208





regulator





210
Tel
NP_001978.1
Transcriptional
Y114
YRSPHSGDVLyELLQHILK
SEQ ID NO. 209





regulator





211
Tel
NP_001978.1
Transcriptional
Y346
LLWDYVyQLLSDSR
SEQ ID NO. 210





regulator





212
Tel
NP_001978.1
Transcriptional
Y401
HyYKLNIIR
SEQ ID NO. 211





regulator





213
Tel
NP_001978.1
Transcriptional
Y402
HYyKLNIIR
SEQ ID NO. 212





regulator





214
TFIIF
NP_004119.1
Transcriptional
Y124
AECRPAASENyMR
SEQ ID NO. 213





regulator





215
TFII-I
NP_001509.2
Transcriptional
Y419
EDLQLDKPASGVKEEWyAR
SEQ ID NO. 214





regulator





216
TORC2
NP_859066.1
Transcriptional
Y488
LPPYPySSPSLVLPTQPHTPK
SEQ ID NO. 215





regulator





217
TRAP150
NP_005110.1
Transcriptional
Y228
ASESSKPWPDATyGTGSASR
SEQ ID NO. 216





regulator





218
Trap170
NP_004220.2
Transcriptional
Y941
DGAySLFDNSK
SEQ ID NO. 217





regulator





219
TRIM22
NP_006065.2
Transcriptional
Y394
SSGFAFDPSVNySK
SEQ ID NO. 218





regulator





220
TRIP11
NP_004230.1
Transcriptional
Y452
LNNEyEVIK
SEQ ID NO. 219





regulator





221
TRIP11
NP_004230.1
Transcriptional
Y1267
LQVDyTGLIQSYEQNETK
SEQ ID NO. 220





regulator





222
TSC22D2
NP_055594.1
Transcriptional
Y703
SHLMyAVR
SEQ ID NO. 221





regulator





223
UXT
NP_004173.1
Transcriptional
Y43
DKVyEQLAK
SEQ ID NO. 222





regulator





224
YY1
NP_003394.1
Transcriptional
Y251
DIDHETVVEEQIIGENSPPDySEYMTGK
SEQ ID NO. 223





regulator





225
YY1
NP_003394.1
Transcriptional
Y254
DIDHETVVEEQIIGENSPPDYSEyMTGK
SEQ ID NO. 224





regulator





226
ZBTB16
NP_005997.2
Transcriptional
Y334
HLGIySVLPNHK
SEQ ID NO. 225





regulator





227
ZNF141
NP_003432.1
Transcriptional
Y423
KIHSADKPyKCK
SEQ ID NO. 226





regulator





228
ZNF281
NP_036614.1
Transcriptional
Y353
CDTCQQyFSR
SEQ ID NO. 227





regulator





229
ZNF281
NP_036614.1
Transcriptional
Y479
KNTDKNyLNFVSPLPDIVGQK
SEQ ID NO. 228





regulator





230
ZNF289
NP_115765.2
Transcriptional
Y278
LAyQELQIDRK
SEQ ID NO. 229





regulator





231
ZNF33A
NP_008885.1
Transcriptional
Y45
DVMLENySNLVSVGYCVHK
SEQ ID NO. 230





regulator





232
TSFM
NP_005717.2
Translational
Y59
TGySFVNCK
SEQ ID NO. 231





regulator





233
TUFM
NP_003312.3
Translational
Y269
DLEKPFLLPVEAVySVPGR
SEQ ID NO. 232





regulator





234
UBC3B
NP_060281.2
Ubiquitin
Y190
VPTTLAEyCIK
SEQ ID NO. 233





conjugating





system





235
UBCE7IP3
NP_006453.1
Ubiquitin
Y220
QQQQEGNyLQHVQLDQR
SEQ ID NO. 234





conjugating





system





236
UBE1L
NP_003326.2
Ubiquitin
Y15
LLDEELySR
SEQ ID NO. 235





conjugating





system





237
ubiquilin2
NP_038472.2
Ubiquitin
Y265
ALSNLESIPGGyNALR
SEQ ID NO. 236





conjugating





system





238
ubiquitin
NP_002945.1
Ubiquitin
Y148
CCLTyCFNKPEDK
SEQ ID NO. 237





conjugating





system





239
UREB1
NP_113584.3
Ubiquitin
Y1658
YTVQFTTMVQVNEETGNR
SEQ ID NO. 238





conjugating





system





240
UREB1
NP_113584.3
Ubiquitin
Y3424
SVPVSAGGEGETSPySLEASPLGQLMNMLSHPVI
SEQ ID NO. 239





conjugating

RR





system





241
UREB1
NP_113584.3
Ubiquitin
Y4078
EMFNPMyALFR
SEQ ID NO. 240





conjugating





system





242
USP10
NP_005144.2
Ubiquitin
Y54
SSVELPPYSGTVLCGTQAVDKLPDGQEyQR
SEQ ID NO. 241





conjugating





system





243
USP11
NP_004642.2
Ubiquitin
Y396
SQFLGyQQHDSQE
SEQ ID NO. 242





conjugating





system





244
USP14
NP_005142.1
Ubiquitin
Y417
YEPFSFADDIGSNNCGyYDLQAVLTHQGR
SEQ ID NO. 243





conjugating





system





245
USP15
NP_006304.1
Ubiquitin
Y239
NSNYCLPSyTAYK
SEQ ID NO. 244





conjugating





system





246
USP20
NP_006667.2
Ubiquitin
Y227
GyAQQDTQEFLR
SEQ ID NO. 245





conjugating





system





247
USP4
NP_003354.2
Ubiquitin
Y192
YMSNTyEQLSK
SEQ ID NO. 246





conjugating





system





248
USP4
NP_003354.2
Ubiquitin
Y476
VSVTFDPFCyLTLPLPLKK
SEQ ID NO. 247





conjugating





system





249
WWP2
NP_008945.2
Ubiquitin
Y369
NyEQWQSQR
SEQ ID NO. 248





conjugating





system





250
SLFN5
XP_496206.1
Unknown function
Y115
AEVENKGySYK
SEQ ID NO. 249





251
SLITRK5
NP_056382.1
Unknown function
Y833
SPAySVSTIEPR
SEQ ID NO. 250





252
SMAP-5
NP_110426.4
Unknown function
Y26
SIDDQSQQSyDYGGSGGPY
SEQ ID NO. 251





253
SMAP-5
NP_110426.4
Unknown function
Y28
SIDDQSQQSYDyGGSGGPY
SEQ ID NO. 252





254
SMAP-5
NP_110426.4
Unknown function
Y67
TGQIyQPTQAY
SEQ ID NO. 253





255
SMBP
NP_064508.2
Unknown function
Y272
YSKEEEMDDMDRDLGDEyGWK
SEQ ID NO. 254





256
SMC6L1
NP_078900.1
Unknown function
Y780
SLKIEAENKyDAIK
SEQ ID NO. 255





257
SMEK2
NP_065196.1
Unknown function
Y171
EKLALALENEGyIK
SEQ ID NO. 256





258
SNF8
NP_009172.2
Unknown function
Y192
NGyVTVSEIK
SEQ ID NO. 257





259
SNX14
NP_065201.1
Unknown function
Y353
SLHEELQKIyKTY
SEQ ID NO. 258





260
SPATA2
NP_006029.1
Unknown function
Y257
SVDAyDSYWESR
SEQ ID NO. 259





261
SPATA2
NP_006029.1
Unknown function
Y260
SVDAYDSyWESR
SEQ ID NO. 260





262
SPATA5
NP_660208.1
Unknown function
Y180
IVLPGNFLyCTFYGRPYK
SEQ ID NO. 261





263
SPATA5
NP_660208.1
Unknown function
Y793
IIyVPLPDAATRR
SEQ ID NO. 262





254
SSX9
NP_777622.1
Unknown function
Y48
SSEKIIyVYMKR
SEQ ID NO. 263





265
SSX9
NP_777622.1
Unknown function
Y50
SSEKIIYVyMKR
SEQ ID NO. 264





266
STS-1
NP_116262.2
Unknown function
Y402
MDVVFGKyWLSQCFDAK
SEQ ID NO. 265





267
SYAP1
NP_116185.2
Unknown function
Y60
DFGNyLFNFASAATK
SEQ ID NO. 266





268
SYAP1
NP_116185.2
Unknown function
Y327
ELQQELQEyEVVTESEKR
SEQ ID NO. 267





269
SYNPO
NP_009217.3
Unknown function
Y222
MSGRAAATTPTKVySE
SEQ ID NO. 268





270
TBC1D12
XP_051081.4
Unknown function
Y800
LyEDILLQMDFIHIAQFLTK
SEQ ID NO. 269





271
TBC1D13
NP_060671.2
Unknown function
Y143
ATDyPCLLILDPQNEFETLR
SEQ ID NO. 270





272
TDRD3
NP_110421.1
Unknown function
Y481
HFNVNTDyQNPVR
SEQ ID NO. 271





273
TDRD3
NP_110421.1
Unknown function
Y644
STRPTQQFyQPPR
SEQ ID NO. 272





274
TIP20
NP_001002836.1
Unknown function
Y150
IHTGEKPyTCPDCGR
SEQ ID NO. 273





275
TIPRL
NP_690866.1
Unknown function
Y209
LYHEADKTyMLR
SEQ ID NO. 274





276
TLCD1
NP_612472.1
Unknown function
Y173
VNKyVNLVMYFLFR
SEQ ID NO. 275





277
TLCD1
NP_612472.1
Unknown function
Y179
VNKYVNLVMyFLFR
SEQ ID NO. 276





278
TNKS1BP1
NP_203754.2
Unknown function
Y855
DSQGTySSRDAELQDQEFGKR
SEQ ID NO. 277





279
TNKS1BP1
NP_203754.2
Unknown function
Y940
DVSLGTyGSR
SEQ ID NO. 278





280
TNKS1BP1
NP_203754.2
Unknown function
Y1122
SyQFGIIGNDR
SEQ ID NO. 279





281
TNRC6B
NP_055903.1
Unknown function
Y1081
DMGTTDSGPyFEKGGSHGLFGNSTAQSR
SEQ ID NO. 280





282
TNRC6B
NP_055903.1
Unknown function
Y1388
GIQNIDPESDPyVTPGSVLGGTATSPIVDTDHQL
SEQ ID NO. 281







LR





283
TOM1
NP_005479.1
Unknown function
Y386
yEAPQATDGLAGALDAR
SEQ ID NO. 282





284
TPD52
NP_005070.1
Unknown function
Y96
GWQDVTATSAyK
SEQ ID NO. 283





285
TPD52L2
NP_003279.2
Unknown function
Y106
SWHDVQVSSAyVK
SEQ ID NO. 284





286
TPR18
NP_660153.2
Unknown function
Y1023
EYEKAKKTyMQACK
SEQ ID NO. 285





287
TRIM16
NP_006461.3
Unknown function
Y371
EQFLQyAYDITFDPDTAHKYLRLQEENRK
SEQ ID NO. 286





288
TRIM34
NP_067629.2
Unknown function
Y391
yRPLFGYWVIGLQNKCK
SEQ ID NO. 287





289
TRIM34
NP_067629.2
Unknown function
Y397
YRPLFGyWVIGLQNKCK
SEQ ID NO. 288





290
TRIM9
NP_443210.1
Unknown function
Y524
TGVSPySKTLVLQTSEGK
SEQ ID NO. 289



iso5





291
TRS85
NP_055754.2
Unknown function
Y179
IQHNSDySYPK
SEQ ID NO. 290





292
TRS85
NP_055754.2
Unknown function
Y181
IQHNSDYSyPK
SEQ ID NO. 291





293
TSC22D2
NP_055594.1
Unknown function
Y57
ATDyGPEEVCER
SEQ ID NO. 292





294
TTC21B
NP_079029.3
Unknown function
Y160
GKEPyTKKALK
SEQ ID NO. 293





295
UBAP2
NP_060919.2
Unknown function
Y199
FSTQGMGTFNPADySDSTSTDVCGTK
SEQ ID NO. 294





296
UBAP2
NP_060919.2
Unknown function
Y1111
SQASKPAyGNSPYWTN
SEQ ID NO. 295





297
UBAP2
NP_060919.2
Unknown function
Y1116
SQASKPAYGNSPyWTN
SEQ ID NO. 296





298
USH3A
NP_443721.1
Unknown function
Y95
HLSEKIANyKEGTY
SEQ ID NO. 297





299
USP47
BAB55063.1
Unknown function
Y81
EGSVGSTSDyVSQSYSYSSILNK
SEQ ID NO. 298





300
USP47
BAB55063.1
Unknown function
Y86
EGSVGSTSDYVSQSySYSSILNK
SEQ ID NO. 299





301
UTX
NP_066963.1
Unknown function
Y928
PPSSPyPPLPKDKLNPPTPSIYLENK
SEQ ID NO. 300





302
VPS13B
NP_060360.3
Unknown function
Y242
LHFTyENLNSK
SEQ ID NO. 301





303
VPS13B
NP_060360.3
Unknown function
Y2100
KIKNAHSLAHSEETSAMSNTMVNKDDLPVSKyYR
SEQ ID NO. 302





304
VPS13B
NP_060360.3
Unknown function
Y2101
KIKNAHSLAHSEETSAMSNTMVNKDDLPVSKYyR
SEQ ID NO. 303





305
VPS13D
NP_056193.2
Unknown function
Y661
VHTSGFGyQSELELR
SEQ ID NO. 304





306
WAPL
NP_055860.1
Unknown function
Y633
REDKELyTVVQHVK
SEQ ID NO. 305





307
WBP2
NP_036610.2
Unknown function
Y231
AAEAAASAyYNPGNPHNVYMPTSQPPPPPYYPPE
SEQ ID NO. 306







DK





308
WBSCR20A
NP_060514.1
Unknown function
Y37
QLyALVCETQR
SEQ ID NO. 307





309
WDR18
NP_077005.2
Unknown function
Y61
NyISAWELQR
SEQ ID NO. 308





310
WDR3
NP_006775.1
Unknown function
Y456
TMTCEyALCSFFVPGDR
SEQ ID NO. 309





311
WDR43
BAA13441.1
Unknown function
Y196
MSLLLVyGSWFQPTIER
SEQ ID NO. 310





312
WDR70
NP_060504.1
Unknown function
Y618
AAEDSPyWVSPAYSK
SEQ ID NO. 311





313
WDR9
NP_061836.2
Unknown function
Y551
PyEKIPDQMFFHTDYR
SEQ ID NO. 312





314
WHSC1
NP_579877.1
Unknown function
Y747
KyPLTVFESRGFR
SEQ ID NO. 313





315
YEATS2
NP_060493.3
Unknown function
Y15
TIKETDPDyEDVSVALPNKR
SEQ ID NO. 314





316
YRDC
NP_078916.3
Unknown function
Y278
YGLLPSHASyL
SEQ ID NO. 315





317
Za2Od3
NP_061879.2
Unknown function
Y185
YSDVHNCSYNyK
SEQ ID NO. 316





318
ZBTB11
NP_055230.1
Unknown function
Y517
SVNEGAyIR
SEQ ID NO. 317





319
ZBTB5
NP_055687.1
Unknown function
Y641
VHTGEKPyACLK
SEQ ID NO. 318





320
ZC3H7A
NP_054872.2
Unknown function
Y681
RyWQNLEANVPGAQVLGNQIMPGFLNMKIK
SEQ ID NO. 319





321
ZDHHC11
NP_079062.1
Unknown function
Y106
LMKNySQPMPLFDRSK
SEQ ID NO. 320





322
ZDHHC13
NP_061901.2
Unknown function
Y59
ATQyGIFER
SEQ ID NO. 321





323
ZDHHC5
NP_056272.2
Unknown function
Y354
yRPGYSSSSTSAAMPHSSSAK
SEQ ID NO. 322





324
ZDHHC5
NP_056272.2
Unknown function
Y358
YRPGySSSSTSAAMPHSSSAK
SEQ ID NO. 323





325
ZDHHC5
NP_056272.2
Unknown function
Y630
GVGSPEPGPTAPyLGR
SEQ ID NO. 324





326
ZDHHC8
NP_037505.1
Unknown function
Y579
SQADSLFGDSGVyDAPSSYSLQQASVLSEGPRGP
SEQ ID NO. 325







ALR





327
ZFP 598
NP_835461.1
Unknown function
Y108
KYDIYFADGKVyALYR
SEQ ID NO. 326





328
ZFP 598
NP_835461.1
Unknown function
Y289
HIDLQFSyAPR
SEQ ID NO. 327





329
ZFP 598
NP_835461.1
Unknown function
Y312
RNEGVVGGEDYEEVDRySR
SEQ ID NO. 328





330
ZFP 598
NP_835461.1
Unknown function
Y471
LKDEDFPSLSASTSSSCSTAATPGPVGLALPyAI
SEQ ID NO. 329







PAR





331
ZFPL1
NP_006773.2
Unknown function
Y235
DDDRTPGLHGDCDDDKyR
SEQ ID NO. 330





332
ZFR
NP_057191.2
Unknown function
Y610
RHRLQyKKKVNPDLQVEVK
SEQ ID NO. 331





333
ZFR
NP_057191.2
Unknown function
Y667
RYEEDMyWR
SEQ ID NO. 332





334
ZNF12
NP_008887.2
Unknown function
Y315
THSGEKPyECSYCGK
SEQ ID NO. 333





335
ZNF183
NP_008909.1
Unknown function
Y120
SAKPVGPEDMGATAVyELDTEKER
SEQ ID NO. 334





336
ZNF183
NP_008909.1
Unknown function
Y153
GKEDDKIyR
SEQ ID NO. 335





337
ZNF198
NP_003444.1
Unknown function
Y502
FCCQSCVSEyK
SEQ ID NO. 336





338
ZNF198
NP_003444.1
Unknown function
Y557
FFDMTQCIGPNGyMEPYCSTACMNSHK
SEQ ID NO. 337





339
ZNF198
NP_003444.1
Unknown function
Y595
NSLPQyQATMPDGKLYNFCNSSCVAK
SEQ ID NO. 338





340
ZNF198
NP_003444.1
Unknown function
Y605
NSLPQYQATMPDGKLyNFCNSSCVAK
SEQ ID NO. 339





341
ZNF198
NP_003444.1
Unknown function
Y671
TCSDDyKKLHCIVTYCEYCQEEK
SEQ ID NO. 340





342
ZNF198
NP_003444.1
Unknown function
Y680
TCSDDYKKLHCIVTyCEYCQEEK
SEQ ID NO. 341





343
ZNF198
NP_003444.1
Unknown function
Y683
LHCIVTYCEyCQEEK
SEQ ID NO. 342





344
ZNF198
NP_003444.1
Unknown function
Y729
CVTCNyCSQLCK
SEQ ID NO. 343





345
ZNF198
NP_003444.1
Unknown function
Y762
KFQDWyYK
SEQ ID NO. 344





346
ZNF198
NP_003444.1
Unknown function
Y819
GPENLHyDQGCQTSR
SEQ ID NO. 345





347
ZNF198
NP_003444.1
Unknown function
Y862
ATyCKPHMQTK
SEQ ID NO. 346





348
ZNF272
NP_006626.3
Unknown function
Y420
HFNIHTGEKPyECLQCGK
SEQ ID NO. 347





349
ZNF291
NP_065894.1
Unknown function
Y1207
ENyTQNTIQVAIQSLR
SEQ ID NO. 348





350
ZNF313
NP_061153.1
Unknown function
Y116
YQNyIMEGVK
SEQ ID NO. 349





351
ZNF313
NP_061153.1
Unknown function
Y186
SVVCPICASMPWGDPNyR
SEQ ID NO. 350





352
ZNF326
NP_892021.1
Unknown function
Y93
SGyGFNEPEQSR
SEQ ID NO. 351





353
ZNF330
NP_055302.1
Unknown function
Y262
NLSSDKyGDTSYHDEEEDEYEAEDDEEEEDEGR
SEQ ID NO. 352





354
ZNF330
NP_055302.1
Unknown function
Y267
YGDTSyHDEEEDEYEAEDDEEEEDEGR
SEQ ID NO. 353





355
ZNF330
NP_055302.1
Unknown function
Y275
YGDTSYHDEEEDEyEAEDDEEEEDEGR
SEQ ID NO. 354





356
ZNF330
NP_055302.1
Unknown function
Y312
TYASGyAHYEEQEN
SEQ ID NO. 355





357
ZNF341
NP_116208.3
Unknown function
Y174
GPPPVQSSLNMHSVPSyLT
SEQ ID NO. 356





358
ZNF425
NP_001001661.1
Unknown function
Y675
PFQCPECDKSyCIR
SEQ ID NO. 357





359
ZNF433
NP_001073880.1
Unknown function
Y132
AyEYQEYGQKPYK
SEQ ID NO. 358





360
ZNF433
NP_001073880.1
Unknown function
Y134
AYEyQEYGQKPYK
SEQ ID NO. 359





361
ZNF433
NP_001073880.1
Unknown function
Y137
AYEYQEyGQKPYK
SEQ ID NO. 360





362
ZNF433
NP_001073880.1
Unknown function
Y142
AYEYQEYGQKPyK
SEQ ID NO. 361





363
ZNF512
NP_115810.2
Unknown function
Y417
IQCPNQGCEAVySSVSGLK
SEQ ID NO. 362





364
ZNF532
NP_060651.2
Unknown function
Y811
SPyTCPECGAICR
SEQ ID NO. 363





365
ZNF563
NP_660319.1
Unknown function
Y237
QCSKAFPFySSYR
SEQ ID NO. 364





366
ZNF563
NP_660319.1
Unknown function
Y240
QCSKAFPFYSSyR
SEQ ID NO. 365





367
ZNF571
NP_057620.3
Unknown function
Y335
VHTGEKPyICKECGK
SEQ ID NO. 366





368
ZNF574
NP_073589.3
Unknown function
Y605
YHHTGEyPYK
SEQ ID NO. 367





369
ZNF577
NP_116068.1
Unknown function
Y41
VLyKEVMLENYINLVSIGYRGTKPDSLFK
SEQ ID NO. 368





370
ZNF592
NP_055445.2
Unknown function
Y768
SPyCCPECGVLCR
SEQ ID NO. 369





371
ZNF609
NP_055857.1
Unknown function
Y796
AEADKIySFTDNAPSPSIGGSSR
SEQ ID NO. 370





372
ZNF622
NP_219482.1
Unknown function
Y284
DHSFFIPDIEyLSDIK
SEQ ID NO. 371





373
ZSWIM4
NP_075560.1
Unknown function
Y548
yLFTALLPHDPDLAYRLALR
SEQ ID NO. 372





374
SNAP29
NP_004773.1
Vesicle protein
Y160
yQASHPNLR
SEQ ID NO. 373





375
SNX1
NP_003090.2
Vesicle protein
Y162
IGDGMNAyVAYK
SEQ ID NO. 374





376
SNX2
NP_003091.2
Vesicle protein
Y203
YLHVGyIVPPAPEK
SEQ ID NO. 375





377
SNX27
NP_112180.4
Vesicle protein
Y186
FVVYNVyMAGR
SEQ ID NO. 376





378
STX16
NP_003754.2
Vesicle protein
Y66
WVDGVDEIQyDVGR
SEQ ID NO. 377





379
STXBP5
NP_640337.2
Vesicle protein
Y502
NKDDRPNTDIVDEDPyAIQIISWCPESR
SEQ ID NO. 378





380
SV2A
NP_055664.2
Vesicle protein
Y65
FEEEDDDDDFPAPSDGyYR
SEQ ID NO. 379





381
SV2A
NP_055664.2
Vesicle protein
Y66
FEEEDDDDDFPAPSDGYyR
SEQ ID NO. 380





382
VPS29
NP_057310.1
Vesicle protein
Y69
GDFDENLNyPEQK
SEQ ID NO. 381





383
VPS35
NP_060676.2
Vesicle protein
Y507
SEDPDQQyLILNTAR
SEQ ID NO. 382





384
VPS4B
NP_004860.2
Vesicle protein
Y40
AGNYEEALQLYQHAVQyFLHVVK
SEQ ID NO. 383









The short name for each protein in which a phosphorylation site has presently been identified is provided in Column A, and its SwissProt accession number (human) is provided Column B. The protein type/group into which each protein falls is provided in Column C. The identified tyrosine residue at which phosphorylation occurs in a given protein is identified in Column D, and the amino acid sequence of the phosphorylation site encompassing the tyrosine residue is provided in Column E (lower case y=the tyrosine (identified in Column D)) at which phosphorylation occurs. Table 1 above is identical to FIG. 2, except that the latter includes the disease and cell type(s) in which the particular phosphorylation site was identified (Columns F and G).


“Antibody” or “antibodies” refers to all classes of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including whole antibodies and any antigen biding fragment thereof (e.g., Fab) or single chains thereof, including chimeric, polyclonal, and monoclonal antibodies. Antibodies are antigen-specific protein molecules produced by lymphocytes of the B cell lineage. Following antigenic stimulation, B cells that have surface immunoglobulin receptors that bind the antigen clonally expand, and the binding affinity for the antigen increases through a process called affinity maturation. The B cells further differentiate into plasma cells, which secrete large quantities of antibodies in to the serum. While the physiological role of antibodies is to protect the host animal by specifically binding and eliminating microbes and microbial pathogens from the body, large amounts of antibodies are also induced by intentional immunization to produce specific antibodies that are used extensively in many biomedical and therapeutic applications.


Antibody molecules are shaped somewhat like the letter “Y”, and consist of 4 protein chains, two heavy (H) and two light (L) chains. Antibodies possess two distinct and spatially separate functional features. The ends of each of the two arms of the “Y” contain the variable regions (variable heavy (V(H)) and variable light (V(L)) regions), which form two identical antigen-binding sites. The variable regions undergo a process of “affinity maturation” during the immune response, leading to a rapid divergence of amino acids within these variable regions. The other end of the antibody molecule, the stem of the “Y”, contains only the two heavy constant (CH) regions, interacts with effector cells to determine the effector functions of the antibody. There are five different CH region genes that encode the five different classes of immunoglobulins: IgM, IgD, IgG, IgA and IgE. These constant regions, by interacting with different effector cells and molecules, determine the immunoglobulin molecule's biological function and biological response.


Each V(H) and V(L) region contains three subregions called complementarity determining regions. These include CDR1-3 of the V(H) domain and CDR1-3 of the V(L) domain. These six CDRs generally form the antigen binding surface, and include those residues that hypermutate during the affinity maturation phase of the immune response. The CDR3 of the V(H) domain seems to play a dominant role in generating diversity oof both the B cell antigen receptor (BCR) and the T cell antigen receptor systems (Xu et al., Immunity 13:37-45 (2000)).


The term “antibody” or “antibodies” refers to all classes of polyclonal or monoclonal immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including whole antibodies and any antigen binding fragment thereof. This includes any combination of immunoglobulin domains or chains that contains a variable region (V(H) or V(L)) that retains the ability to bind the immunogen. Such fragments include F(ab)2 fragments (V(H)-C(H1), V(L)-C(L))2; monovalent Fab fragments (V(H)-C(H1), V(L)-C(L)); Fv fragment (V(H)-V(L); single-chain Fv fragments (Kobayashi et al., Steroids July; 67(8):733-42 (2002).


Monoclonal antibodies refer to clonal antibodies produced from fusions between immunized murine, rabbit, human, or other vertebrate species, and produced by classical fusion technology Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975 Aug. 7; 256(5517):495-7 or by alternative methods which may isolate clones of immunoglobulin secreting cells from transformed plasma cells.


When used with respect to an antibody's binding to one phospho-form of a sequence, the expression “does not bind” means that a phospho-specific antibody either does not apparently bind to the non-phospho form of the antigen as ascertained in commonly used experimental detection systems (Western blotting, IHC, Immunofluorescence, etc.). One of skill in the art will appreciate that the expression may be applicable in those instances when (1) a phospho-specific antibody either does not apparently bind to the non-phospho form of the antigen as ascertained in commonly used experimental detection systems (Western blotting, IHC, Immunofluorescence, etc.); (2) where there is some reactivity with the surrounding amino acid sequence, but that the phosphorylated residue is an immunodominant feature of the reaction. In cases such as these, there is an apparent difference in affinities for the two sequences. Dilutional analyses of such antibodies indicates that the antibodies apparent affinity for the phosphorylated form is at least 10-100 fold higher than for the non-phosphorylated form; or where (3) the phospho-specific antibody reacts no more than an appropriate control antibody would react under identical experimental conditions. A control antibody preparation might be, for instance, purified immunoglobulin from a pre-immune animal of the same species, an isotype- and species-matched monoclonal antibody. Tests using control antibodies to demonstrate specificity are recognized by one of skill in the art as appropriate and definitive.


In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. As used in this specification, the singular forms “a,” “an” and “the” specifically also encompass the plural forms of the terms to which they refer, unless the content clearly dictates otherwise. As used herein, unless specifically indicated otherwise, the word “or” is used in the “inclusive” sense of “and/or” and not the “exclusive” sense of “either/or.”


The term “about” is used herein to mean approximately, in the region of, roughly, or around. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20%.


As used herein, the recitation of a numerical range for a variable is intended to convey that the invention may be practiced with the variable equal to any of the values within that range. Thus, for a variable that is inherently discrete, the variable can be equal to any integer value of the numerical range, including the end-points of the range. Similarly, for a variable that is inherently continuous, the variable can be equal to any real value of the numerical range, including the end-points of the range. As an example, a variable that is described as having values between 0 and 2, can be 0, 1 or 2 for variables which are inherently discrete, and can be 0.0, 0.1, 0.01, 0.001, or any other real value for variables which are inherently continuous.


As used in this specification, whether in a transitional phrase or in the body of the claim, the terms “comprise(s)” and “comprising” are to be interpreted as having an open-ended meaning. That is, the terms are to be interpreted synonymously with the phrases “having at least” or “including at least”. When used in the context of a process, the term “comprising” means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound or composition, the term “comprising” means that the compound or composition includes at least the recited features or components, but may also include additional features or components.


“Antibody” or “antibodies” refers to all classes of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including whole antibodies and any antigen biding fragment thereof (e.g., Fab) or single chains thereof, including chimeric, polyclonal, and monoclonal antibodies. Antibodies are antigen-specific protein molecules produced by lymphocytes of the B cell lineage. Following antigenic stimulation, B cells that have surface immunoglobulin receptors that bind the antigen clonally expand, and the binding affinity for the antigen increases through a process called affinity maturation. The B cells further differentiate into plasma cells, which secrete large quantities of antibodies in to the serum. While the physiological role of antibodies is to protect the host animal by specifically binding and eliminating microbes and microbial pathogens from the body, large amounts of antibodies are also induced by intentional immunization to produce specific antibodies that are used extensively in many biomedical and therapeutic applications.


Antibody molecules are shaped somewhat like the letter “Y”, and consist of 4 protein chains, two heavy (H) and two light (L) chains. Antibodies possess two distinct and spatially separate functional features. The ends of each of the two arms of the “Y” contain the variable regions (variable heavy (V(H)) and variable light (V(L)) regions), which form two identical antigen-binding sites. The variable regions undergo a process of “affinity maturation” during the immune response, leading to a rapid divergence of amino acids within these variable regions. The other end of the antibody molecule, the stem of the “Y”, contains only the two heavy constant (CH) regions, interacts with effector cells to determine the effector functions of the antibody. There are five different CH region genes that encode the five different classes of immunoglobulins: IgM, IgD, IgG, IgA and IgE. These constant regions, by interacting with different effector cells and molecules, determine the immunoglobulin molecule's biological function and biological response.


Each V(H) and V(L) region contains three subregions called complementarity determining regions. These include CDR1-3 of the V(H) domain and CDR1-3 of the V(L) domain. These six CDRs generally form the antigen binding surface, and include those residues that hypermutate during the affinity maturation phase of the immune response. The CDR3 of the V(H) domain seems to play a dominant role in generating diversity oof both the B cell antigen receptor (BCR) and the T cell antigen receptor systems (Xu et al., Immunity 13:37-45 (2000)).


The term “antibody” or “antibodies” refers to all classes of polyclonal or monoclonal immunoglobulins, including IgG, IgM, IgA, IgD, and IgE, including whole antibodies and any antigen binding fragment thereof. This includes any combination of immunoglobulin domains or chains that contains a variable region (V(H) or V(L)) that retains the ability to bind the immunogen. Such fragments include F(ab)2 fragments (V(H)-C(H1), V(L)-C(L))2; monovalent Fab fragments (V(H)-C(H1), V(L)-C(L)); Fv fragment (V(H)-V(L); single-chain Fv fragments (Kobayashi et al., Steroids July; 67(8):733-42 (2002).


Monoclonal antibodies refer to clonal antibodies produced from fusions between immunized murine, rabbit, human, or other vertebrate species, and produced by classical fusion technology Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975 Aug. 7; 256(5517):495-7 or by alternative methods which may isolate clones of immunoglobulin secreting cells from transformed plasma cells.


When used with respect to an antibody's binding to one phospho-form of a sequence, the expression “does not bind” means that a phospho-specific antibody either does not apparently bind to the non-phospho form of the antigen as ascertained in commonly used experimental detection systems (Western blotting, IHC, Immunofluorescence, etc.). One of skill in the art will appreciate that the expression may be applicable in those instances when (1) a phospho-specific antibody either does not apparently bind to the non-phospho form of the antigen as ascertained in commonly used experimental detection systems (Western blotting, IHC, Immunofluorescence, etc.); (2) where there is some reactivity with the surrounding amino acid sequence, but that the phosphorylated residue is an immunodominant feature of the reaction. In cases such as these, there is an apparent difference in affinities for the two sequences. Dilutional analyses of such antibodies indicates that the antibodies apparent affinity for the phosphorylated form is at least 10-100 fold higher than for the non-phosphorylated form; or where (3) the phospho-specific antibody reacts no more than an appropriate control antibody would react under identical experimental conditions. A control antibody preparation might be, for instance, purified immunoglobulin from a pre-immune animal of the same species, an isotype- and species-matched monoclonal antibody. Tests using control antibodies to demonstrate specificity are recognized by one of skill in the art as appropriate and definitive.


“Target signaling protein/polypeptide” means any protein (or polypeptide derived therefrom) enumerated in Column A of Table 1/FIG. 2, which is disclosed herein as being phosphorylated in one or more cell line(s). Target signaling protein(s)/polypeptide(s) may be tyrosine kinases, such as TTN or BCR, or serine/threonine kinases, or direct substrates of such kinases, or may be indirect substrates downstream of such kinases in signaling pathways. Target signaling protein/polypeptide where elucidated in leukemia cell lines, however one of skill in the art will appreciate that a target signaling protein/polypeptide may also be phosphorylated in other cell lines (non-leukemic) 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.


Technical and scientific terms used herein have the meaning commonly understood by one of skill in the art to which the present invention pertains, unless otherwise defined. Reference is made herein to various methodologies and materials known to those of skill in the art. Standard reference works setting forth the general principles of recombinant DNA technology include Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, New York (1989); Kaufman et al., Eds., Handbook of Molecular and Cellular Methods in Biology in Medicine, CRC Press, Boca Raton (1995); McPherson, Ed., Directed Mutagenesis: A Practical Approach, IRL Press, Oxford (1991). Standard reference works setting forth the general principles of pharmacology include Goodman and Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., McGraw Hill Companies Inc., New York (2006).


A. Identification of Phosphorylation Sites. The target signaling protein/polypeptide phosphorylation sites disclosed herein and listed in Table 1/FIG. 2 were discovered by employing the modified peptide isolation and characterization techniques described in U.S. Pat. No. 7,198,896 using cellular extracts from the following human cancer cell lines, tissues and patient samples: 01364548-cll, 223-CLL, 293T, 3T3 TrkB, 3T3-Src, 3T3-TrkA, 3T3-wt, 577, A172, AML-4833, AML-6246, AML-6735, AML-7592, BaF3-10ZF, BaF3-4ZF, BaF3-APR, BaF3-FLT3(D842V), BaF3-FLT3(D842Y), BaF3-FLT3(K663Q), BaF3-FLT3(WT), BaF3-FLT3/ITD, BaF3-PRTK, BaF3-TDII, BaF3-Tel/FGFR3, Baf3, Baf3-V617F-jak2, Baf3/E255K, Baf3/H396P, Baf3/Jak2(IL-3 dep), Baf3/M35 IT, Baf3/T315I, Baf3/TpoR, Baf3/TpoR-Y98F, Baf3/Tyk2, Baf3/V617F-jak2 (IL-3), Baf3/Y253F, Baf3/cc-TpoR-IV, Baf3/p210wt, CHRF, CI-1, CMK, CTV-1, DMS 53, DND41, DU-528, DU145, ELF-153, EOL-1, GDM-1, H1703, H1734, H1793, H1869, H1944, H1993, H2023, H226, H3255, H358, H520, H82, H838, HCC1428, HCC1435, HCC1806, HCC1937, HCC366, HCC827, HCT116, HEL, HL107B, HL117B, HL131A, HL131B, HL133A, HL53B, HL59b, HL60, HL61a, HL61b, HL66B, HL68A, HL75A, HL84A, HL97B, HL98A, HT29, HU-3, HUVEC, Jurkat, K562, KG-1, KG1-A, KMS11, KMS18, KMS27, KOFT-K1, KY821, Karpas 299, Karpas-1106p, M-07e, M01043, M059K, MC-116, MCF-10A (Y561F), MCF-10A(Y969F), MDA-MB-453, MDA-MB-468, MEC-2, MKPL-1, ML-1, MO-91, MOLT15, MV4-11, Me-F2, Molm 14, Monomac 6, NCI-N87, Nomo-1, OCI-M1, OCI-ly4, OCI-ly8, OCI/AML2, OPM-1, PL21, Pfeiffer, RC-K8, RI-1, SCLC T1, SEM, SK-N-AS, SK-N-MC, SKBR3, SR-786, SU-DHL1, SUP-M2, SUPT-13, SuDHL5, T17, TRE-cll patient, TS, UT-7, VAL, Verona, Verona 1, Verona 4, WSU-NHL, XG2, Z-55, cs001, cs015, cs025, cs041, cs042, gz21, gz68, gz73, gz74, gzB1, h1144b, h1152b, lung tumor T26, lung tumor T57, normal human lung, pancreatic xenograft, patient 1, rat brain, sw480. The isolation and identification of phosphopeptides from these cell lines, using an immobilized general phosphotyrosine-specific antibody, or an antibody recognizing the phosphorylated motif PXpSP is described in detail in Example 1 below. In addition to the protein phosphorylation sites (tyrosine) described herein, many known phosphorylation sites were also identified (not described herein). The immunoaffinity/mass spectrometric technique described in the '896 Patent (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). 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.


In the IAP method as employed herein, a general phosphotyrosine-specific monoclonal antibody (commercially available from Cell Signaling Technology, Inc., Beverly, Mass., 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 following human cancer cell lines, tissues and patient samples were employed: 01364548-cll, 223-CLL, 293T, 3T3 TrkB, 3T3-Src, 3T3-TrkA, 3T3-wt, 577, A172, AML-4833, AML-6246, AML-6735, AML-7592, BaF3-10ZF, BaF3-4ZF, BaF3-APR, BaF3-FLT3(D842V), BaF3-FLT3(D842Y), BaF3-FLT3(K663Q), BaF3-FLT3(WT), BaF3-FLT3/ITD, BaF3-PRTK, BaF3-TDII, BaF3-Tel/FGFR3, Baf3, Baf3-V617F-jak2, Baf3/E255K, Baf3/H396P, Baf3/Jak2(IL-3 dep), Baf3/M351T, Baf3/T315I, Baf3/TpoR, Baf3/TpoR-Y98F, Baf3/Tyk2, Baf3/V617F-jak2 (IL-3), Baf3/Y253F, Baf3/cc-TpoR-IV, Baf3/p210wt, CHRF, CI-1, CMK, CTV-1, DMS 53, DND41, DU-528, DU145, ELF-153, EOL-1, GDM-1, H1703, H1734, H1793, H1869, H1944, H1993, H2023, H226, H3255, H358, H520, H82, H838, HCC1428, HCC1435, HCC1806, HCC1937, HCC366, HCC827, HCT116, HEL, HL107B, HL117B, HL131A, HL131B, HL133A, HL53B, HL59b, HL60, HL61a, HL61b, HL66B, HL68A, HL75A, HL84A, HL97B, HL98A, HT29, HU-3, HUVEC, Jurkat, K562, KG-1, KG1-A, KMS11, KMS18, KMS27, KOPT-K1, KY821, Karpas 299, Karpas-1106p, M-07e, M01043, M059K, MC-116, MCF-10A (Y561F), MCF-10A(Y969F), MDA-MB-453, MDA-MB-468, MEC-2, MKPL-1, ML-1, MO-91, MOLT15, MV4-11, Me-F2, Molm 14, Monomac 6, NCI-N87, Nomo-1, OCI-M1, OCI-ly4, OCI-1y8, OCI/AML2, OPM-1, PL21, Pfeiffer, RC-K8, RI-1, SCLC T1, SEM, SK-N-AS, SK-N-MC, SKBR3, SR-786, SU-DHL1, SUP-M2, SUPT-13, SuDHL5, T17, TRE-cll patient, TS, UT-7, VAL, Verona, Verona 1, Verona 4, WSU-NHL, XG2, Z-55, cs001, cs015, cs025, cs041, cs042, gz21, gz68, gz73, gz74, gzB1, h1144b, h1152b, lung tumor T26, lung tumor T57, normal human lung, pancreatic xenograft, patient 1, rat brain and sw480.


As described in more detail in the Examples, lysates were prepared from these cells and digested with trypsin after treatment with DTT and iodoacetamide to redue and alkylate cysteine residues. Before the immunoaffinity step, peptides were pre-fractionated by reversed-phase solid phase extraction using Sep-Pak C18 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 IP buffer and treated with phosphotyrosine (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 either a LCQ or ThermoFinnigan LTQ ion trap mass spectrometer. Peptides were eluted from a 10 cm×75 μm 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.


This revealed the tyrosine phosphorylation sites in signaling pathways affected by kinase activation or active in leukemia cells. The identified phosphorylation sites and their parent proteins are enumerated in Table 1/FIG. 2. The tyrosine at which phosphorylation occurs is provided in Column D, and the peptide sequence encompassing the phosphorylatable tyrosine residue at the site is provided in Column E. If a phosphorylated tyrosine was found in mouse, the orthologous site in human was identified using either Homologene or BLAST at NCBI; the sequence reported in column E is the phosphorylation site flanked by 7 amino acids on each side. FIG. 2 also shows the particular type of leukemic disease (see Column G) and cell line(s) (see Column F) in which a particular phosphorylation site was discovered.


As a result of the discovery of these phosphorylation sites, phospho-specific antibodies and AQUA peptides for the detection of and quantification of these sites and their parent proteins may now be produced by standard methods, as described below. These new reagents will prove highly useful in, e.g., studying the signaling pathways and events underlying the progression of leukemias and the identification of new biomarkers and targets for diagnosis and treatment of such diseases in a mammal.


The methods of the present invention are intended for use with any mammal that may experience the benefits of the methods of the invention. Foremost among such mammals are humans, although the invention is not intended to be so limited, and is applicable to veterinary uses. Thus, in accordance with the invention, “mammals” or “mammal in need” include humans as well as non-human mammals, particularly domesticated animals including, without limitation, cats, dogs, and horses.


B. Antibodies and Cell Lines. Isolated phosphorylation site-specific antibodies that specifically bind a target signaling protein/polypeptide 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/FIG. 2 may 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. The TAGLN3 cytoskeletal protein phosphorylation site (tyrosine 192) (see Row 66 of Table 1/FIG. 2) is presently disclosed. Thus, an antibody that specifically binds this novel TAGLN3 cytoskeletal site can now be produced, e.g. by immunizing an animal with 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 Row 66, Column E, of Table 1, SEQ ID NO: 65, respectively) (which encompasses the phosphorylated tyrosine at position 192 in TAGLN3, to produce an antibody that only binds TAGLN3 cytoskeletal protein when phosphorylated at that site.


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 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. For example, a peptide antigen corresponding to all or part of the novel TES cytoskeletal phosphorylation site disclosed herein (SEQ ID NO: 71=RTQYSCyCCK, encompassing phosphorylated tyrosine 237 (see Row 72 of Table 1)) may be employed to produce antibodies that only bind TES when phosphorylated at Tyr 237. Similarly, 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. If an antibody that only binds the protein when phosphorylated at the disclosed site is desired, 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)).


It will be appreciated by those of skill in the art that longer or shorter phosphopeptide antigens may be employed. See Id. For example, a peptide antigen may comprise the full sequence disclosed in Column E of Table 1/FIG. 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”). Typically, 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, for example, may be produced as described in U.S. Pat. No. 5,675,063. 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 preferable 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)).


An 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 WDR1 tyrosine 98 phosphorylation site sequence disclosed in Row 83, Column E of Table 1), and antibodies of the invention thus specifically bind a target signal protein/polypepetide comprising such epitopic sequence. 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.


Included in the scope of the invention are equivalent 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 Fab 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 al., 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 or U.S. Pat. No. 4,816,567. The antibodies may also be chemically constructed by specific antibodies made according to the method disclosed in U.S. Pat. No. 4,676,980.


The invention also provides immortalized cell lines that produce an antibody of the invention. For example, hybridoma clones, constructed as described above, that produce monoclonal antibodies to the protein phosphorylation sites disclosed herein are also provided. Similarly, 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, whether polyclonal or monoclonal, 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. 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 target signal protein/polypepetide. 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.


In an exemplary embodiment, phage display libraries containing more than 1010 phage clones are used for high-throughput production of monoclonal antibodies that target post-translational modification sites (e.g., phosphorylation sites) and, for validation and quality control, high-throughput immunohistochemistry is utilized to screen the efficacy of these antibodies. Western blots, protein microarrays and flow cytometry can also be used in high-throughput screening of phosphorylation site-specific polyclonal or monoclonal antibodies of the present invention. See, e.g., Blow N., Nature, 447: 741-743 (2007).


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 Target signaling protein/polypeptide epitope for which the antibody of the invention is specific.


In certain cases, polyclonal antisera may exhibit some undesirable general cross-reactivity to phosphotyrosine or phosphoserine 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 (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 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) staining using normal and diseased tissues to evaluate 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 al., 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° 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 target signal protein/polypepetide 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.


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.


Phosphorylation-site specific antibodies of the invention specifically bind to a target signaling protein/polypeptide 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 or identical phosphorylation sites in respective Target signaling protein/polypeptide 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 Target signaling protein/polypeptide phosphorylation sites disclosed herein.


C. Heavy-Isotope Labeled Peptides (AQUA Peptides). The 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, Gerber et al., Proc. Natl. Acad. Sci. U.S.A. 100: 6940-5 (2003).


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. Briefly, 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.


Generally, to develop a suitable internal standard, 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 (13C, 15N). 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.


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. (See Gerber et al., supra.) 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. In addition, 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-phosphorylated form of the residue developed. In this way, 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. Preferably, the size of the peptide is selected to minimize the chances that the peptide sequence will be repeated elsewhere in other non-target proteins. Thus, a peptide is preferably at least about 6 amino acids. The size of the peptide is also optimized to maximize ionization frequency. A workable range 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. Alternatively, 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). For example, 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. Preferably, 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. As a result, the labeled amino acids and peptides are readily distinguished from unlabeled ones by the ion/mass pattern in the resulting mass spectrum. Preferably, 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 2H, 13C, 15N, 17O, 18O, or 34S, are sutable labels. Pairs of peptide internal standards that incorporate a different isotope label may also be prepared. 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. The fragments are then analyzed, for example by multi-stage mass spectrometry (MSn) to obtain a fragment ion spectrum, to obtain a peptide fragmentation signature. Preferably, 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 MS3 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. Any biological sample potentially containing a target protein/peptide of interest may be assayed. Crude or partially purified cell extracts may be employed. Generally, 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 method contemplated.


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 MSn 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.


In accordance with the present invention, AQUA internal peptide standards (heavy-isotope labeled peptides) may now be produced, as described above, for any of the phosphorylation sites disclosed herein. Peptide standards for a given phosphorylation site (e.g., the tyrosine 328 in TOP2A—see Row 87 of Table 1) may be produced for both the phosphorylated and non-phosphorylated forms of the site (e.g., see PKCD site sequence in Column E, Row 123 of Table 1 (SEQ ID NO: 122) and such standards employed in the AQUA methodology to detect and quantify both forms of such phosphorylation site in a biological sample.


AQUA peptides of the invention may comprise all, or part of, a phosphorylation site peptide sequence disclosed herein (see Column E of Table 1/FIG. 2). In an embodiment, an AQUA peptide of the invention comprises a phosphorylation site sequence disclosed herein in Table 1/FIG. 2. For example, an AQUA peptide of the invention for detection/quantification of SMRT transcriptional regulator protein when phosphorylated at tyrosine Y2249 may comprise the sequence SAVyPLLYR (y=phosphotyrosine), which comprises phosphorylatable tyrosine 2249 (see Row 177, Column E; (SEQ ID NO: 176)). Heavy-isotope labeled equivalents of the peptides enumerated in Table 1/FIG. 2 (both in phosphorylated and unphosphorylated form) 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 (see Column E of Table 1/FIG. 2) are 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. Thus, heavy-isotope labeled equivalents of these peptides (both in phosphorylated and unphosphorylated form) 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.


Accordingly, the invention provides heavy-isotope labeled peptides (AQUA peptides) for the detection and/or quantification of any of the phosphorylation sites disclosed in Table 1/FIG. 2 (see Column E) and/or their corresponding parent proteins/polypeptides (see Column A). A phosphopeptide sequence comprising any of the phosphorylation sequences listed in Table 1 may be considered an AQUA peptide of the invention. For example, an AQUA peptide comprising the sequence SAVyPLLYR (SEQ ID NO: 176) (where y may be either phosphotyrosine or tyrosine, and where V=labeled valine (e.g., 14C)) is provided for the quantification of phosphorylated (or non-phosphorylated) diaphanous (Tyr2249) in a biological sample (see Row 177 of Table 1, tyrosine 2249 being the phosphorylatable residue within the site). It will be appreciated that a larger AQUA peptide comprising a disclosed phosphorylation site sequence (and additional residues downstream or upstream of it) may also be constructed. Similarly, a smaller 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 of Table 1/FIG. 2) may alternatively be constructed. Such larger or shorter AQUA peptides are within the scope of the present invention, and the selection and production of AQUA peptides may be carried out as described above (see Gygi et al., Gerber et al., supra.).


Certain subsets of AQUA peptides provided by the invention are described above (corresponding to particular protein types/groups in Table 1, for example, tyrosine protein 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. For example, the above-described AQUA peptides corresponding to both the phosphorylated and non-phosphorylated forms of the disclosed Tel transcriptional regulator protein tyrosine 402 phosphorylation site (see Row 213 of Table 1/FIG. 2) may be used to quantify the amount of phosphorylated Tel (Tyr 402) 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 Target signaling protein/polypeptide disclosed in Table 1/FIG. 2), and, optionally, a second detecting reagent conjugated to a detectable group. For example, 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 useful in the further study of signal transduction anomalies associated with diseases such as for example cancer, including leukemias, and in identifying diagnostic/bio-markers of these diseases, new potential drug targets, and/or in monitoring the effects of test compounds on Target Signaling Proteins/Polypeptides and pathways.


D. Immunoassay Formats. 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.


In a heterogeneous assay approach, 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. For example, if the antigen to be detected contains a second binding site, 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. Examples of 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-Immunoassay, (1980) (CRC Press, Inc., Boca Raton, Fla.); see also, e.g., U.S. Pat. No. 4,727,022; U.S. Pat. No. 4,659,678; U.S. Pat. No. 4,376,110. Conditions suitable for the formation of reagent-antibody complexes are well described. See id. Monoclonal antibodies of the invention may be used in a “two-site” or “sandwich” assay, with a single cell line serving as a source for both the labeled monoclonal antibody and the bound monoclonal antibody. Such assays are described in U.S. Pat. No. 4,376,110. The concentration of detectable reagent should be sufficient such that the binding of a Target signaling protein/polypeptide is detectable compared to background.


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., 35S, 125I, 131I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein) in accordance with known techniques.


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 signaling protein/polypeptide in patients before, during, and after treatment with a drug targeted at inhibiting phosphorylation of such a protein at the phosphorylation site disclosed herein. For example, bone marrow cells or peripheral blood cells from patients may be analyzed by flow cytometry for Target signaling protein/polypeptide 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. Chow et al., Cytometry (Communications in Clinical Cytometry) 46: 72-78 (2001). Briefly and by way of example, the following protocol for cytometric analysis may be employed: fixation of the cells with 1% para-formaldehyde for 10 minutes at 37° 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 Target Signaling Protein(s)/Polypeptide(s) in the malignant cells and reveal the drug response on the targeted protein.


Alternatively, antibodies of the invention may be employed in immunohistochemical (IHC) 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, Luminex™ and/or Bioplex™ assay formats, or otherwise optimized for antibody array formats, such as reversed-phase array applications (see, e.g., Paweletz et al., Oncogene 20(16): 1981-89 (2001)). Accordingly, in another embodiment, the invention provides a method for the multiplex detection of 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 proteins enumerated in Column A of Table 1/FIG. 2. In an embodiment, two to five antibodies or AQUA peptides of the invention are employed in the method. In another embodiment, six to ten antibodies or AQUA peptides of the invention are employed, while in another 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 Target signaling protein/polypeptide disclosed in Table 1/FIG. 2), and, optionally, a second antibody conjugated to a detectable group. In some embodies, 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.


Reference is made hereinafter in detail to specific embodiments of the invention. While the invention will be described in conjunction with these specific embodiments, it will be understood that it is not intended to limit the invention to such specific embodiments. On the contrary, it is intended to cover alternatives, modifications, and equivalents as may be included within the spirit and scope of the invention as defined by the appended claims. In the description, numerous specific details are set forth in order to provide a thorough understanding of the present invention. The present invention may be practiced without some or all of these specific details. In other instances, well known process operations have not been described in detail, in order not to unnecessarily obscure the present invention.


The following examples are intended to further illustrate certain embodiments of the invention and are not limiting in nature. Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific substances and procedures described herein.


Any suitable materials and/or methods known to those of skill can be utilized in carrying out the present invention. However, materials and methods are described. Materials, reagents and the like to which reference is made in the following description and examples are obtainable from commercial sources, unless otherwise noted.


Example 1
Isolation of Phosphotyrosine-Containing Peptides from Extracts of Cancer Cell Lines and Identification of Phosphorylation Sites

IAP isolation techniques were employed to identify phosphotyrosine containing peptides in cell extracts from the following human cancer cell lines, tissues and patient cell lines: 01364548-cll, 223-CLL, 293T, 3T3 TrkB, 3T3-Src, 3T3-TrkA, 3T3-wt, 577, A 172, AML-4833, AML-6246, AML-6735, AML-7592, BaF3-10ZF, BaF3-4ZF, BaF3-APR, BaF3-FLT3(D842V), BaF3-FLT3(D842Y), BaF3-FLT3(K663Q), BaF3-FLT3(WT), BaF3-FLT3/ITD, BaF3-PRTK, BaF3-TDII, BaF3-Tel/FGFR3, Baf3, Baf3-V617F-jak2, Baf3/E255K, Baf3/H396P, Baf3/Jak2(IL-3 dep), Baf3/M351T, Baf3/T315I, Baf3/TpoR, Baf3/TpoR-Y98F, Baf3/Tyk2, Baf3/V617F-jak2 (IL-3), Baf3/Y253F, Baf3/cc-TpoR-IV, Baf3/p210wt, CHRF, CI-1, CMK, CTV-1, DMS 53, DND41, DU-528, DU145, ELF-153, EOL-1, GDM-1, H1703, H1734, H1793, H1869, H1944, H1993, H2023, H226, H3255, H358, H520, H82, H838, HCC1428, HCC1435, HCC1806, HCC1937, HCC366, HCC827, HCT116, HEL, HL107B, HL117B, HL131A, HL131B, HL133A, HL53B, HL59b, HL60, HL61a, HL61b, HL66B, HL68A, HL75A, HL84A, HL97B, HL98A, HT29, HU-3, HUVEC, Jurkat, K562, KG-1, KG1-A, KMS11, KMS18, KMS27, KOPT-K 1, KY821, Karpas 299, Karpas-1106p, M-07e, M01043, M059K, MC-116, MCF-10A (Y561F), MCF-10A(Y969F), MDA-MB-453, MDA-MB-468, MEC-2, MKPL-1, ML-1, MO-91, MOLT15, MV4-11, Me-F2, Molm 14, Monomac 6, NCI-N87, Nomo-1, OCI-M1, OCI-ly4, OCI-1y8, OCI/AML2, OPM-1, PL21, Pfeiffer, RC-K8, RI-1, SCLC T1, SEM, SK-N-AS, SK-N-MC, SKBR3, SR-786, SU-DHL1, SUP-M2, SUPT-13, SuDHL5, T17, TRE-cll patient, TS, UT-7, VAL, Verona, Verona 1, Verona 4, WSU-NHL, XG2, Z-55, cs001, cs015, cs025, cs041, cs042, gz21, gz68, gz73, gz74, gzB1, h1144b, hl 152b, lung tumor T26, lung tumor T57, normal human lung, pancreatic xenograft, patient 1, rat brain and sw480.


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.


Suspension cells were harvested by low speed centrifugation. After complete aspiration of medium, cells were resuspended in 1 mL lysis buffer per 1.25×108 cells (20 mM HEPES pH 8.0, 9 M urea, 1 mM sodium vanadate, supplemented or not with 2.5 mM sodium pyro-phosphate, 1 mM β-glycerol-phosphate) and sonicated.


Sonicated cell lysates were cleared by centrifugation at 20,000×g, and proteins were reduced with DTT at a final concentration of 4.1 mM and alkylated with iodoacetamide at 8.3 mM. For digestion with trypsin, protein extracts were diluted in 20 mM HEPES pH 8.0 to a final concentration of 2 M urea and soluble TLCK®-trypsin (Worthington® Biochemcial Corporation, Lakewood, N.J.) was added at 10-20 μg/mL. Digestion was performed for 1-2 days at room temperature.


Trifluoroacetic acid (TFA) was added to protein digests to a final concentration of 1%, precipitate was removed by centrifugation, and digests were loaded onto Sep-Pak® C18 columns (provided by Waters Corporation, Milford, Mass.) equilibrated with 0.1% TFA. A column volume of 0.7-1.0 ml was used per 2×108 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 II 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 2×108 cells were dissolved in 1 ml of IAP buffer (20 mM Tris/HCl or 50 mM MOPS pH 7.2, 10 mM sodium phosphate, 50 mM NaCl) and insoluble material was removed by centrifugation. IAP was performed on each peptide fraction separately. The phosphotyrosine monoclonal antibody P-Tyr-100 (Cell Signaling Technology®, Inc., Danvers, Mass. catalog number 9411) was coupled at 4 mg/ml beads to protein G or protein A agarose (Roche®, Basel, Switzerland), respectively. Immobilized antibody (15 μl, 60 μg) was added as 1:1 slurry in IAP buffer to 1.4 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 μl of 0.1% TFA at room temperature for 10 minutes.


Alternatively, 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 NaCl) and insoluble material was removed by centrifugation. Immobilized antibody (40 μl, 160 μg) 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 40 μl of 0.15% TFA at room temperature for 10 min (eluate 1), followed by a wash of the beads (eluate 2) with 40 μl of 0.15% TFA. Both eluates were combined.


Analysis by LC-MS/MS Mass Spectrometry.

40 μl or more of IAP eluate were purified by 0.2 μl StageTips (Proxeon, Staermosegaardsvej 6, DK-5230 Odense M, Denmark) or ZipTips® (produced by Millipore®, Billerica Mass.). Peptides were eluted from the microcolumns with 1 μl of 40% MeCN, 0.1% TFA (fractions I and II) or 1 μl of 60% MeCN, 0.1% TFA (fraction III) into 7.6 μl of 0.4% acetic acid/0.005% heptafluorobutyric acid. This sample was loaded onto a 10 cm×75 μm PicoFrit® capillary column (produced by New Objective, Woburn, Mass.) packed with Michrom Magic Bullets® C18 AQ reversed-phase resin (Michrom Bioresources, Auburn Calif.) using a Famos™ autosampler with an inert sample injection valve (Dionex®, Sunnyvale, Calif.). The column was then developed with a 45-min linear gradient of acetonitrile delivered at 200 nl/min (using an Ultimate® pump, Dionex®, Sunnyvale, Calif.), and tandem mass spectra were collected in a data-dependent manner with an LTQ® (produced by Thermo® Finnigan® San, Jose, Calif.), ion trap mass spectrometer essentially as described by Gygi et al., supra.


Database Analysis & Assignments.

MS/MS spectra were evaluated using TurboSequest™ in the Sequest® (owned by Thermo® Finnigan® San Jose, Calif.) Browser package (v. 27, rev. 12) supplied as part of BioWorks™ 3.0 (Thermo® Finnigan®, San Jose, Calif.). Individual MS/MS spectra were extracted from the raw data file using the Sequest® Browser program CreateDta™ (owned by Thermo® Finnigan® San Jose, Calif.), with the following settings: bottom MW, 700; top MW, 4,500; minimum number of ions, 20; minimum TIC, 4×105; 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™ (owned by Thermo® Finnigan® San Jose, Calif.) 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 (as released on Aug. 24, 2004 and containing 27, 960 protein sequences). 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. Furthermore, it should be noted that certain peptides were originally isolated in mouse and later normalized to human sequences as shown by Table 1/FIG. 2.


In proteomics research, it is desirable to validate protein identifications based solely on the observation of a single peptide in one experimental result, in order to indicate that the protein is, in fact, present in a sample. This has led to the development of statistical methods for validating peptide assignments, which are not yet universally accepted, and guidelines for the publication of protein and peptide identification results (see Carr et al., Mol. Cell. Proteomics 3: 531-533 (2004)), which were followed in this Example. However, because the immunoaffinity strategy separates phosphorylated peptides from unphosphorylated peptides, observing just one phosphopeptide from a protein is a common result, since many phosphorylated proteins have only one tyrosine-phosphorylated site. For this reason, it is appropriate to use additional criteria to validate phosphopeptide assignments. Assignments are likely to be correct if any of these additional criteria are met: (i) the same sequence is assigned to co-eluting ions with different charge states, since the MS/MS spectrum changes markedly with charge state; (ii) the site is found in more than one peptide sequence context due to sequence overlaps from incomplete proteolysis or use of proteases other than trypsin; (iii) the site is found in more than one peptide sequence context due to homologous but not identical protein isoforms; (iv) the site is found in more than one peptide sequence context due to homologous but not identical proteins among species; and (v) sites validated by MS/MS analysis of synthetic phosphopeptides corresponding to assigned sequences, since the ion trap mass spectrometer produces highly reproducible MS/MS spectra. 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. The following 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.


In the first step, 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).


In the second step, assignments with below-threshold scores should be accepted if the low-scoring spectrum shows a high degree of similarity to a high-scoring spectrum collected in another study, which simulates a true reference library-searching strategy.


Example 2
Production of Phospho-Specific Polyclonal Antibodies for the Detection of Target Signal Protein/Polypepetide Phosphorylation

Polyclonal antibodies that specifically bind a target signal protein/polypepetide only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1/FIG. 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.


A. VCP (Tyrosine 644).

A 13 amino acid phospho-peptide antigen, LDKLIy*IPLPDEK (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 644 phosphorylation site in human VCP cell cycle regulation protein (see Row 28 of Table 1; SEQ ID NO: 27), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. This peptide is then coupled to KLH and used to immunize animals to produce (and subsequently screen) phospho-specific VCP (tyr643) polyclonal antibodies as described in Immunization/Screening below.


B. HSP90B (Tyrosine 192).

An 16 amino acid phospho-peptide antigen, VILHLKEDQTEy*LEER (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 192 phosphorylation site in human HSP90B chaperone protein (see Row 30 of Table 1 (SEQ ID NO: 29)), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. This peptide is then coupled to KLH and used to immunize animals to produce (and subsequently screen) phospho-specific HSP90B (tyr 191) polyclonal antibodies as described in Immunization/Screening below.


C. TSN (Tyrosine 210).

A 12 amino acid phospho-peptide antigen, KVEEVVy*DLSIR (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 210 phosphorylation site in human catalase chromatin or DNA binding/repair/replication protein (see Row 44 of Table 1 (SEQ ID NO: 43), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. This peptide is then coupled to KLH and used to immunize animals to produce (and subsequently screen) phospho-specific TSN (tyr 210) antibodies as described in Immunization/Screening below.


Immunization/Screening.

A synthetic phospho-peptide antigen as described in A-C above is coupled to KLH, and rabbits are injected intradermally (ID) on the back with antigen in complete Freunds adjuvant (384 μg antigen per rabbit). The rabbits are boosted with same antigen in incomplete Freund adjuvant (250 μg 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. After washing the column extensively, 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 antibody storage buffer.


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 VCP, HSP90B or TSN), for example, CTV, CMK and MOLT15 cells, 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° C. for 5 minutes. 20 μl (10 μg protein) of sample is then added onto 7.5% SDS-PAGE gel.


A standard Western blot may be performed according to the Immunoblotting 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. TSN is not bound when not phosphorylated at tyrosine 210).


In order to confirm the specificity of the isolated antibody, different cell lysates containing various phosphorylated signal transduction proteins other than the target protein are prepared. The Western blot assay is performed again using these cell lysates. The phospho-specific polyclonal antibody isolated as described above is used (1:1000 dilution) to test reactivity with the different phosphorylated non-target proteins on Western blot membrane. The phospho-specific antibody does not significantly cross-react with other phosphorylated signal transduction proteins, although occasionally slight binding with a highly homologous phosphorylation-site on another protein may be observed. In such case the antibody may be further purified using affinity chromatography, or the specific immunoreactivity cloned by rabbit hybridoma technology.


Example 3
Production of Phospho-Specific Monoclonal Antibodies for the Detection of Target Signal Protein/Polypepetide Phosphorylation

Monoclonal antibodies that specifically bind a target signal protein/polypepetide only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1/FIG. 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, and harvesting spleen cells from such animals to produce fusion hybridomas, as further described below. Production of exemplary monoclonal antibodies is provided below.


A. WRN (Tyrosine 849).

An 11 amino acid phospho-peptide antigen, DMESYy*QEIGR (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 849 phosphorylation site in human WRN chromatin or DNA binding/repair/replication protein (see Row 51 of Table 1 (SEQ ID NO: 50)), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. 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 WRN (tyr 849) antibodies as described in Immunization/Fusion/Screening below.


B. SPTA1 (Tyrosine 1538).

An 11 amino acid phospho-peptide antigen, DATNIQRKy*LK (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 1538 phosphorylation site in human SPTA1 cytoskeletal protein (see Row 63 of Table 1 (SEQ ID NO: 62)), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. 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 SPTA1 (tyr1538) antibodies as described in Immunization/Fusion/Screening below.


C. SPTBN1 (tyrosine 1667).


A 15 amino acid phospho-peptide antigen, VDKLy*AGLKDLAEER (where y*=phosphotyrosine) that corresponds to the sequence encompassing the tyrosine 1667 phosphorylation site in human SPTBN1 cytoskeletal protein (see Row 61 of Table 1 (SEQ ID NO: 60), plus cysteine on the C-terminal for coupling, is constructed according to standard synthesis techniques using, e.g., a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra. 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 SPTBN1 (tyr1667) antibodies as described in Immunization/Fusion/Screening below.


Immunization/Fusion/Screening.

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 μg antigen per mouse). The mice are boosted with same antigen in incomplete Freund adjuvant (e.g. 25 μg 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 WRN, SFTA1 or SPTBN1 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. SPTA1 phosphorylated at tyrosine 1538).


Example 4
Production and Use of Aqua Peptides for the Quantification of Target Signal Protein/Polypepetide Phosphorylation

Heavy-isotope labeled peptides (AQUA peptides (internal standards)) for the detection and quantification of a target signal protein/polypepetide only when phosphorylated at the respective phosphorylation site disclosed herein (see Table 1/FIG. 2) are produced according to the standard AQUA methodology (see Gygi et al., 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 MSn 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.


A. VASP (Tyrosine 15).

An AQUA peptide comprising the sequence, ATVMLy*DDGNKR (y*=phosphotyrosine; sequence incorporating 14C/15N-labeled leucine (indicated by bold L), which corresponds to the tyrosine 16 phosphorylation site in human VASP cytoskeletal protein (see Row 79 in Table 1 (SEQ ID NO: 78)), 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 VASP (tyr 16) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated VASP (tyr 16) in the sample, as further described below in Analysis & Quantification.


B. TOP2B (Tyrosine 230).

An AQUA peptide comprising the sequence IKHFDGEDy*TCITFTQPDLSK (y*=phosphotyrosine; sequence incorporating 14C/15N-labeled leucine (indicated by bold L), which corresponds to the tyrosine 230 phosphorylation site in human TOP2B chromatin or DNA binding/repair/replication protein (see Row 42 in Table 1 (SEQ ID NO: 41)), 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 TOP2B (tyr230) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated TOP2B (tyr230) in the sample, as further described below in Analysis & Quantification.


C. PKCD (Tyrosine 374)

An AQUA peptide comprising the sequence GRGEy*FAIK (y*=phosphotyrosine; sequence incorporating 14C/15N-labeled phenylalanine (indicated by bold F), which corresponds to the tyrosine 374 phosphorylation site in human PKCD protein kinase (Ser/Thr) (see Row 123 in Table 1 (SEQ ID NO: 122)), 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 PKCD (tyr374) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated PKCD (tyr374) in the sample, as further described below in Analysis & Quantification.


D. TAGLN3 (Tyrosine 192).

An AQUA peptide comprising the sequence, GASQAGMTGy*GMPR (y*=phosphotyrosine; sequence incorporating 14C/15N-labeled proline (indicated by bold P), which corresponds to the tyrosine 133 phosphorylation site in human TAGLN3 cytoskeletal protein (see Row 66 in Table 1 (SEQ ID NO: 65)), 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 TAGLN3 (tyr192) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated TAGLN3 (tyr192) in the sample, as further described below in Analysis & Quantification.


Synthesis & MS/MS Spectra.

Fluorenylmethoxycarbonyl (Fmoc)-derivatized amino acid monomers may be obtained from AnaSpec (San Jose, Calif.). Fmoc-derivatized stable-isotope monomers containing one 15N and five to nine 13C atoms may be obtained from Cambridge Isotope Laboratories (Andover, Mass.). Preloaded Wang resins may be obtained from Applied Biosystems. Synthesis scales may vary from 5 to 25 μmol. Amino acids are activated in situ with 1-H-benzotriazolium, 1-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 by-products. 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, Mass.) and ion-trap (ThermoFinnigan, LCQ DecaXP) MS.


MS/MS spectra for each AQUA peptide should exhibit a strong γ-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 Ř150-220 mm) are prepared according to standard methods. An Agilent 1100 liquid chromatograph may be used to develop and deliver a solvent gradient 10.4% 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. 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.


Analysis & Quantification.

Target protein (e.g. a phosphorylated protein of A-D above) in a biological sample is quantified using a validated 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, Calif.) mass spectrometer (LTQ ion trap or TSQ Quantum triple quadrupole). On the LTQ, parent ions are isolated at 1.6 m/z width, the ion injection time being limited to 100 ms per microscan, with one microscans per peptide, and with an AGC setting of 1×105; 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. On both instruments, 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., 384 fmol).

Claims
  • 1. (canceled)
  • 2. (canceled)
  • 3. (canceled)
  • 4. (canceled)
  • 5. (canceled)
  • 6. (canceled)
  • 7. (canceled)
  • 8. (canceled)
  • 9. (canceled)
  • 10. (canceled)
  • 11. (canceled)
  • 12. (canceled)
  • 13. (canceled)
  • 14. (canceled)
  • 15. (canceled)
  • 16. (canceled)
  • 17. (canceled)
  • 18. (canceled)
  • 19. (canceled)
  • 20. (canceled)
  • 21. (canceled)
  • 22. (canceled)
  • 23. (canceled)
  • 24. (canceled)
  • 25. (canceled)
  • 26. (canceled)
  • 27. (canceled)
  • 28. (canceled)
  • 29. (canceled)
  • 30. (canceled)
  • 31. (canceled)
  • 32. (canceled)
  • 33. (canceled)
  • 34. (canceled)
  • 35. (canceled)
  • 36. (canceled)
  • 37. (canceled)
  • 38. (canceled)
  • 39. (canceled)
  • 40. (canceled)
  • 41. (canceled)
  • 42. (canceled)
  • 43. (canceled)
  • 44. (canceled)
  • 45. (canceled)
  • 46. An isolated phosphorylation site-specific antibody that specifically binds a human signaling protein selected from Column A of Table 1, Rows 123, 66, 30, 140 and 194 only when phosphorylated at the tyrosine 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: 122, 65, 29, 139 and 193), wherein said antibody does not bind said signaling protein when not phosphorylated at said tyrosine.
  • 47. An isolated phosphorylation site-specific antibody that specifically binds a human signaling protein selected from Column A of Table 1, Rows 123, 66, 30, 140 and 194 only when not phosphorylated at the tyrosine 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: 122, 65, 29, 139 and 193), wherein said antibody does not bind said signaling protein when phosphorylated at said tyrosine.
  • 48. A method selected from the group consisting of: (a) a method for detecting a human signaling protein selected from Column A of Table 1, Rows 123, 66, 30, 140 and 194 wherein said human signaling protein is phosphorylated at the tyrosine 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: 122, 65, 29, 139 and 193), comprising the step of adding an isolated phosphorylation-specific antibody according to claim 46, to a sample comprising said human signaling protein under conditions that permit the binding of said antibody to said human signaling protein, and detecting bound antibody;(b) a method for quantifying the amount of a human signaling protein listed in Column A of Table 1, Rows 123, 66, 30, 140 and 194 that is phosphorylated at the corresponding tyrosine listed in Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 122, 65, 29, 139 and 193), in a sample using a heavy-isotope labeled peptide (AQUA™ peptide), said labeled peptide comprising a phosphorylated tyrosine at said corresponding lysine listed Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 as an internal standard; and(c) a method comprising step (a) followed by step (b).
  • 49. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding PKCD only when phosphorylated at Y374, comprised within the phosphorylatable peptide sequence listed in Column E, Row 123, of Table 1 (SEQ ID NO: 122), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • 50. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding PKCD only when not phosphorylated at Y374, comprised within the phosphorylatable peptide sequence listed in Column E, Row 123, of Table 1 (SEQ ID NO: 122), wherein said antibody does not bind said protein when phosphorylated at said tyrosine.
  • 51. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding TAGLN3 only when phosphorylated at Y192, comprised within the phosphorylatable peptide sequence listed in Column E, Row 66, of Table 1 (SEQ ID NO: 65), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • 52. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding TAGLN3 only when not phosphorylated at Y192, comprised within the phosphorylatable peptide sequence listed in Column E, Row 66, of Table 1 (SEQ ID NO: 65), wherein said antibody does not bind said protein when phosphorylated at said tyrosine.
  • 53. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding HSP90B only when phosphorylated at Y192, comprised within the phosphorylatable peptide sequence listed in Column E, Row 30, of Table 1 (SEQ ID NO: 29), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • 54. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding HSP90B only when not phosphorylated at Y192, comprised within the phosphorylatable peptide sequence listed in Column E, Row 30, of Table 1 (SEQ ID NO: 29), wherein said antibody does not bind said protein when phosphorylated at said tyrosine.
  • 55. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding Tyro3 only when phosphorylated at Y685, comprised within the phosphorylatable peptide sequence listed in Column E, Row 140, of Table 1 (SEQ ID NO: 139), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • 56. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding Tyro3 only when not phosphorylated at Y685, comprised within the phosphorylatable peptide sequence listed in Column E, Row 140, of Table 1 (SEQ ID NO: 139), wherein said antibody does not bind said protein when phosphorylated at said tyrosine.
  • 57. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding STAT5B only when phosphorylated at Y683, comprised within the phosphorylatable peptide sequence listed in Column E, Row 194, of Table 1 (SEQ ID NO: 193), wherein said antibody does not bind said protein when not phosphorylated at said tyrosine.
  • 58. The method of claim 48, wherein said isolated phosphorylation-specific antibody is capable of specifically binding STAT5B only when not phosphorylated at Y683, comprised within the phosphorylatable peptide sequence listed in Column E, Row 194, of Table 1 (SEQ ID NO: 193), wherein said antibody does not bind said protein when phosphorylated at said tyrosine.
RELATED APPLICATIONS

Pursuant to 35 U.S.C. §119(e) this application claims the benefit of, and priority to, provisional application U.S. Ser. No. 60/830,724, filed Jul. 13, 2006, the disclosure of which is incorporated herein, in its entirety, by reference.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US07/73537 7/13/2007 WO 00 2/4/2010
Provisional Applications (1)
Number Date Country
60830724 Jul 2006 US