Tyrosine phosphorylation sites

Abstract
The invention discloses 482 novel phosphorylation sites identified in carcinoma and/or leukemia, peptides (including AQUA peptides) comprising a phosphorylation site of the invention, antibodies specifically bind to a novel phosphorylation site of the invention, and diagnostic and therapeutic uses of the above.
Description
FIELD OF THE INVENTION

The invention relates generally to novel tyrosine phosphorylation sites, methods and compositions for detecting, quantitating and modulating same.


BACKGROUND OF THE INVENTION

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. Protein phosphorylation, for example, plays a critical role in the etiology of many pathological conditions and diseases, including to mention but a few: cancer, developmental disorders, autoimmune diseases, and diabetes. Yet, in spite of the importance of protein modification, it is not yet well understood at the molecular level, due to the extraordinary complexity of signaling pathways, and the slow development of technology necessary to unravel it.


Protein phosphorylation on a proteome-wide scale is extremely complex as a result of three factors: the large number of modifying proteins, e.g., kinases, encoded in the genome, the much larger number of sites on substrate proteins that are modified by these enzymes, and the dynamic nature of protein expression during growth, development, disease states, and aging. The human genome, for example, encodes over 520 different protein kinases, making them the most abundant class of enzymes known. (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.


Many of these phosphorylation sites regulate critical biological processes and may prove to be important diagnostic or therapeutic targets for molecular medicine. 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 disease states like cancer.


Carcinoma and/or leukemia is one of the two main categories of cancer, and is generally characterized by the formation of malignant tumors or cells of epithelial tissue original, such as skin, digestive tract, glands, etc. Carcinoma and/or leukemias are malignant by definition, and tend to metastasize to other areas of the body. The most common forms of carcinoma and/or leukemia are skin cancer, lung cancer, breast cancer, and colon cancer, as well as other numerous but less prevalent carcinoma and/or leukemias. Current estimates show that, collectively, various carcinoma and/or leukemias will account for approximately 1.65 million cancer diagnoses in the United States alone, and more than 300,000 people will die from some type of carcinoma and/or leukemia during 2005. (Source: American Cancer Society (2005)). The worldwide incidence of carcinoma and/or leukemia is much higher.


As with many cancers, deregulation of receptor tyrosine kinases (RTKs) appears to be a central theme in the etiology of carcinoma and/or leukemias. Constitutively active RTKs can contribute not only to unrestricted cell proliferation, but also to other important features of malignant tumors, such as evading apoptosis, the ability to promote blood vessel growth, the ability to invade other tissues and build metastases at distant sites (see Blume-Jensen et al., Nature 411: 355-365 (2001)). These effects are mediated not only through aberrant activity of RTKs themselves, but, in turn, by aberrant activity of their downstream signaling molecules and substrates.


The importance of RTKs in carcinoma and/or leukemia progression has led to a very active search for pharmacological compounds that can inhibit RTK activity in tumor cells, and more recently to significant efforts aimed at identifying genetic mutations in RTKs that may occur in, and affect progression of, different types of carcinoma and/or leukemias (see, e.g., Bardell et al., Science 300: 949 (2003); Lynch et al., N. Eng. J. Med. 350: 2129-2139 (2004)). For example, non-small cell lung carcinoma and/or leukemia patients carrying activating mutations in the epidermal growth factor receptor (EGFR), an RTK, appear to respond better to specific EGFR inhibitors than do patients without such mutations (Lynch et al., supra.; Paez et al., Science 304: 1497-1500 (2004)).


Clearly, identifying activated RTKs and downstream signaling molecules driving the oncogenic phenotype of carcinoma and/or leukemias would be highly beneficial for understanding the underlying mechanisms of this prevalent form of cancer, identifying novel drug targets for the treatment of such disease, and for assessing appropriate patient treatment with selective kinase inhibitors of relevant targets when and if they become available. The identification of key signaling mechanisms is highly desirable in many contexts in addition to cancer.


Leukemia, another form of cancer, is a disease in which a number of underlying signal transduction events have been elucidated and which 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.).


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 ST1571 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, 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)).


Although a few key RTKs and various other signaling proteins involved in carcinoma and/or leukemia and leukemia progression are, known, there is relatively scarce information about kinase-driven signaling pathways and phosphorylation sites that underlie the different types of cancer. Therefore there is presently an incomplete and inaccurate 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 ontogenesis in cancer by identifying the downstream signaling proteins mediating cellular transformation in these cancers.


Presently, diagnosis of many types of cancer is often made by tissue biopsy and detection of different cell surface markers. However, misdiagnosis can occur since certain types of cancer 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 cancer 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 diseases including for example, carcinoma and/or 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 many diseases.


SUMMARY OF THE INVENTION

The present invention provides in one aspect novel tyrosine phosphorylation sites (Table 1) identified in carcinoma and/or leukemia. The novel sites occur in proteins such as: Adaptor/Scaffold proteins, adhesion/extra cellular matrix proteins, cell cycle regulation, chaperone proteins, chromatin or DNA binding/repair/proteins, cytoskeleton proteins, endoplasmic reticulum or golgi proteins, enzyme proteins, g proteins or regulator proteins, kinases, protein kinases receptor/channel/transporter/cell surface proteins, transcriptional regulators, ubiquitan conjugating proteins, RNA processing proteins, secreted proteins, motor or contractile proteins, apoptosis proteins of unknown function and vesicle proteins.


In another aspect, the invention provides peptides comprising the novel phosphorylation sites of the invention, and proteins and peptides that are mutated to eliminate the novel phosphorylation sites.


In another aspect, the invention provides modulators that modulate tyrosine phosphorylation at a novel phosphorylation site of the invention, including small molecules, peptides comprising a novel phosphorylation site, and binding molecules that specifically bind at a novel phosphorylation site, including but not limited to antibodies or antigen-binding fragments thereof.


In another aspect, the invention provides compositions for detecting, quantitating or modulating a novel phosphorylation site of the invention, including peptides comprising a novel phosphorylation site and antibodies or antigen-binding fragments thereof that specifically bind at a novel phosphorylation site. In certain embodiments, the compositions for detecting, quantitating or modulating a novel phosphorylation site of the invention are Heavy-Isotype Labeled Peptides (AQUA peptides) comprising a novel phosphorylation site.


In another aspect, the invention discloses phosphorylation site specific antibodies or antigen-binding fragments thereof. In one embodiment, the antibodies specifically bind to an amino acid sequence comprising a phosphorylation site identified in Table 1 when the tyrosine identified in Column D is phosphorylated, and do not significantly bind when the tyrosine is not phosphorylated. In another embodiment, the antibodies specifically bind to an amino acid sequence comprising a phosphorylation site when the tyrosine is not phosphorylated, and do not significantly bind when the tyrosine is phosphorylated.


In another aspect, the invention provides a method for making phosphorylation site-specific antibodies.


In another aspect, the invention provides compositions comprising a peptide, protein, or antibody of the invention, including pharmaceutical compositions.


In a further aspect, the invention provides methods of treating or preventing carcinoma and/or leukemia in a subject, wherein the carcinoma and/or leukemia is associated with the phosphorylation state of a novel phosphorylation site in Table 1, whether phosphorylated or dephosphorylated. In certain embodiments, the methods comprise administering to a subject a therapeutically effective amount of a peptide comprising a novel phosphorylation site of the invention. In certain embodiments, the methods comprise administering to a subject a therapeutically effective amount of an antibody or antigen-binding fragment thereof that specifically binds at a novel phosphorylation site of the invention.


In a further aspect, the invention provides methods for detecting and quantitating phosphorylation at a novel tyrosine phosphorylation site of the invention.


In another aspect, the invention provides a method for identifying an agent that modulates tyrosine phosphorylation at a novel phosphorylation site of the invention, comprising: contacting a peptide or protein comprising a novel phosphorylation site of the invention with a candidate agent, and determining the phosphorylation state or level at the novel phosphorylation site. A change in the phosphorylation state or level at the specified tyrosine in the presence of the test agent, as compared to a control, indicates that the candidate agent potentially modulates tyrosine phosphorylation at a novel phosphorylation site of the invention.


In another aspect, the invention discloses immunoassays for binding, purifying, quantifying and otherwise generally detecting the phosphorylation of a protein or peptide at a novel phosphorylation site of the invention.


Also provided are pharmaceutical compositions and kits comprising one or more antibodies or peptides of the invention and methods of using them.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a diagram depicting the immuno-affinity isolation and mass-spectrometric characterization methodology (IAP) used in the Examples to identify the novel phosphorylation sites disclosed herein.



FIG. 2 is a table (corresponding to Table 1) summarizing the 482 novel phosphorylation sites of the invention: Column A=the parent proteins from which the phosphorylation sites are derived; Column B=the SwissProt accession number for the human homologue of the identified parent proteins; Column C=the protein type/classification; Column D=the tyrosine residues at which phosphorylation occurs (each number refers to the amino acid residue position of the tyrosine in the parent human protein, according to the published sequence retrieved by the SwissProt accession number); Column E=flanking sequences of the phosphorylatable tyrosine residues; sequences (SEQ ID NOs: 1-19, 21-22, 25-27, 29-136, 138-144, 147-204, 206-232, 234-257, 259-277, 279-301, 303-374, 376-393, 395-403, 405-435, 437-447, 449-461, 463-500) were identified using Trypsin digestion of the parent proteins; in each sequence, the tyrosine (see corresponding rows in Column D) appears in lowercase; Column F=the type of carcinoma and/or leukemia in which each of the phosphorylation site was discovered; Column G=the cell type(s)/Tissue/Patient Sample in which each of the phosphorylation site was discovered; and Column H=the SEQ ID NOs of the trypsin-digested peptides identified in Column E.



FIG. 3 is an exemplary mass spectrograph depicting the detection of the phosphorylation of tyrosine 522 in FGFR1, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (corresponds to lowercase “y” in Column E of Table 1; SEQ ID NO: 261).



FIG. 4 is an exemplary mass spectrograph depicting the detection of the phosphorylation of tyrosine 1194 in Met, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (corresponds to lowercase “y” in Column E of Table 1; SEQ ID NO: 277).



FIG. 5 is an exemplary mass spectrograph depicting the detection of the phosphorylation of tyrosine 176 in epsin 3, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (corresponds to lowercase “y” in Column E of Table 1; SEQ ID NO: 497).



FIG. 6 is an exemplary mass spectrograph depicting the detection of the phosphorylation of tyrosine 216 in FBP2, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); Y* (and pY) indicates the phosphorylated tyrosine (corresponds to lowercase “y” in Column E of Table 1; SEQ ID NO: 225).





DETAILED DESCRIPTION OF THE INVENTION

The inventors have discovered and disclosed herein novel tyrosine phosphorylation sites in signaling proteins extracted from carcinoma and/or leukemia cells. The newly discovered phosphorylation sites significantly extend our knowledge of kinase substrates and of the proteins in which the novel sites occur. The disclosure herein of the novel phosphorylation sites and reagents including peptides and antibodies specific for the sites add important new tools for the elucidation of signaling pathways that are associate with a host of biological processes including cell division, growth, differentiation, developmental changes and disease. Their discovery in carcinoma and/or leukemia cells provides and focuses further elucidation of the disease process. And, the novel sites provide additional diagnostic and therapeutic targets.


1. Novel Phosphorylation Sites in Carcinoma and/or Leukemia


In one aspect, the invention provides 482 novel tyrosine phosphorylation sites in signaling proteins from cellular extracts from a variety of human carcinoma and/or leukemia-derived cell lines and tissue samples (such as H1993, lung HCC827, etc., as further described below in Examples), identified using the techniques described in “Immunoaffinity Isolation of Modified Peptides From Complex Mixtures,” U.S. Patent Publication No. 20030044848, Rush et al, using Table 1 summarizes the identified novel phosphorylation sites.


These phosphorylation sites thus occur in proteins found in carcinoma and/or leukemia. The sequences of the human homologues are publicly available in SwissProt database and their Accession numbers listed in Column B of Table 1. The novel sites occur in proteins such as: adaptor/scaffold proteins; adhesion or extracellular matrix proteins; cytoskeletal proteins; enzyme proteins; G proteins or regulator proteins; non-protein kinase proteins; motor or contractile proteins; phosphatase proteins; protein kinases; receptor, channel, transporter or cell surface proteins; RNA processing proteins; and transcriptional regulator proteins. (see Column C of Table 1).


The novel phosphorylation sites of the invention were identified according to the methods described by Rush et al., U.S. Patent Publication No. 20030044848, which are herein incorporated by reference in its entirety. Briefly, phosphorylation sites were isolated and characterized by immunoaffinity isolation and mass-spectrometric characterization (IAP) (FIG. 1), using the following human carcinoma and/or leukemia-derived cell lines and tissue samples: 101206; 143.98.2; 23132/87; 23132/87: 10% serum; 3T3(EGFR|deletion∥EGF); 42-MG-BA; 5637; 639L; 8-MG-BA; A172; A498; A549; A704; AML-30410; B13_AML; B18_AML; BC-3C; BC004; BC005; BC007; BJ629; BJ630; BJ631; BJ635; BJ665; BT1; BT2; Baf3(FGFR1|truncation: 10ZF); Baf3(FLT3|D835Y); Baf3(FLT3|K663Q); Baf3(Jak2|Jak2|V617F); Baf3(TEL-FGFR3); CAKI-2; CAL-29; CAL-51; CAL-85-1; CCF-STTG1; CHP-212; CHP126; CHRF; CMK; CML-06/164; COLO-699; CTV-1; Caki-2; Cal-148; Calu-3; CaoV4; Colo680N; DK-MG; DMS 53; DMS 79; DU.528; DV-90; Detroit562; EFM-19; EFO-21; ENT01; ENT02; ENT03; ENT05; ENT10; ENT12; ENT14; ENT15; ENT19; ENT7; EOL-1; ES2; EVSA-T; FUOV1; GAMG; GI-CA-N; GI-LI-N; GMS-10; H1435; H1437; H1568; H1650; H1651; H1703; H1734; H1781; H1838; H1915; H1975; H2023; H2052; H2066; H2085; H2135; H2172; H2342; H2452; H28; H3255; H358; H4; H446; H4; H520; H524; H596; H810; HCC1143; HCC1395; HCC1428; HCC15; HCC1806; HCC1937; HCC827; HCT 116; HCT15; HCT8; HD-MyZ; HDLM-2; HEL; HL130A; HL131B; HL145A; HL146A; HL152A; HL183A; HL183B; HL184A; HL184B; HL1881; HL213A; HL226A; HL226B; HL233B; HL234A; HL25A; HL53A; HL53B; HL55A; HL59A; HL66A; HL68A; HL83A; HL84A; HL98A; HP28; HT29; Hs746T; IMR32; J82; Jurkat; K562; KATO III; KELLY; KG-1; KMS-11; KPL-1; Kyse140; Kyse270; Kyse410; Kyse450; Kyse510; Kyse70; L428; L540; LAN-1; LAN-5; LCLC-103H; LN-405; LN18; LXF-289; M059J; M059K; MDAH2774; MHH-NB-11; MKN-45; MKPL-1; ML-1; MT-3; MV4-11; Me-F2; MiaPaca; Molm 14; N06BJ505(2); N06BJ573(9); N06BJ591(11); N06BJ593(13); N06BJ606(19); N06CS02; N06CS06; N06CS103; N06CS106; N06CS107; N06CS16; N06CS17; N06CS22(2)-R; N06CS22-1; N06CS22-2; N06CS23; N06CS39; N06CS40; N06CS75; N06CS77; N06CS87; N06CS90; N06CS91; N06CS93-2; N06CS94; N06CS97; N06CS98; N06CS98-2; N06CS98-R; N06N101; N06N102; N06N103; N06N106; N06N121; N06N127; N06N128; N06N131; N06N80; N06N90; N06N93; N06bj523(3); N06bj567(7); N06bj594(14); N06bj595(15); N06bj638(26); N06bj639(27); N06bj667(29); N06c144; N06c78; N06cs108; N06cs109; N06cs110; N06cs110-R; N06cs112; N06cs113; N06cs117; N06cs121; N06cs122; N06cs123; N06cs123(2); N06cs126; N06cs128; N06cs132; N06cs132-1; N06cs59; N06cs63; N06cs72; N06cs76; N06cs88; N06cs92; NALM-19; NCI-H716; OPM-1; OV90; PA-1; RSK2-3; RSK2-4; S 2; SCLC T1; SCLC T4; SEM; SK-ES-1; SK-N-AS; SK-N-BE(2); SK-N-DZ; SK-N-FI; SK-OV-3; SNB-19; SNU-1; SNU-16; SNU-5; SNU-C2B; SUP-T13; SW1088; SW1710; SW480; SW620; Scaber; T17; T98G; TOV112D; TOV21G; U118 MG; UM-UC-1; UT-7; ZR-75-30; brain; cs012; cs015; cs018; cs019; cs024; cs025; cs026; cs037; cs041; cs042; cs057; cs068; cs069; cs070; cs103; cs104; cs105; cs106; cs107; cs110; cs114; cs133; cs136; csC44; csC45; csC50; csC52; csC56; csC60; csC66; csC71; gz21; gz30; gz33; gz41; gz42; gz7; gz73; gz74; gz75; gzB1; h2073; sw48. In addition to the newly discovered phosphorylation sites (all having a phosphorylatable tyrosine), many known phosphorylation sites were also identified.


The immunoaffinity/mass spectrometric technique described in Rush et al, i.e., the “IAP” method, is described in detail in the Examples and briefly summarized below.


The IAP method 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, e.g., using SILAC or AQUA, may also be used to quantify isolated peptides in order to compare peptide levels in a sample to a baseline.


In the IAP method as disclosed herein, a general phosphotyrosine-specific monoclonal antibody (commercially available from Cell Signaling Technology, Inc., Beverly, Mass., Cat #9411 (p-Tyr-100)) may be used in the immunoaffinity step to isolate the widest possible number of phospho-tyrosine containing peptides from the cell extracts.


As described in more detail in the Examples, lysates may be prepared from various carcinoma and/or leukemia cell lines or tissue samples and digested with trypsin after treatment with DTT and iodoacetamide to alkylate cysteine residues. Before the immunoaffinity step, peptides may be pre-fractionated (e.g., by reversed-phase solid phase extraction using Sep-Pak C18 columns) to separate peptides from other cellular components. The solid phase extraction cartridges may then be eluted (e.g., with acetonitrile). Each lyophilized peptide fraction can be redissolved and treated with phosphotyrosine-specific antibody (e.g., P-Tyr-100, CST #9411) immobilized on protein Agarose. Immunoaffinity-purified peptides can be eluted and a portion of this fraction may be concentrated (e.g., with Stage or Zip tips) and analyzed by LC-MS/MS (e.g., using a ThermoFinnigan LCQ Deca XP Plus ion trap mass spectrometer or LTQ). MS/MS spectra can be evaluated using, e.g., the program Sequest with the NCBI human protein database.


The novel phosphorylation sites identified are summarized in Table 1/FIG. 2. Column A lists the parent (signaling) protein in which the phosphorylation site occurs. Column D identifies the tyrosine residue at which phosphorylation occurs (each number refers to the amino acid residue position of the tyrosine in the parent human protein, according to the published sequence retrieved by the SwissProt accession number). Column E shows flanking sequences of the identified tyrosine residues (which are the sequences of trypsin-digested peptides). FIG. 2 also shows the particular type of carcinoma and/or leukemia (see Column G) and cell line(s) (see Column F) in which a particular phosphorylation site was discovered.










TABLE 1







Novel Phosphorylation Sites in Carcinoma and/or leukemia.
















Protein


Phospho-





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





  2
Cbl
NP_005179.2
Adaptor/scaffold
Y141
MyEENSQPR
SEQ ID NO: 1






  3
Cbl-b
NP_733762.2
Adaptor/scaffold
Y337
SyNPDLTGLCEPTPHDHIK
SEQ ID NO: 2





  4
CNKSR
NP_055742.2
Adaptor/scaffold
Y671
INMLTAGyAER
SEQ ID NO: 3



2





  5
CSDE1
NP_001007554.1
Adaptor/scaffold
Y138
VFyLTYTPEDVE
SEQ ID NO: 4





  6
CSDE1
NP_001007554.1
Adaptor/scaffold
Y141
VFYLTyTPEDVE
SEQ ID NO: 5





  7
CTNND
NP_001322.1
Adaptor/scaffold
Y208
NFHyPPDGYSR
SEQ ID NO: 6



1





  8
DLG3
NP_066943.2
Adaptor/scaffold
Y808
QIIEDQSGHyIWVPSPEKL
SEQ ID NO: 7





  9
DNMBP
NP_056036.1
Adaptor/scaffold
Y430
SQYySTVGGSHPHSEQYPDLLPLEAR
SEQ ID NO: 8





 10
Dok4
NP_060580.2
Adaptor/scaffold
Y165
LQITHENIyLWDIHNPR
SEQ ID NO: 9





 11
Dok4
NP_060580.2
Adaptor/scaffold
Y220
MCDAGEGLYTFQTQEGEQIyQR
SEQ ID NO: 10





 12
ENTH
NP_055481.1
Adaptor/scaffold
Y172
ySERYDPEPK
SEQ ID NO: 11





 13
ENTH
NP_055481.1
Adaptor/scaffold
Y176
YSERyDPEPK
SEQ ID NO: 12





 14
EPB41L
NP_036288.2
Adaptor/scaffold
Y68
MEEKDySEADGLSER
SEQ ID NO: 13



1





 15
FAM125
NP_612410.1
Adaptor/scaffold
Y210
RNDSIYEASSLyGISAMDGVPFTLHPR
SEQ ID NO: 14



A





 16
FAM125
NP_612410.1
Adaptor/scaffold
Y254
SLADIEEEyNYGFVVEK
SEQ ID NO: 15



A





 17
FAM125
NP_612410.1
Adaptor/scaffold
Y256
SLADIEEEYNyGFVVEK
SEQ ID NO: 16



A





 18
FCHSD
NP_055639.1
Adaptor/scaffold
Y603
PHASLPPLPLyDQPPSSPYPSPDKR
SEQ ID NO: 17



2





 19
FNBP1L
NP_060207.2
Adaptor/scaffold
Y291
SGFEPPGDFPFEDYSQHIyR
SEQ ID NO: 18





 20
FRS2
NP_006645.3
Adaptor/scaffold
Y150
TPTTPGFAAQNLPNGyPR
SEQ ID NO: 19





 21
FRS2
NP_006645.3
Adaptor/scaffold
Y59
RDSVKWHyLCLR
SEQ ID NO: 21





 22
Gab1
NP_002030.2
Adaptor/scaffold
Y47
SGRLTGDPDVLEyYK
SEQ ID NO: 22





 23
KPNA3
NP_002258.2
Adaptor/scaffold
Y506
ATQGGTyNFDPTANLQTKE
SEQ ID NO: 25





 24
KPNA4
NP_002259.1
Adaptor/scaffold
Y66
DSDIDGDyRVQNTSLE
SEQ ID NO: 26





 25
MACF1
NP_149033.2
Adaptor/scaffold
Y2230
GALDTTDGyMGVNQAPEKLDK
SEQ ID NO: 27





 26
P1300a
NP_055382.2
Adaptor/scaffold
Y653
FTSQDSPDGQyENSEGGWMEDYDYVH
SEQ ID NO: 29



s



LQGK





 27
PAR3-
NP_689739.4
Adaptor/scaffold
Y1034
GGPADPVDyLPAAPR
SEQ ID NO: 30



beta





 28
PEX14
NP_004556.1
Adaptor/scaffold
Y290
GSTVTyHLLGPQEE
SEQ ID NO: 31





 29
PSD-93
NP_001355.2
Adaptor/scaffold
Y223
GLGFSIAGGVGNQHIPGDNSIyVTK
SEQ ID NO: 32





 30
PSD-93
NP_001355.2
Adaptor/scaffold
Y750
FIEAGQyNDNLYGTSVQSVR
SEQ ID NO: 33





 31
PSD-95
NP_001356.1
Adaptor/scaffold
Y233
GLGFSIAGGVGNQHIPGDNSIyVTK
SEQ ID NO: 34





 32
PSD-95
NP_001356.1
Adaptor/scaffold
Y576
EDSVLSYETVTQMEVHyARPIIILGPTK
SEQ ID NO: 35





 33
SAPAP
NP_001073887.1
Adaptor/scaffold
Y823
EAEDyELPEEILEK
SEQ ID NO: 36



3





 34
SHANK
NP_057232.2
Adaptor/scaffold
Y186
FLEyVQLGTSDK
SEQ ID NO: 37



1





 35
SHANK
XP_037493.7
Adaptor/scaffold
Y122
FMDyVQLHSTDK
SEQ ID NO: 38



3





 36
Shc2
XP_944665.2
Adaptor/scaffold
Y412
GPPDHEEHLyVNTQGLDAPEPEDSPK
SEQ ID NO: 39





 37
SHD
NP_064594.2
Adaptor/scaffold
Y144
GVQLyDTPYEEQDPETADGPPSGQKPR
SEQ ID NO: 40





 38
SNCG
NP_003078.1
Adaptor/scaffold
Y39
EGVMyVGAK
SEQ ID NO: 41





 39
syntenin
NP_001007068.1
Adaptor/scaffold
Y50
SANPANPAILSEASAPIPHDGNLYPRLyP
SEQ ID NO: 42







ELSQY





 40
CDH1
NP_004351.1
Adhesion or
Y876
KLADMyGGGEDD
SEQ ID NO: 43





extracellular





matrix protein





 41
CDH1
NP_001788.2
Adhesion or
Y702
KDIKPEYQyMPR
SEQ ID NO: 44





extracellular





matrix protein





 42
CDH23
NP_071407.2
Adhesion or
Y1672
ITIQALDLDEGPNGTVTy
SEQ ID NO: 45





extracellular





matrix protein





 43
CGNL1
NP_116255.2
Adhesion or
Y108
ENSEELQLPENPyAQPSPIR
SEQ ID NO: 46





extracellular





matrix protein





 44
claudin
NP_057453.1
Adhesion or
Y254
TEDEVQSyPSKHDYV
SEQ ID NO: 47



18

extracellular





matrix protein





 45
CNTN6
NP_055276.1
Adhesion or
Y225
TDGVMGEyEPK
SEQ ID NO: 48





extracellular





matrix protein





 46
COL17A
NP_000485.3
Adhesion or
Y40
LTSLPPKGGTSNGyAK
SEQ ID NO: 49



1

extracellular





matrix protein





 47
DCBLD
NP_563615.3
Adhesion or
Y569
TEGTYDLPyWDR
SEQ ID NO: 50



2

extracellular





matrix protein





 48
DCBLD
NP_563615.3
Adhesion or
Y649
KPEEGKEAGyADLDPY
SEQ ID NO: 51



2

extracellular





matrix protein





 49
DSC2
NP_077740.1
Adhesion or
Y839
LGEKVyLCNQDENHK
SEQ ID NO: 52





extracellular





matrix protein





 50
Erbin
NP_061165.1
Adhesion or
Y425
VLTNyMFPQQPR
SEQ ID NO: 53





extracellular





matrix protein





 51
FN1
NP_002017.1
Adhesion or
Y2319
RPGGEPSPEGTTGQSyNQYSQR
SEQ ID NO: 54





extracellular





matrix protein





 52
FN1
NP_002017.1
Adhesion or
Y2322
RPGGEPSPEGTTGQSYNQySQR
SEQ ID NO: 55





extracellular





matrix protein





 53
FRAS1
NP_079350.4
Adhesion or
Y3985
NVNILSEPEAAyTFK
SEQ ID NO: 56





extracellular





matrix protein





 54
laminin
NP_001005472.1
Adhesion or
Y39
FLAAGTHLGGTNLDFQMEQYIyKR
SEQ ID NO: 57



receptor

extracellular



1

matrix protein





 55
LRRC7
NP_065845.1
Adhesion or
Y425
VLTNyMFPQQPR
SEQ ID NO: 58





extracellular





matrix protein





 56
occludin
NP_002529.1
Adhesion or
Y337
FyPESSYK
SEQ ID NO: 59





extracellular





matrix protein





 57
occludin
NP_002529.1
Adhesion or
Y342
FYPESSyK
SEQ ID NO: 60





extracellular





matrix protein





 58
PVRL3
NP_056295.1
Adhesion or
Y510
FERPMDyYEDLK
SEQ ID NO: 61





extracellular





matrix protein





 59
SEMA4
NP_004254.2
Adhesion or
Y450
EyDVLYLGTEDGHLHR
SEQ ID NO: 62



F

extracellular





matrix protein





 60
SEMA4
NP_004254.2
Adhesion or
Y454
EYDVLyLGTEDGHLHR
SEQ ID NO: 63



F

extracellular





matrix protein





 61
BNIP3L
NP_004322.1
Apoptosis
Y219
LSTPSASTy
SEQ ID NO: 64





 62
catalase
NP_001743.1
Apoptosis
Y379
LGPNYLHIPVNCPyR
SEQ ID NO: 65





 63
CYCS
NP_061820.1
Apoptosis
Y49
TGQAPGYSyTAANK
SEQ ID NO: 66





 64
FAIM3
NP_005440.1
Apoptosis
Y315
SQNNIySACPR
SEQ ID NO: 67





 65
CALB2
NP_001731.1
Calcium-binding
Y126
SGyIEANELK
SEQ ID NO: 68





protein





 66
CALB2
NP_001731.1
Calcium-binding
Y35
HFDADGNGyIEGK
SEQ ID NO: 69





protein





 67
CALCO
NP_005822.1
Calcium-binding
Y376
GGARQNPGLAyGNPYSGIQE
SEQ ID NO: 70



CO2

protein





 68
CALCO
NP_005822.1
Calcium-binding
Y380
GGARQNPGLAYGNPYSGIQE
SEQ ID NO: 71



C02

protein





 69
calse-
NP_001223.2
Calcium-binding
Y178
SEDSEyYKAFEEAAEHFQPYIK
SEQ ID NO: 72



questrin

protein



2





 70
FREQ
NP_055101.2
Calcium-binding
Y115
LYDLDNDGyITR
SEQ ID NO: 73





protein





 71
Cdc27
NP_001247.2
Cell cycle
Y740
ESLVyFLIGK
SEQ ID NO: 74





regulation





 72
CEP350
NP_055625.3
Cell cycle
Y2612
EKDVSEYFyEK
SEQ ID NO: 75





regulation





 73
CEP350
NP_055625.3
Cell cycle
Y337
KVATAPPAPAyK
SEQ ID NO: 76





regulation





 74
ch-TOG
NP_055571.2
Cell cycle
Y268
LEQQQSAGGDAEGGGDDGDEVPQIDAy
SEQ ID NO: 77





regulation

ELLEAVEILSK





 75
HCAP-
NP_071741.2
Cell cycle
Y929
GNKEFGDQAEAAQDATLTTTTFQNEDE
SEQ ID NO: 78



G

regulation

KNKEVyMTPLR





 76
SKB1
NP_006100.2
Cell cycle
Y283
EFCSYLQyLEYLSQNR
SEQ ID NO: 79





regulation






 77
CCT6B
NP_006575.2
Chaperone
Y239
SLEyEKTEVNSG
SEQ ID NO: 80





 78
CCT7
NP_001009570.1
Chaperone
Y59
VHTVEDyQAIVDAEWNILYDKLEK
SEQ ID NO: 81





 79
CCT-
NP_006421.2
Chaperone
Y269
TDMDNQIVVSDyAQMDR
SEQ ID NO: 82



delta





 80
DNAJB5
NP_036398.3
Chaperone
Y52
EIAEAyDVLSDPK
SEQ ID NO: 83





 81
DNAJB6
NP_490647.1
Chaperone
Y53
QVAEAyEVLSDAK
SEQ ID NO: 84





 82
ARID1A
NP_006006.3
Chromatin,
Y762
NPQMPQySSPQPGSALSPR
SEQ ID NO: 85





DNA-binding,





DNA repair or





DNA replication





protein





 83
C14orf4
NP_919254.2
Chromatin,
Y158
GSPHPGVGVPTyYNHPEALKR
SEQ ID NO: 86



3

DNA-binding,





DNA repair or





DNA replication





protein





 84
CHD1L
NP_004275.2
Chromatin,
Y709
LDyQDPDATSLKYVSGDVTHPQAGAE
SEQ ID NO: 87





DNA-binding,





DNA repair or





DNA replication





protein





 85
CHD-6
NP_115597.3
Chromatin,
Y1640
LyESLTYSQMSR
SEQ ID NO: 88





DNA-binding,





DNA repair or





DNA replication





protein





 86
CHD-6
NP_115597.3
Chromatin,
Y1645
LYESLTySQMSR
SEQ ID NO: 89





DNA-binding,





DNA repair or





DNA replication





protein





 87
H2BK
NP_542160.1
Chromatin,
Y41
ESYSVyVYK
SEQ ID NO: 90





DNA-binding,





DNA repair or





DNA replication





protein





 88
H2BL
NP_003510.1
Chromatin,
Y41
ESYSVyVYK
SEQ ID NO: 91





DNA-binding,





DNA repair or





DNA replication





protein





 89
HIST2H
NP_001019770.1
Chromatin,
Y41
ESYSVyVYK
SEQ ID NO: 92



2BF

DNA-binding,





DNA repair or





DNA replication





protein





 90
HMGB1
NP_002119.1
Chromatin,
Y78
EMKTyIPPKGETKK
SEQ ID NO: 93





DNA-binding,





DNA repair or





DNA replication





protein





 91
NAP1L1
NP_004528.1
Chromatin,
Y377
GDEENDPDyDPKKDQNPAE
SEQ ID NO: 94





DNA-binding,





DNA repair or





DNA replication





protein





 92
ARVCF
NP_001661.1
Cytoskeletal
Y201
DSPSyGSLSR
SEQ ID NO: 95





protein





 93
calponin
NP_001830.1
Cytoskeletal
Y316
HGEYQDDyPRDY
SEQ ID NO: 96



3

protein





 94
CAPZA
NP_006127.1
Cytoskeletal
Y198
IQVHyYEDGNVQLVSHK
SEQ ID NO: 97



2

protein





 95
CCDC6
NP_005427.2
Cytoskeletal
Y336
MDDERyFNE
SEQ ID NO: 98





protein





 96
CDK5R
NP_060719.4
Cytoskeletal
Y1111
VSVMGTDQSESINTSNETEyLKQK
SEQ ID NO: 99



AP2

protein





 97
CLASP2
NP_055912.1
Cytoskeletal
Y1150
NMNSEDIySSLRGVTE
SEQ ID NO: 100





protein





 98
claudin
NP_003268.1
Cytoskeletal
Y212
RPTATGDyDKKNYV
SEQ ID NO: 101



5

protein





 99
claudin
NP_003268.1
Cytoskeletal
Y217
RPTATGDYDKKNyV
SEQ ID NO: 102



5

protein





100
cofilin
NP005498.1
Cytoskeletal
Y82
MLPDKDCRyALYDATYETKESK
SEQ ID NO: 103



1

protein





101
cofilin
NP_068733.1
Cytoskeletal
Y82
LLPLNDCRyALYDATYETK
SEQ ID NO: 104



2

protein





102
cordon-
NP_056013.2
Cytoskeletal
Y1143
LSyTEAEGER
SEQ ID NO: 105



bleu

protein





103
cordon-
NP_056013.2
Cytoskeletal
Y652
VKDKVyGCADGER
SEQ ID NO: 106



bleu

protein





104
cortactin
NP_005222.2
Cytoskeletal
Y538
yGLFPANYVELRQ
SEQ ID NO: 107





protein





105
COTL1
NP_066972.1
Cytoskeletal
Y137
LKKAGGANyDAQTE
SEQ ID NO: 108





protein





106
CTNND
NP_001323.1
Cytoskeletal
Y1090
KTDyECTGSNATYHGAK
SEQ ID NO: 109



2

protein





107
DBN1
NP_004386.2
Cytoskeletal
Y32
SAADWALyTYEDGSDDLKLAASGE
SEQ ID NO: 110





protein





108
dystro-
NP_004014.1
Cytoskeletal
Y894
MHyPMVEYCTPTTSGEDVR
SEQ ID NO: 111



phin

protein





109
EB1
NP_036457.1
Cytoskeletal
Y268
GFVIPDEGGPQEEQEEy
SEQ ID NO: 112





protein





110
EHM2
NP_061987.3
Cytoskeletal
Y447
TNPEVHNyQPQYHPNIHPSQPR
SEQ ID NO: 113





protein





111
EHM2
NP_061987.3
Cytoskeletal
Y451
TNPEVHNYQPQyHPNIHPSQPR
SEQ ID NO: 114





protein





112
EML4
NP_061936.2
Cytoskeletal
Y265
LKLEWAyGYR
SEQ ID NO: 115





protein





113
EML4
NP_061936.2
Cytoskeletal
Y954
GSGDLGEPLyEEPCNE
SEQ ID NO: 116





protein





114
eplin
NP_057441.1
Cytoskeletal
Y429
LSLGTyASLHGR
SEQ ID NO: 117





protein





115
EXOC7
NP_001013861.1
Cytoskeletal
Y90
TLSCLDHVISyYHVASDTEK
SEQ ID NO: 118





protein





116
ezrin
NP_003370.2
Cytoskeletal
Y499
SLQDEGAEPTGySAE
SEQ ID NO: 119





protein





117
FLNB
NP_001448.2
Cytoskeletal
Y596
IEyNDQNDGSCDVK
SEQ ID NO: 120





protein





118
FLNC
NP_001449.3
Cytoskeletal
Y1303
VLNPSGAKTDTyVTDNGDGTYR
SEQ ID NO: 121





protein





119
FLNC
NP_001449.3
Cytoskeletal
Y1312
VLNPSGAKTDTYVTDNGDGTyR
SEQ ID NO: 122





protein





120
FNBP1
NP_055848.1
Cytoskeletal
Y234
MGESMKTyAEVDR
SEQ ID NO: 123





protein





121
FNBP1
NP_055848.1
Cytoskeletal
Y287
SGFEPPGDIEFEDyTQPMKR
SEQ ID NO: 124





protein





122
INA
NP_116116.1
Cytoskeletal
Y425
FSTSGLSISGLNPLPNPSyLLPPR
SEQ ID NO: 125





protein





123
K1
NP_006112.3
Cytoskeletal
Y566
GSYGSGGSSYGSGGGSyGSGGGGGGH
SEQ ID NO: 126





protein

GSYGSGSSSGGYR





124
RP1
NP_055083.1
Cytoskeletal
Y162
FYDANyDGKEYDPVEAR
SEQ ID NO: 127





protein





125
SAPAP
NP_004737.2
Cytoskeletal
Y317
SCQyLQVPQDEWTGYTPR
SEQ ID NO: 128



1

protein





126
SHRM
NP_065910.2
Cytoskeletal
Y500
ESGyIAPQGACNK
SEQ ID NO: 129





protein





127
talin 2
NP_055874.1
Cytoskeletal
Y1854
GTFVDyQTTVVK
SEQ ID NO: 130





protein





128
talin 2
NP_055874.1
Cytoskeletal
Y49
ERVPEAQTGQASDyGLFLSDEDPR
SEQ ID NO: 131





protein





129
OSBPL
NP_056365.1
Endoplasmic
Y47
GEMNyTQEPPVQK
SEQ ID NO: 132



3

reticulum or





golgi





130
CA9
NP_001207.1
Enzyme, misc.
Y449
GGVSyRPAEVAETGA
SEQ ID NO: 133





131
CCBL1
NP_004050.3
Enzyme, misc.
Y15
MAKQLQARRLDGIDyNPWVEFVK
SEQ ID NO: 134





132
CHD2
NP_001262.3
Enzyme, misc.
Y596
LKFNALITTyEILLKDK
SEQ ID NO: 135





133
CNP
NP_149124.3
Enzyme, misc.
Y373
LySLGNGR
SEQ ID NO: 136





134
CPT1A
NP_001867.2
Enzyme, misc.
Y514
GDINPNIPyPTR
SEQ ID NO: 138





135
CTPS
NP_001896.1
Enzyme, misc.
Y96
IYQyVINK
SEQ ID NO: 139





136
DDHD1
NP_085140.1
Enzyme, misc.
Y89
GEPGLHLAPGTDDHNHHLALDPCLSDE
SEQ ID NO: 140







NyDFSSAESGSSLR





137
DDX10
NP_004389.2
Enzyme, misc.
Y273
LSLKNPEyVWVHEK
SEQ ID NO: 141





138
DDX42
NP_031398.2
Enzyme, misc.
Y160
GIRDDIEEEDDQEAyFR
SEQ ID NO: 142





139
DDX9
NP_001348.2
Enzyme, misc.
Y21
KMTPSyEIR
SEQ ID NO: 143





140
DEGS1
NP_003667.1
Enzyme, misc.
Y14
VSREDFEWVyTDQPHADR
SEQ ID NO: 144





141
DHCR7
NP_001351.2
Enzyme, misc.
Y382
VIECSyTSADGQR
SEQ ID NO: 147





142
DHX33
NP_064547.2
Enzyme, misc.
Y380
KyNPDSGLEVLAVQR
SEQ ID NO: 148





143
DHX36
NP_065916.1
Enzyme, misc.
Y1006
NFPPRFQDGyYS
SEQ ID NO: 149





144
Diminuto
NP_055577.1
Enzyme, misc.
Y299
LNSIGNyYK
SEQ ID NO: 150





145
Diminuto
NP_055577.1
Enzyme, misc.
Y300
LNSIGNYyK
SEQ ID NO: 151





146
DUS3L
NP_064560.1
Enzyme, misc.
Y308
LyLAPLTTCGNLPFR
SEQ ID NO: 152





147
EPRS
NP_004437.2
Enzyme, misc.
Y684
RGFFICDQPyEPVSPYSCK
SEQ ID NO: 153





148
FBPase
NP_000498.2
Enzyme, misc.
Y259
TLVyGGIFLYPANK
SEQ ID NO: 154





149
G6PI
NP_000166.2
Enzyme, misc.
Y92
MFNGEKINyTEGR
SEQ ID NO: 155





150
IMPDH2
NP_000875.2
Enzyme, misc.
Y459
FVPyLIAGIQHSCQDIGAK
SEQ ID NO: 156





151
LDH-B
NP_002291.1
Enzyme, misc.
Y173
FRyLMAEK
SEQ ID NO: 157





152
LSD1
NP_055828.2
Enzyme, misc.
Y135
EMDESLANLSEDEyYSEEER
SEQ ID NO: 158





153
LSD1
NP_055828.2
Enzyme, misc.
Y136
EMDESLANLSEDEYySEEER
SEQ ID NO: 159





154
ACSM2
NP_001010845.1
Enzyme, misc.
Y303
FDPLVILKTLSSyPIK
SEQ ID NO: 160



A





155
NTE
NP_006693.3
Enzyme, misc.
Y431
EQPAGACEySYCEDESATGGCPFGPYQ
SEQ ID NO: 161







GR





156
PLD1
NP_002653.1
Enzyme, misc.
Y42
ELHFEGEEVDyDVSPSDPK
SEQ ID NO: 162





157
SCLY
NP_057594.2
Enzyme, misc.
Y280
GLGEFTPLyPMLFGGGQER
SEQ ID NO: 163





158
Cdc42
NP_001782.1
G protein or
Y32
TTNKFPSEyVPTVF
SEQ ID NO: 164





regulator





159
Cdc42E
NP_689449.1
G protein or
Y130
NAISLPQLNQAAyDSLVVGK
SEQ ID NO: 165



P1

regulator





160
Cdc42E
NP_689449.1
G protein or
Y331
HWGAGWDGGHHyPEMDAR
SEQ ID NO: 166



P1

regulator





161
CENTD
NP_056045.2
G protein or
Y867
KAGQSLQMEFLyHNK
SEQ ID NO: 167



1

regulator





162
CENTD
NP_056057.1
G protein or
Y119
yFDSNKDAYSK
SEQ ID NO: 168



2

regulator





163
CENTD
NP_056057.1
G protein or
Y127
YFDSNKDAySK
SEQ ID NO: 169



2

regulator





164
ARHGA
NP_004058.1
G protein or
Y153
YISKMTTNPIyE
SEQ ID NO: 170



P3

regulator





165
ARHGA
NP_004058.1
G protein or
Y158
MTTNPIYEHIGyATLLR
SEQ ID NO: 171



P3

regulator





166
DDEF2
NP_003878.1
G protein or
Y724
EDRPISFyQLGSNQLQSNAVSLAR
SEQ ID NO: 172





regulator





167
DOCK1
NP_055504.1
G protein or
Y1126
EDQLEyQEELR
SEQ ID NO: 173



0

regulator





168
DOCK9
NP_056111.1
G protein or
Y340
LFyLDPDAQK
SEQ ID NO: 174





regulator





169
ephexin
NP_062824.1
G protein or
Y288
LVTSEASyYKSLNLLVSHFMENE
SEQ ID NO: 175



1

regulator





170
ephexin
NP_062824.1
G protein or
Y289
LVTSEASYyKSLNLLVSHFMENE
SEQ ID NO: 176



1

regulator





171
ephexin
NP_062824.1
G protein or
Y537
QIPGDKyQVFDSAPR
SEQ ID NO: 177



1

regulator





172
EPS8L2
NP_073609.2
G protein or
Y678
VySQLTMQK
SEQ ID NO: 178





regulator





173
EVI5L
NP_660288.1
G protein or
Y788
LAAPySQGLDN
SEQ ID NO: 179





regulator





174
FGD5
NP_689749.2
G protein or
Y603
QQSADQDAESAyTEPYK
SEQ ID NO: 180





regulator





175
G-
NP_002064.1
G protein or
Y75
EYKPLIIyNAIDSLTR
SEQ ID NO: 181



alpha(z)

regulator





176
G-
NP_002061.1
G protein or
Y155
EYQLNDSMyYLNDLER
SEQ ID NO: 182



alpha2(i)

regulator





177
GPSM3
NP_071390.1
G protein or
Y108
EQLySTILSHQCQR
SEQ ID NO: 183





regulator





178
IQGAP1
NP_003861.1
G protein or
Y1284
FNVDEySDLVTLTK
SEQ ID NO: 184





regulator





179
IQGAP1
NP_003861.1
G protein or
Y694
GGyYYYHNLETQEGGWDEPPNFVQNS
SEQ ID NO: 185





regulator

MQLSR





180
IQGAP1
NP_003861.1
G protein or
Y695
GGYyYYHNLETQEGGWDEPPNFVQNS
SEQ ID NO: 186





regulator

MQLSR





181
IQGAP1
NP_003861.1
G protein or
Y697
GGYYYyHNLETQEGGWDEPPNFVQNS
SEQ ID NO: 187





regulator

MQLSR





182
IQGAP2
AAB37765.1
G protein or
Y630
ESSWVTPESCFyKESWLTGK
SEQ ID NO: 188





regulator





183
IQGAP3
NP_839943.2
G protein or
Y1268
FAVDEySDMVAVAKPMVYITVGELVNTH
SEQ ID NO: 189





regulator

R





184
RICS
NP_055530.2
G protein or
Y1424
GPVMSQyDNMTPAVQDDLGGIYVIHLR
SEQ ID NO: 190





regulator





185
RRAS2
NP_036382.2
G protein or
Y105
GSFEEIyKFQR
SEQ ID NO: 191





regulator





186
SIPA1L
NP_055888.1
G protein or
Y1265
GEPQySSHSSSNTLSSNASSSHSDDR
SEQ ID NO: 192



3

regulator





187
SIPA1L
NP_055888.1
G protein or
Y1316
GGSSDSGIDTTLyTSSPSCMSLAK
SEQ ID NO: 193



3

regulator





188
SRGAP
NP_056141.2
G protein or
Y699
GPVYSRGGSMEDyCDSPHGE
SEQ ID NO: 194



2

regulator





189
ITIH1
NP_002206.1
Inhibitor
Y431
FPLyNLGFGHNVDFNFLEVMSMENNGR
SEQ ID NO: 195





protein





190
EM55
NP_002427.1
Kinase (non-
Y316
FVYPVPyTTRPPRKSEEDGK
SEQ ID NO: 196





protein)





191
PIK3CA
NP_006209.2
Kinase (non-
Y294
ESLySQLPMDCFTMPSYSR
SEQ ID NO: 197





protein)





192
PIK3R1
NP_852664.1
Kinase (non-
Y76
GDFPGTYVEyIGR
SEQ ID NO: 198





protein)





193
PIK3R2
NP_005018.1
Kinase (non-
Y460
SREyDQLYEEYTR
SEQ ID NO: 199





protein)





194
PIP5K
NP_055855.2
Kinase (non-
Y154
GKSQDSDLKQyWMPDSQCKE
SEQ ID NO: 200





protein)





195
PIP5KG
NP_036530.1
Kinase (non-
Y354
ALySTAMESIQGGAAR
SEQ ID NO: 201





protein)





196
ephrin-
NP_004420.1
Ligand, receptor
Y79
PYEYYKLyLVR
SEQ ID NO: 202



B1

tyrosine kinase





197
FRMD6
NP_689543.1
Lipid binding
Y262
QLLyDFPWTNVGK
SEQ ID NO: 203





protein





198
PLEKH
NP_001001974.1
Lipid binding
Y345
GFyESLAK
SEQ ID NO: 204



A1

protein





199
DCTN3
NP_009165.1
Motor or
Y67
KIEDLIKyLDPEYIDR
SEQ ID NO: 206





contractile





protein





200
DNCH1
NP_001367.2
Motor or
Y970
ITNQVIyLNPPIEECR
SEQ ID NO: 207





contractile





protein





201
KIF2A
NP_004511.1
Motor or
Y168
KRAQDVDATNPNyE
SEQ ID NO: 208





contractile





protein





202
KNSL8
NP_958929.1
Motor or
Y205
GQGATAAQQGGyEIPAR
SEQ ID NO: 209





contractile





protein





203
MRLC2
NP_000423.2
Motor or
Y118
GVLKADyVR
SEQ ID NO: 210



V

contractile





protein





204
MYH1
NP_005954.3
Motor or
Y1856
KVKELTyQTEEDRK
SEQ ID NO: 211





contractile





protein





205
MYH1
NP_005954.3
Motor or
Y424
GQTVQQVyNAVGALAK
SEQ ID NO: 212





contractile





protein





206
MYH2
NP_060004.2
Motor or
Y1858
ELTyQTEEDRKNILR
SEQ ID NO: 213





contractile





protein





207
MYH2
NP_060004.2
Motor or
Y822
EAIFCIQyNIR
SEQ ID NO: 214





contractile





protein





208
MYH4
NP_060003.2
Motor or
Y1856
ELTyQTEEDRK
SEQ ID NO: 215





contractile





protein





209
MYH4
NP_060003.2
Motor or
Y424
GQTVQQVyNAVGALAK
SEQ ID NO: 216





contractile





protein





210
MYH7
NP_000248.2
Motor or
Y422
GQNVQQVIyATGALAK
SEQ ID NO: 217





contractile





protein





211
MYH8
NP_002463.1
Motor or
Y424
GQTVQQVyNAVGALAK
SEQ ID NO: 218





contractile





protein





212
MYO10
NP_036466.1
Motor or
Y585
FDFIyDLFEHVSSR
SEQ ID NO: 219





contractile





protein





213
MYO1C
NP_203693.3
Motor or
Y438
SEQEEYEAEGIAWEPVQyFNNK
SEQ ID NO: 220





contractile





protein





214
MYO1F
NP_036467.2
Motor or
Y438
WTPIQyFNNK
SEQ ID NO: 221





contractile





protein





215
CD45
NP_002829.2
Phosphatase
Y681
NRyVDILPYDYNR
SEQ ID NO: 222





216
DARPP-
NP_115568.2
Phosphatase
Y116
ELGyPREEDEEEEEDDEEEEEEEDSQAE
SEQ ID NO: 223



32



VLK





217
DUSP3
NP_004081.1
Phosphatase
Y23
LSVQDLNDLLSDGSGCySLPSQPCNE
SEQ ID NO: 224





218
FBP2
NP_003828.2
Phosphatase
Y216
IYSLNEGyAK
SEQ ID NO: 225





219
FBP2
NP_003828.2
Phosphatase
Y259
TLVyGGIFLYPANQK
SEQ ID NO: 226





220
INPP5F
NP_055752.1
Phosphatase
Y430
FENVQTLTDAIyDIILDMK
SEQ ID NO: 227





221
PTPN14
NP_005392.2
Phosphatase
Y496
ERHPyTVPYGPQGVYSNK
SEQ ID NO: 228





222
CMA1
NP_001827.1
Protease
Y35
PyMAYLEIVTSNGPSK
SEQ ID NO: 229





223
CMA1
NP_001827.1
Protease
Y38
PYMAyLEIVTSNGPSK
SEQ ID NO: 230





224
CNDP2
NP_060705.1
Protease
Y311
WRyPSLSLHGIEGAFSGSGAK
SEQ ID NO: 231





225
PSMB4
NP_002787.2
Protease
Y75
FEGGVVIAADMLGSyGSLAR
SEQ ID NO: 232





226
DYRK4
NP_003836.1
Protein kinase,
Y356
yPDSKDLTMVLK
SEQ ID NO: 234





dual-speciticity





227
CKS1
NP_001817.1
Protein kinase,
Y12
QIYYSDKyDDEEFEYR
SEQ ID NO: 235





regulatory





subunit





228
CaMK2-
NP_001212.2
Protein kinase,
Y14
FTDEyQLFEELGK
SEQ ID NO: 236



delta

Ser/Thr (non-





receptor)





229
CAMKK
NP_006540.3
Protein kinase,
Y190
LAYNENDNTyYAMK
SEQ ID NO: 237



2

Ser/Thr (non-





receptor)





230
Cdc2
NP_001777.1
Protein kinase,
Y4
MEDyTKIEKIGEGTYGVVYK
SEQ ID NO: 238





Ser/Thr (non-





receptor)





231
CDK9
NP_001252.1
Protein kinase,
Y287
LKAYVRDPyALDLIDKLLVLDPAQR
SEQ ID NO: 239





Ser/Thr (non-





receptor)





232
CK2-
NP_001886.1
Protein kinase,
Y50
GKySEVFEAINITNNEK
SEQ ID NO: 240



alpha1

Ser/Thr (non-





receptor)





233
CRIK
NP_009105.1
Protein kinase,
Y1759
QyTLEEFLDK
SEQ ID NO: 241





Ser/Thr (non-





receptor)





234
Nek1
NP_036356.1
Protein kinase,
Y443
GQyEHYHAIFDQMQQQR
SEQ ID NO: 242





Ser/Thr (non-





receptor)





235
Nek1
NP_036356.1
Protein kinase,
Y446
GQYEHyHAIFDQMQQQR
SEQ ID NO: 243





Ser/Thr (non-





receptor)





236
PLK2
NP_006613.2
Protein kinase,
Y297
EARyTMPSSLLAPAKHLIASMLSK
SEQ ID NO: 244





Ser/Thr (non-





receptor)





237
ROCK1
NP_005397.1
Protein kinase,
Y913
GLLEEQYFELTQESK
SEQ ID NO: 245





Ser/Thr (non-





receptor)





238
RSK2
NP_004577.1
Protein kinase,
Y433
EDIGVGSySVCK
SEQ ID NO: 246





Ser/Thr (non-





receptor)





239
RSK2
NP_004577.1
Protein kinase,
Y644
FSLSGGyWNSVSDTAK
SEQ ID NO: 247





Ser/Thr (non-





receptor)





240
RSK3
NP_001006933.1
Protein kinase,
Y434
EDIGVGSySVCK
SEQ ID NO: 248





Ser/Thr (non-





receptor)





241
RSK3
NP_001006933.1
Protein kinase,
Y707
GAMAATYFALNR
SEQ ID NO: 249





Ser/Thr (non-





receptor)





242
RSK4
NP_055311.1
Protein kinase,
Y437
EDIGVGSySVCK
SEQ ID NO: 250





Ser/Thr (non-





receptor)





243
SRPK1
NP_003128.3
Protein kinase,
Y62
GSAPHSESDLPEQEEEILGSDDDEQEDP
SEQ ID NO: 251





Ser/Thr (non-

NDyCK





receptor)





244
FRK
NP_002022.1
Protein kinase,
Y221
IQVPAPFDLSyK
SEQ ID NO: 252





Tyr (non-





receptor)





245
FRK
NP_002022.1
Protein kinase,
Y317
HGSLQEyLQNDTGSK
SEQ ID NO: 253





Tyr (non-





receptor)





246
FRK
NP_002022.1
Protein kinase,
Y368
NVLVGEHNIyKVADFGLAR
SEQ ID NO: 254





Tyr (non-





receptor)





247
Lyn
NP_002341.1
Protein kinase,
Y321
EEPIYIITEyMAK
SEQ ID NO: 255





Tyr (non-





receptor)





248
EphA2
NP_004422.2
Protein kinase,
Y735
YLANMNyVHR
SEQ ID NO: 256





Tyr (receptor)





249
EphA3
NP_005224.2
Protein kinase,
Y937
EIFTGVEySSCDTIAK
SEQ ID NO: 257





Tyr (receptor)





250
EphA4
NP_004429.1
Protein kinase,
Y798
WTAPEAIAyR
SEQ ID NO: 259





Tyr (receptor)





251
EphA6
XP_914973.6
Protein kinase,
Y831
VLEDDPEAAyTTTGGK
SEQ ID NO: 260





Tyr (receptor)





252
EphA6
NP_001073917.2
Protein kinase,
Y934
WTAPEAIAyR
SEQ ID NO: 261





Tyr (receptor)





253
EphB1
NP_004432.1
Protein kinase,
Y575
EAVySDKLQHYSTGR
SEQ ID NO: 262





Tyr (receptor)





254
EphB1
NP_004432.1
Protein kinase,
Y582
EAVYSDKLQHySTGR
SEQ ID NO: 263





Tyr (receptor)





255
EphB1
NP_004432 1
Protein kinase,
Y798
WTAPEAIAyR
SEQ ID NO: 264





Tyr (receptor)





256
EphB3
NP_004434.2
Protein kinase,
Y812
WTAPEAIAyR
SEQ ID NO: 265





Tyr (receptor)





257
FGFR1
NP_075594.1
Protein kinase,
Y522
GMEyLASKK
SEQ ID NO: 266





Tyr (receptor)





258
FGFR2
NP_000132.1
Protein kinase,
Y466
LSSTADTPMLAGVSEyELPEDPKWEFPR
SEQ ID NO: 267





Tyr (receptor)





259
FGFR2
NP_000132.1
Protein kinase,
Y586
RPPGMEySYDINR
SEQ ID NO: 268





Tyr (receptor)





260
FGFR2
NP_000132.1
Protein kinase,
Y588
RPPGMEYSyDINR
SEQ ID NO: 269





Tyr (receptor)





261
FGFR2
NP_000132.1
Protein kinase,
Y616
GMEyLASQK
SEQ ID NO: 270





Tyr (receptor)





262
FGFR2
NP_000132.1
Protein kinase,
Y733
MDKPANCTNELyMMMR
SEQ ID NO: 271





Tyr (receptor)





263
FGFR2
NP_000132.1
Protein kinase,
Y805
SSCSSGDDSVFSPDPMPyEPCLPQYPHI
SEQ ID NO: 272





Tyr (receptor)

NGSVK





264
FGFR2
NP_000132.1
Protein kinase,
Y812
SSCSSGDDSVFSPDPMPYEPCLPQyPHI
SEQ ID NO: 273





Tyr (receptor)

NGSVK





265
FGFR3
NP_000133.1
Protein kinase,
Y607
GMEyLASQK
SEQ ID NO: 274





Tyr (receptor)





266
FLT3
NP_004110.2
Protein kinase,
Y865
WMAPESLFEGIyTIK
SEQ ID NO: 275





Tyr (receptor)





267
Lmr2
NP_055731.2
Protein kinase,
Y500
GHLDEGLSyTSIFYPVEVFESSLSDPGPG
SEQ ID NO: 276





Tyr (receptor)

K





268
Met
NP_000236.2
Protein kinase,
Y1194
GMKyLASKK
SEQ ID NO: 277





Tyr (receptor)





269
CD229
NP_002339.2
Receptor,
Y583
GAGHDPAPEGQADyDPVTPYVTE
SEQ ID NO: 279





channel,





transporter or





cell surface





protein





270
CD229
NP_002339.2
Receptor,
Y589
GAGHDPAPEGQADYDPVTPyVTE
SEQ ID NO: 280





channel,





transporter or





cell surface





protein





271
CD82
NP_002222.1
Receptor,
Y261
HVHSEDySKVPKY
SEQ ID NO: 281





channel,





transporter or





cell surface





protein





272
CD82
NP_002222.1
Receptor,
Y267
HVHSEDYSKVPKy
SEQ ID NO: 282





channel,





transporter or





cell surface





protein





273
CLCC1
NP_001041675.1
Receptor,
Y412
GQMGPTEQGPyAK
SEQ ID NO: 283





channel,





transporter or





cell surface





protein





274
CLCC1
NP_001041675.1
Receptor,
Y531
SEAAGSPDQGSTySPAR
SEQ ID NO: 284





channel,





transporter or





cell surface





protein





275
CR2
NP_001006659.1
Receptor,
Y1083
EVySVDPYNPAS
SEQ ID NO: 285





channel,





transporter or





cell surface





protein





276
Cx43
NP_000156.1
Receptor,
Y301
NyNKQASEQNWANYSAEQNR
SEQ ID NO: 286





channel,





transporter or





cell surface





protein





277
DNAJC
NP_071760.2
Receptor,
Y295
TTyIQSYDHGTSIEE
SEQ ID NO: 287



1

channel,





transporter or





cell surface





protein





278
DNAJC
NP_071760.2
Receptor,
Y299
TTYIQSyDHGTSIEE
SEQ ID NO: 288



1

channel,





transporter or





cell surface





protein





279
DNER
NP_620711.3
Receptor,
Y677
PAyEEFYNCR
SEQ ID NO: 289





channel,





transporter or





cell surface





protein





280
DNER
NP_620711.3
Receptor,
Y681
PAYEEFyNCR
SEQ ID NO: 290





channel,





transporter or





cell surface





protein





281
DNER
NP_620711.3
Receptor,
Y711
SRPAMyDVSPIAYEDYSPDDKPLVTLIK
SEQ ID NO: 291





channel,





transporter or





cell surface





protein





282
DNER
NP_620711.3
Receptor,
Y718
SRPAMYDVSPIAyEDYSPDDKPLVTLIK
SEQ ID NO: 292





channel,





transporter or





cell surface





protein





283
DNER
NP_620711.3
Receptor,
Y721
SRPAMYDVSPIAYEDySPDDKPLVTLIK
SEQ ID NO: 293





channel,





transporter or





cell surface





protein





284
DYSF
NP_003485.1
Receptor,
Y1650
ITLYDyDLLSKDEK
SEQ ID NO: 294





channel,





transporter or





cell surface





protein





285
EDG4
NP_004711.2
Receptor,
Y325
ESVHyTSSAQGGASTR
SEQ ID NO: 295





channel,





transporter or





cell surface





protein





286
Emelin
NP_036451.2
Receptor,
Y487
YESQLSTNEEKVDTDDRTEGyLR
SEQ ID NO: 296





channel,





transporter or





cell surface





protein





287
FZD5
NP_003459.2
Receptor,
Y556
SGGAMAAGDyPEASAALTGR
SEQ ID NO: 297





channel,





transporter or





cell surface





protein





288
FZD5
NP_003459.2
Receptor,
Y576
TGPPGPAATyHK
SEQ ID NO: 298





channel,





transporter or





cell surface





protein





289
GPA33
NP_005805.1
Receptor,
Y301
EREEEDDyRQEEQR
SEQ ID NO: 299





channel,





transporter or





cell surface





protein





290
GPIP13
NP_005889.3
Receptor,
Y78
GKLDDyQER
SEQ ID NO: 300



7

channel,





transporter or





cell surface





protein





291
HMMR
NP_036617.1
Receptor,
Y698
GNTNCyRAPMECQE
SEQ ID NO: 301





channel,





transporter or





cell surface





protein





292
IL2RG
NP_000197.1
Receptor,
Y363
GGALGEGPGASPCNQHSPYWAPPCyTL
SEQ ID NO. 303





channel,

KPET





transporter or





cell surface





protein





293
LRP4
NP_002325.1
Receptor,
Y1764
FTDPGMGNLTySNPSYR
SEQ ID NO: 304





channel,





transporter or





cell surface





protein





294
NKCC1
NP_001037.1
Receptor,
Y1211
GNHQSVLTFyS
SEQ ID NO: 305





channel,





transporter or





cell surface





protein





295
OR4C4
NP_001004703.1
Receptor,
Y273
AVAIFyTMITPMLNPLIYTLKNAQMK
SEQ ID NO: 306



6

channel,





transporter or





cell surface





protein





296
OR4C4
NP_001004703.1
Receptor,
Y285
AVAIFYTMITPMLNPLIyTLKNAQMK
SEQ ID NO: 307



6

channel,





transporter or





cell surface





protein





297
ORAI1
NP_16179.2
Receptor,
Y300
GDHPLTPGSHyA
SEQ ID NO: 308





channel,





transporter or





cell surface





protein





298
PTDSS
NP_110410.1
Receptor,
Y366
EIyDFMDDPK
SEQ ID NO: 309



2

channel,





transporter or





cell surface





protein





299
SLC11A
NP_000608.1
Receptor,
Y31
GNINPAySNPSLSQSPGDSEEY
SEQ ID NO: 310



2

channel,





transporter or





cell surface





protein





300
SLC4A2
NP_003031.2
Receptor,
Y1234
EGVDEyNEMPMPV
SEQ ID NO: 311





channel,





transporter or





cell surface





protein





301
SLC4A2
NP_003031.2
Receptor,
Y66
GGEEPGRSyGEEDFEYHR
SEQ ID NO: 312





channel,





transporter or





cell surface





protein





302
SLC4A4
NP_003750.1
Receptor,
Y994
GSLDSDNDDSDCPySEK
SEQ ID NO: 313





channel,





transporter or





cell surface





protein





303
SLC5A3
NP_008864.3
Receptor,
Y631
EEGNPVASLGHSEAETPVDAySNGQAAL
SEQ ID NO: 314





channel,

MGEK





transporter or





cell surface





protein





304
SLITRK
NP_055741.2
Receptor,
Y904
ERPQPAPCTVGFVDCLyGTVPK
SEQ ID NO: 315



3

channel,





transporter or





cell surface





protein





305
CIRBP
NP_001271.1
RNA processing
Y135
SGGyGGSRDYYSSR
SEQ ID NO: 316





306
CPSF1
NP_037423.2
RNA processing
Y748
SGPEAEGLGSETSPTVDDEEEMLyGDS
SEQ ID NO: 317







GSLFSPSKEEAR


307
DBR1
NP_057300.2
RNA processing
Y533
NQAIyAAVDDDDDDAA
SEQ ID NO: 318





308
DCP2
NP_689837.2
RNA processing
Y370
TDAVyDLPSSSE
SEQ ID NO: 319





309
DDX1
NP_004930.1
RNA processing
Y628
VWyHVCSSR
SEQ ID NO: 320





310
DDX1
NP_004930.1
RNA processing
Y731
EAQTSFLHLGyLPNQLFR
SEQ ID NO: 321





311
DDX3
NP_001347.3
RNA processing
Y163
LFSGGNTGINFEKyDDIPVEATGNNCPP
SEQ ID NO: 322







HIE





312
DDX3
NP_001347.3
RNA processing
Y283
ELAVQIyEEAR
SEQ ID NO: 323





313
DDX3
NP_001347.3
RNA processing
Y466
KGADSLEDFLYHEGyACTSIHGDR
SEQ ID NO: 324





314
DDX39
NP_005795.2
RNA processing
Y13
NDLLDyDEEEEPQAPQE
SEQ ID NO: 325





315
DDX3Y
NP_004651.2
RNA processing
Y241
TAAFLLPILSQIyTDGPGEALK
SEQ ID NO: 326





316
DDX3Y
NP_004651.2
RNA processing
Y281
ELAVQIYEEAR
SEQ ID NO: 327





317
DDX3Y
NP_004651.2
RNA processing
Y464
KGADSLEDFLYHEGyACTSIHGDR
SEQ ID NO: 328





318
DDX5
NP_004387.1
RNA processing
Y442
TGTAyTFFTPNNIK
SEQ ID NO: 329





319
HNRPU
NP_001073027.1
RNA processing
Y660
SRGQGyVGGQR
SEQ ID NO: 330



L2





320
HNRPU
NP_001073027.1
RNA processing
Y743
NYYGyQGYR
SEQ ID NO: 331



L2





321
HNRPU
NP_001073027.1
RNA processing
Y746
NYYGYQGyR
SEQ ID NO: 332



L2





322
DRBP1
NP_694453.2
RNA processing
Y204
SSEQDyYSNMRQE
SEQ ID NO: 333





323
E1B-
NP_008971.2
RNA processing
Y111
QNQFyDTQVIKQENESGYER
SEQ ID NO: 334



AP5





324
ELAVL1
NP_001410.2
RNA processing
Y26
TNLIVNyLPQNMTQDELR
SEQ ID NO: 335





325
EXOSC
NP_057130.1
RNA processing
Y32
HGyIFSSLAGCLMK
SEQ ID NO: 336



1





326
FXR1
NP_005078.2
RNA processing
Y353
ESIGNVQVLLEyHIAYLK
SEQ ID NO: 337





327
FXR1
NP_005078.2
RNA processing
Y357
ESIGNVQVLLEYHIAyLK
SEQ ID NO: 338





328
hnRNP
NP_036339.1
RNA processing
Y100
GFyDPPRR
SEQ ID NO: 339



2H9





329
hnRNP
NP_036339.1
RNA processing
Y156
GGDGYDGGYGGFDDYGGyNNYGYGND
SEQ ID NO: 340



2H9



GFDDR





330
hnRNP
NP_036339.1
RNA processing
Y181
GMGGHGyGGAGDASSGFHGGHFVHMR
SEQ ID NO: 341



2H9





331
hnRNP
NP_005511.1
RNA processing
Y236
GAyGGGYGGYDDYNGYNDGYGFGSDR
SEQ ID NO: 342



H





332
hnRNP
NP_005511.1
RNA processing
Y240
GAYGGGyGGYDDYNGYNDGYGFGSDR
SEQ ID NO: 343



H





333
hnRNP
NP_005511.1
RNA processing
Y253
GAYGGGYGGYDDYNGYNDGyGFGSDR
SEQ ID NO: 344



H





334
hnRNP
NP_062543.1
RNA processing
Y298
GLPyRATENDIYNFFSPLNPMR
SEQ ID NO: 345



H





335
hnRNP
NP_005817.1
RNA processing
Y624
FyQDTYGQQWK
SEQ ID NO: 346



R





336
hnRNP
NP_005817.1
RNA processing
Y628
FYQDTyGQQWK
SEQ ID NO: 347



R





337
hnRNP
NP_004492.2
RNA processing
Y635
GNFTLPEVAECFDEITyVELQKEEAQK
SEQ ID NO: 348



U





338
hnRNP-
NP_112420.1
RNA processing
Y244
GGGGYGGSGDGyNGFGNDGGYGGGG
SEQ ID NO: 349



A1



PGYSGGSR





339
hnRNP-
NP_112420.1
RNA processing
Y253
GGGGYGGSGDGYNGFGNDGGyGGGG
SEQ ID NO: 350



A1



PGYSGGSR





340
hnRNP-I
NP_002810.1
RNA processing
Y456
EGQEDQGLTKDyGNSPLHR
SEQ ID NO: 351





341
HUMAG
NP_037418.3
RNA processing
Y379
GQGGAyAFLK
SEQ ID NO: 352



CGB





342
IGF2BP
NP_006538.2
RNA processing
Y39
TGyAFVDCPDESWALK
SEQ ID NO: 353



3





343
MPHOS
NP_005782.1
RNA processing
Y452
PKEDAyEYK
SEQ ID NO: 354



PH10





344
MPHOS
NP_005782.1
RNA processing
Y454
PKEDAYEyK
SEQ ID NO: 355



PH10





345
NOL5A
NP_006383.2
RNA processing
Y210
IINDNATyCR
SEQ ID NO: 356





346
PABPN
NP_004634.1
RNA processing
Y46
GAPGGAGDyGNGLE
SEQ ID NO: 357



1





347
PSF
NP_005057.1
RNA processing
Y691
GMGPGTPAGyGR
SEQ ID NO: 358





348
RBM10
NP_005667.2
RNA processing
Y435
GTWATSEEPPVDySYYQQDE
SEQ ID NO: 359





349
RBM14
NP_006319.1
RNA processing
Y558
GQPGNAYDGAGQPSAAyLSMSQGAVA
SEQ ID NO: 360







NANSTPPPYER





350
RBM15
NP_073605.4
RNA processing
Y336
ERDYPFyER
SEQ ID NO: 361





351
RBM15
NP_073605.4
RNA processing
Y416
GQTSTyGFLK
SEQ ID NO: 362





352
SFRS5
NP_008856.2
RNA processing
Y55
FEDPRDADDAVyE
SEQ ID NO: 363





353
SFRS9
NP_003760.1
RNA processing
Y17
GGEGDGRIyVGNLPTDVR
SEQ ID NO: 364





354
SFRS9
NP_003760.1
RNA processing
Y35
EKDLEDLFyKYGR
SEQ ID NO: 365





355
SLU7
NP_006416.3
RNA processing
Y297
ENPyANAGKNPDEVSYAGDNFVR
SEQ ID NO: 366





356
snRNP
NP_003080.2
RNA processing
Y414
GLGNDSRDMyME
SEQ ID NO: 367



70





357
CGB
NP_001810.1
Secreted protein
Y526
LGELFNPYyDPLQWK
SEQ ID NO: 368





358
DEFA1
NP_004075.1
Secreted protein
Y80
yGTCIYQGR
SEQ ID NO: 369





359
DEFA3
NP_005208.1
Secreted protein
Y80
yGTCIYQGR
SEQ ID NO: 370





360
DEFA3
NP_005208.1
Secreted protein
Y85
YGTCIyQGR
SEQ ID NO: 371





361
FBS1
NP_071897.1
Secreted protein
Y308
LyGLEPAHPLLYSR
SEQ ID NO: 372





362
FBS1
NP_071897.1
Secreted protein
Y318
LYGLEPAHPLLySR
SEQ ID NO: 373





363
FGA
NP_000499.1
Secreted protein
Y277
GGSTSyGTGSETESPR
SEQ ID NO: 374





364
CEBPZ
NP_005751.2
Transcriptional
Y192
WYDLEYSNEySLKPQPQDVVSK
SEQ ID NO: 376





regulator





365
CNOT7
NP_037486.2
Transcriptional
Y260
YCGHLyGLGSGSSYVQNGTGNAYEEEA
SEQ ID NO: 377





regulator

NKQS





366
DTX2
NP_065943.1
Transcriptional
Y75
FGLGSLAHSIPLGQADPSLAPyIIDLPSWT
SEQ ID NO: 378





regulator

QFR





367
E2A
NP_003191.1
Transcriptional
Y150
GTSQYyPSYSGSSR
SEQ ID NO: 379





regulator





368
E2A
NP_003191.1
Transcriptional
Y153
GTSQYYPSySGSSR
SEQ ID NO: 380





regulator





369
Ets-1
NP_005229.1
Transcriptional
Y140
EDVKPyQVNGVNPAYPESR
SEQ ID NO: 381





regulator





370
FBP1
NP_003893.2
Transcriptional
Y242
ITGDPyKVQQAK
SEQ ID NO: 382





regulator





371
FBP1
NP_003893.2
Transcriptional
Y625
QQAAyYAQTSPQGMPQHPPAPQGQ
SEQ ID NO: 383





regulator





372
FHL2
NP_001441.4
Transcriptional
Y97
EDQLLCTDCYSNEySSK
SEQ ID NO: 384





regulator





373
FLI1
NP_002008.2
Transcriptional
Y451
HPNTHVPSHLGSyY
SEQ ID NO: 385





regulator





374
JunB
NP_002220.1
Transcriptional
Y68
GPGPEGGGGGSyFSGQGSDTGASLK
SEQ ID NO: 386





regulator





375
MTA2
NP_004730.2
Transcriptional
Y437
GHLSRPEAQSLSPyTTSANR
SEQ ID NO: 387





regulator





376
NFkB-
NP_002493.3
Transcriptional
Y55
FRyGCEGPSHGGLPGASSEK
SEQ ID NO: 388



p100

regulator





377
SPT5
NP_003160.2
Transcriptional
Y765
RPGGMTSTyGR
SEQ ID NO: 389





regulator





378
SPT5
NP_003160.2
Transcriptional
Y771
TPMyGSQTPMYGSGSR
SEQ ID NO: 390





regulator





379
SSRP1
NP_003137.1
Transcriptional
Y438
EGMNPSyDEYADSDEDQHDAYLER
SEQ ID NO: 391





regulator





380
eEF1A-
AAH71619.1
Translational
Y233
LPLQDVyK
SEQ ID NO: 392



1

regulator





381
eEF1A-
NP_001949.1
Translational
Y254
LPLQDVyK
SEQ ID NO: 393



2

regulator





382
eIF2B-
NP_003898.2
Translational
Y319
WVyPLTPEANFTDSTTQSCTHSR
SEQ ID NO: 395



epsilon

regulator





383
eIF3-
NP_003748.1
Translational
Y300
SySSGGEDGYVR
SEQ ID NO: 396



beta

regulator





384
eIF3S6I
NP_057175.1
Translational
Y247
QLEVyTSGGDPESVAGEYGR
SEQ ID NO: 397



P

regulator





385
eIF3S6I
NP_057175.1
Translational
Y287
LHSLLGDyYQAIK
SEQ ID NO: 398



P

regulator





386
eIF3S6I
NP_057175.1
Translational
Y288
LHSLLGDYyQAIK
SEQ ID NO: 399



P

regulator





387
eIF3S6I
NP_057175.1
Translational
Y357
TTYKyEMINK
SEQ ID NO: 400



P

regulator





388
eIF3-
NP_003744.1
Translational
Y506
yLILKDPNK
SEQ ID NO: 401



zeta

regulator





389
eIF4B
NP_001408.2
Translational
Y258
yDDRGSRDYDRGYDSR
SEQ ID NO: 402





regulator





390
PES1
NP_055118.1
Translational
Y265
AKAGEGTyALDSE
SEQ ID NO: 403





regulator





391
RPL18a
NP_000971.1
Translational
Y46
FWyFVSQLK
SEQ ID NO: 405





regulator





392
FAT
NP_005236.2
Tumor
Y4489
FNLNQyLPNFYPLDMSEPQTK
SEQ ID NO: 406





suppressor





393
FAT
NP_005236.2
Tumor
Y4519
GTGENSTCREPHAPyPPGYQR
SEQ ID NO: 407





suppressor





394
FAT
NP_005236.2
Tumor
Y4523
GTGENSTCREPHAPYPPGyQR
SEQ ID NO: 408





suppressor





395
COPS3
NP_003644.2
Ubiquitin
Y422
SMGSQEDDSGNKPSSyS
SEQ ID NO: 409





conjugating





system





396
CYLD
NP_056062.1
Ubiquitin
Y181
GQGFTDGVyQGK
SEQ ID NO: 410





conjugating





system





397
CYLD
NP_056062.1
Ubiquitin
Y356
SELFyTLNGSSVDSQPQSK
SEQ ID NO: 411





conjugating





system





398
ETEA
NP_055428.1
Ubiquitin
Y297
QQQDEAyLASLR
SEQ ID NO: 412





conjugating





system





399
HECW2
NP_065811.1
Ubiquitin
Y208
GMFFNPDPyLK
SEQ ID NO: 413





conjugating





system





400
RC3H1
NP_742068.1
Ubiquitin
Y593
GSQLYPAQQTDVYyQDPR
SEQ ID NO: 414





conjugating





system





401
SMURF
NP_065162.1
Ubiquitin
Y413
EEIFEESyRQIMK
SEQ ID NO: 415



1

conjugating





system





402
ACAD1
NP_115545.3
Unknown
Y324
MAGIAQGVySR
SEQ ID NO: 416



1

function





403
ANKRD
EAW96919.1
Unknown
Y207
GyGLLHTAAASGQIEWKYLLR
SEQ ID NO: 417



52

function





404
C17orf7
NP_060619.4
Unknown
Y309
LQHALEDQIyR
SEQ ID NO: 418



1

function





405
C18orf8
NP_037458.3
Unknown
Y610
QTEDNMLFyTIFR
SEQ ID NO: 419





function





406
C19orf2
NP_775752.1
Unknown
Y7
VTRyPILGIPQAHR
SEQ ID NO: 420



1

function





407
C1orf10
NP_776168.1
Unknown
Y23
ySTNSPNYR
SEQ ID NO: 421



1

function





408
C1orf10
NP_776168.1
Unknown
Y30
YSTNSPNyR
SEQ ID NO: 422



1

function





409
C1orf16
NP 777556.1
Unknown
Y120
LSSIPGESLTyASTTFK
SEQ ID NO: 423



2

function





410
C1orf16
NP_777556.1
Unknown
Y148
SNHLAENHSADFDPIVyAQIK
SEQ ID NO: 424



2

function





411
C1orf21
NP_110433.1
Unknown
Y34
NYQNGDVFGDEyR
SEQ ID NO: 425





function





412
C1orf32
NP_955383.1
Unknown
Y566
SASYyAWSPPGTYK
SEQ ID NO: 426





function





413
C1orf82
NP_079089.1
Unknown
Y127
TNKVyDITER
SEQ ID NO: 427





function





414
C1orf82
NP_079089.1
Unknown
Y319
LKASENSESEySR
SEQ ID NO: 428





function





415
C22orf5
NP_036396.2
Unknown
Y243
ELLSPySPVLKFFMVK
SEQ ID NO: 429





function





416
C22orf9
NP_056079.1
Unknown
Y66
LAySGSESGADGR
SEQ ID NO: 430





function





417
C2orf33
NP_064579.3
Unknown
Y35
IQyEMEYTEGISQR
SEQ ID NO: 431





function





418
C2orf33
NP_064579.3
Unknown
Y39
IQYEMEyTEGISQR
SEQ ID NO: 432





function





419
C3orf24
NP_775743.1
Unknown
Y163
SILLLyATYK
SEQ ID NO: 433





function





420
C3orf24
NP_775743.1
Unknown
Y166
QMLRSILLLYATyKK
SEQ ID NO: 434





function





421
C3orf58
NP_775823.1
Unknown
Y92
NVyFAQYGEPREGGRRR
SEQ ID NO: 435





function





422
C5orf32
NP_115788.1
Unknown
Y64
TTVyVVEDQR
SEQ ID NO: 437





function





423
C6orf14
NP_001010872.1
Unknown
Y523
FEGyDNPENLK
SEQ ID NO: 438



3

function





424
C6orf14
NP_001010872.1
Unknown
Y685
HYVySTLTR
SEQ ID NO: 439



3

function





425
C6orf14
NP_001010872.1
Unknown
Y896
FNTEQIQyR
SEQ ID NO: 440



3

function





426
C6orf14
NP_001010872.1
Unknown
Y986
SSPLLNYNTGVyR
SEQ ID NO: 441



3

function





427
C6orf14
NP_065141.3
Unknown
Y26
ESKRFSAyNYRTYAVR
SEQ ID NO: 442



9

function





428
C6orf14
NP_065141.3
Unknown
Y28
ESKRFSAYNyRTYAVR
SEQ ID NO: 443



9

function





429
C6orf14
NP_065141.3
Unknown
Y31
ESKRFSAYNYRTyAVR
SEQ ID NO: 444



9

function





430
CCDC1
NP_296375.1
Unknown
Y398
SSEVLyERPQPTPAFSSR
SEQ ID NO: 445



20

function





431
CCDC1
NP_996769.2
Unknown
Y884
SEEVYCLQK
SEQ ID NO: 446



8

function





432
CHORD
NP_036256.1
Unknown
Y292
SyVTMTATKIEITMR
SEQ ID NO: 447



C1

function





433
COBLL1
NP_055715.3
Unknown
Y533
STDGQEPHSVVyDTSNGKK
SEQ ID NO: 449





function





434
COBLL1
NP_055715.3
Unknown
Y742
IDKNSTASYLKNyPLYR
SEQ ID NO: 450





function





435
DAZAP
NP_055579.1
Unknown
Y165
KGNFFMGGSDGGyTIW
SEQ ID NO: 451



2

function





436
DENND
NP_056504.2
Unknown
Y370
TLSEENVyEDILDPPMK
SEQ ID NO: 452



2A

function





437
DENND
NP_940861.3
Unknown
Y195
SLENIySEPEGQECGPSINPLPKPR
SEQ ID NO: 453



2C

function





438
DEPDC
NP_631899.2
Unknown
Y300
ELLFDAIGRyYSSR
SEQ ID NO: 454



7

function





439
DKFZP4
CAD89901.1
Unknown
Y48
TLMLNEDKPSDDySAVLQR
SEQ ID NO: 455



51C023

function





440
MIER3
NP_689835.3
Unknown
Y331
YDyFAQQTR
SEQ ID NO: 456





function





441
DYX1C
NP_570722.2
Unknown
Y128
EDQKyALSVMMK
SEQ ID NO: 457



1

function





442
FAM102
NP_001030331.1
Unknown
Y376
VSSGVyEPVVIESH
SEQ ID NO: 458



A

function





443
FAM81
NP_689663.1
Unknown
Y21
HSQSLTMAPySSVSLVEQLEDR
SEQ ID NO: 459



A

function





444
FAM83
NP_060178.1
Unknown
Y100
QEPSGMAEGATTADVDAGSLSyWPGQS
SEQ ID NO: 460



E

function

EQPAPVLR





445
FLJ002
NP_689619.1
Unknown
Y126
NAADLPPPLPNKPPPEDyYEEALPLGPG
SEQ ID NO: 461



58

function

K





446
AFAP1L
NP_115939.1
Unknown
Y459
TDPEEFTYDyVDADR
SEQ ID NO: 463



2

function





447
FLJ147
NP_115734.1
Unknown
Y283
NLHHTQELLyESTKDFLQLR
SEQ ID NO: 464



32

function





448
FLJ206
NP_060377.1
Unknown
Y138
IAAyAYSALSQIR
SEQ ID NO: 465



25

function





449
FLJ220
NP_060395.4
Unknown
Y288
KLyPQLSSVHQK
SEQ ID NO: 466



52

function





450
FLJ220
NP_060395.4
Unknown
Y920
TTATVDTyESLLSDSNSNQSR
SEQ ID NO: 467



52

function





451
FLJ309
NP_659462.1
Unknown
Y415
TTLCNMLAENyKGK
SEQ ID NO: 468



76

function





452
FLJ346
NP_689578.1
Unknown
Y216
GSEEyYSFHESDLDLPEMGSGSMSSR
SEQ ID NO: 469



33

function





453
FLJ346
NP_689578.1
Unknown
Y217
GSEEYySFHESDLDLPEMGSGSMSSR
SEQ ID NO: 470



33

function





454
FRMPD
NP_055543.1
Unknown
Y596
HLyIDNAYSSDGLNQQLSQPGEAPCEAD
SEQ ID NO: 471



4

function

YR





455
FRMPD
NP_055543.1
Unknown
Y601
HLYIDNAySSDGLNQQLSQPGEAPCEAD
SEQ ID NO: 472



4

function

YR





456
FRYL
NP_055845.1
Unknown
Y93
QNGTEDESyEYRPR
SEQ ID NO: 473





function





457
FRYL
NP_055845.1
Unknown
Y95
RQNGTEDESYEyRPR
SEQ ID NO: 474





function





458
HSPA12
NP_079291.2
Unknown
Y21
ETAPTSAySSPAR
SEQ ID NO: 475



A

function





459
KIAA03
NP_056145.1
Unknown
Y808
GRVyNYMNAVER
SEQ ID NO: 476



76

function





460
KIAA03
NP_056145.1
Unknown
Y810
GRVYNyMNAVER
SEQ ID NO: 477



76

function





461
KIAA11
XP_371706.5
Unknown
Y3846
FQTNyASTTHLMTGK
SEQ ID NO: 478



09

function





462
KIAA12
XP_049078.7
Unknown
Y1332
GEIIySLDGSDCVHK
SEQ ID NO: 479



39

function





463
LIMD2
NP_085053.1
Unknown
Y102
GNyDEGFGR
SEQ ID NO: 480





function





464
LSR7
NP_061029.2
Unknown
Y158
GPVNyNVTTEFEK
SEQ ID NO: 481





function





465
NSUN2
NP_060225.4
Unknown
Y646
KLSSETySQAK
SEQ ID NO: 482





function





466
PHF8
NP_055922.1
Unknown
Y267
GEKIFyLIR
SEQ ID NO: 483





function





467
POF1B
NP_079197.2
Unknown
Y166
GSHFFPGNNVIyEK
SEQ ID NO: 484





function





468
QSER1
NP_001070254.1
Unknown
Y1411
EFAATNSyLGYFGDAK
SEQ ID NO: 485





function





469
SNX22
NP_079074.2
Unknown
Y92
GLEQRRQGLEAYIQGILyLNQEVPK
SEQ ID NO: 486





function





470
JIP4
NP_003962.3
Unknown
Y900
GNAGSAEDTVDISQTGVyTE
SEQ ID NO: 487





function





471
ST5
NP_005409.3
Unknown
Y308
GLPQLPSSCySVDR
SEQ ID NO: 488





function





472
SYF2
NP_056299.1
Unknown
Y226
FyGKYTAEIK
SEQ ID NO: 489





function





473
SYF2
NP_056299.1
Unknown
Y229
FYGKyTAEIK
SEQ ID NO: 490





function





474
CLTC
NP_004850.1
Vesicle protein
Y883
IyIDSNNNPER
SEQ ID NO: 491





475
CLTA
NP_001824.1
Vesicle protein
Y94
SNGPTDSyAAISQVDRLQSEPE
SEQ ID NO: 492





476
CSP
NP_079495.1
Vesicle protein
Y17
SLSTSGESLyHVLGLDK
SEQ ID NO: 493





477
CSP
NP_079495.1
Vesicle protein
Y192
TTQLTADSHPSyHTDGFN
SEQ ID NO: 494





478
EHBP1
NP_056067.1
Vesicle protein
Y319
VQTPQyLNPFDEPE
SEQ ID NO: 495





479
EHD2
NP_055416.2
Vesicle protein
Y458
YDEIFyNLAPADGK
SEQ ID NO: 496





480
epsin 3
NP_060427.1
Vesicle protein
Y176
RYGEDySR
SEQ ID NO: 497





481
EXOC1
NP_060731.2
Vesicle protein
Y769
VIySLGQPLEKLNHF
SEQ ID NO: 498





482
NSF
NP_006169.1
Vesicle protein
Y259
GILLyGPPGCGK
SEQ ID NO: 499





483
NSF
NP_006169.1
Vesicle protein
Y499
GDFLASLENDIKPAFGTNQEDyASYIMN
SEQ ID NO: 500







GIIK









One of skill in the art will appreciate that, in many instances the utility of the instant invention is best understood in conjunction with an appreciation of the many biological roles and significance of the various target signaling proteins/polypeptides of the invention. The foregoing is illustrated in the following paragraphs summarizing the knowledge in the art relevant to a few non-limiting representative peptides containing selected phosphorylation sites according to the invention.


CDH1 (Cadherin 1, E-cadherin, uvomorulin), phosphorylated at Y876, is among the proteins listed in this patent. It is a calcium-dependent glycoprotein that mediates cell-cell adhesion and migration and is necessary for epithelial morphogenesis (Nature Cell Biology 2002; 4:E101-E108.). Formation of adherens junctions between cells depends on the CDH1 cytoplasmic domain in which Y876 is located. When situated in an adherens junction, the cytoplasmic domain forms complexes with proteins such as catenins, Cdc42, PAR, atypical PKC, serves as a scaffold for forming adjacent tight junctions, activates PI(3) kinase, promotes actin polymerization and stabilizes microtubules (Nature Cell Biology 2002; 4:E101-E108.). Increased tyrosine phosphorylation of cadherins has been shown to suppress adhesion, suggesting a role for pY876 in regulating the stability of the adherens junction (J Cell Biol. 1995 August; 130(4):977-86.). 33-55% of sporadic diffuse-type gastric cancers carry somatic mutations of CDH1, and germline mutations in the gene cause the disease (Cancer Cell 2004 February; 5(2):121-125.). In addition, altered expression of CDH1 may be therapeutic for breast and colon cancer (Human PSD™, Biobase Corporation, Beverly, Mass.). Molecular probes for pY876 may be useful for diagnostic and/or therapeutic purposes for lung neoplasms (Anticancer Res 2003 July-August; 23(4):3367-71.) and gastric cancer (Cancer Cell 2004 February; 5(2):121-125. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


FGFR2, phosphorylated at Y466, Y586, Y588, Y616, Y733, Y805, and Y812, is among the proteins listed in this patent. Fibroblast growth factor receptor 2 is a receptor tyrosine kinase of the highly-conserved FGFR family that binds fibroblast growth factor (FGF) and acts in induction of apoptosis, skeletal development, cell migration and differentiation (Human PSD™, Biobase Corporation, Beverly, Mass.). Consistent with its role in development, mutations in the FGFR2 gene are known to cause at least three craniosynostotic conditions—Crouzon syndrome (Nat. Genet. 1994 September; 8(1):98-103. Nat. Genet. 1994 November; 8(3):275-9), Apert syndrome (Science. 2003 Aug. 1; 301(5633):643-6. Nat. Genet. 1996 May; 13(1):48-53. Nat. Genet. 1995 February; 9(2):165-72), Pfeiffer syndrome (Eur J Hum Genet. 2006 March; 14(3):289-98)—as well as autosomal dominant lacrimoauriculodentodigital (LADD) syndrome (Nat. Genet. 2006 April; 38(4):414-7). The gene is also associated with gastric cancer (Cancer Res. 2001 May 1; 61(9):3541-3) and breast cancer (Nature. 2007 Jun. 28; 447(7148):1087-93. Nat. Genet. 2007 July; 39(7):870-4). Y586, Y588, Y616, and Y733 are all within the kinase catalytic domain, and their phosphorylation may affect catalytic activity or recognition of substrate. Molecular probes to these and other sites on the protein would provide valuable tools to study the function of this protein in normal and pathological states (PhosphoSite®, Cell Signaling Technology, Danvers, Mass.).


CaMK2-delta, phosphorylated at Y14, is among the proteins listed in this patent. Calcium calmodulin dependent protein kinase II delta is a member of the CaMKII enzyme complex which is composed of four different chains: alpha, beta, gamma, and delta. The different chains assemble into homo- or heteromultimeric holoenzymes composed of 8 to 12 subunits (J Biol. Chem. 2007 Mar. 9; 282(10):7219-31. PhosphoSite®, Cell Signaling Technology, Danvers, Mass.). CAMK2-delta differs from the other subunits in having a variable domain, generated by alternative splicing, that acts as a nuclear localization signal. When the CAMK2-delta subunit predominates in the holoenzyme, as it does in the adult heart, the complex is expressed in the nucleus (J Biol. Chem. 2007 Mar. 9; 282(10):7219-31). Each of the CaMK2 chains contains a site at the N-terminus of the kinase catalytic domain that is paralogous to but distinct from Y14, suggesting that regulation of this site may have been evolutionarily conserved. The sequence differences surrounding the phosphorylation site may distinguish substrates of different kinases or affect the phosphorylation efficiency of a single kinase. An antibody to Y14 would be useful in clarifying these possibilities. Disregulation of CAMK2-delta is associated with cardiac hypertrophy and dilated cardiomyopathy (Circ Res. 2003 May 2; 92(8):912-9. J Biol. Chem. 2002 Jan. 11; 277(2):1261-7), and molecular probes specific for this protein and its modification would be valuable in assessing its role in this disease (Human PSD™, Biobase Corporation, Beverly, Mass.).


Cdc42, phosphorylated at Y32, is among the proteins listed in this patent. Cell division cycle 42 (GTP binding protein 25 kD) is a Rho GTPase which regulates signaling pathways that control diverse cellular functions including cell morphology, migration, endocytosis and cell cycle progression (PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.). In particular, Cdc42 activity is prominent at the leading edge of motile cells (Science 2004 Sep. 10; 305(5690):1615-1619. Mol Cell Biol. 2002 September; 22(18):6582-91) where it is controlled by LKB1. Disregulation of Cdc42 in the absence of LKB1 is associated with non-small cell lung cancer (Cancer Res. 2008 Feb. 1; 68(3):740-8) and with breast neoplasms (Br J Cancer 2002 Sep. 9; 87(6):635-44.). Y32 is located within the docking site for guanine nucleotide exchange factor, and is a potential target for therapeutic intervention in cancer. Molecular probes to this site may provide insight into regulation of Cdc42 activity.


SKB1 (protein arginine methyltransferase 5; PRMT5), phosphorylated at Y283, is among the proteins listed in this patent. By methylating its target proteins on arginine residues, SKB1 modulates the accessibility of DNA for transcription and replication (Human PSD™, Biobase Corporation, Beverly, Mass.) and enables small nuclear ribonucleoproteins to function within the spliceosome (J Cell Biol 2007; 178(5):733-740.). Abnormally high levels of SKB1 expression are found in lymphoid cancer, mantle cell lymphoma and gastric cancer samples (EMBO J 2007; 26:3558-3569. Human PSD™, Biobase Corporation, Beverly, Mass.). Y283 resides in the catalytic domain of the enzyme, and molecular probes of its phosphorylation state may provide insight into its activity in normal and pathological tissues. The SKB1 protein has potential diagnostic and/or therapeutic implications based on association with lymphoid cancer, mantle cell lymphoma, gastric cancer and breast neoplasms (EMBO J 2007; 26:3558-3569. Biochem Biophys Res Commun 2005 Apr. 8; 329(2):522-30. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


Three ribosomal protein S6 kinase family members are among the proteins listed in this patent. RSK2 Y433 (SEQ ID NO: 246), RSK3 Y434 (SEQ ID NO: 248), and RSK4 Y437 (SEQ ID NO: 250) are paralogous sites. RSK2 Y644 (SEQ ID NO: 247) is unique to this member. RSK3 Y707 (SEQ ID NO: 249) has paralogous sites in the other members that are not described here. The ribosomal S6 kinases participate in the transduction of signals from extracellular stimuli, such as hormones and neurotransmitters (Mol Cell Endocrinol 1999 May 25; 151(1-2):65-77.). The RSKs are distantly related to CK2-alpha1, also among the proteins listed in this patent. Based on the CK2-alpha1 crystal structure, RSK2 Y433, RSK3 Y434, RSK4 Y437 and CK2-alpha1 (SEQ ID NO: 240) are within the phosphate anchor element of the active site (EMBO J 1998; 17:2451-2462.). Phosphorylation of these sites may affect kinase activity. Mutations in the RSK2 gene cause Coffin-Lowry mental retardation syndrome, and the protein is prominently expressed in brain structures that are essential for cognitive function and learning (Am J Hum Genet 1998 December; 63(6):1631-40. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.). Molecular probes to these proteins would provide insight into the activation of cells and into their effects on development of the nervous system.


Claudin 5, phosphorylated at Y212 and Y217, is among the proteins listed in this patent. The claudin family of proteins contributes to tight junction function, and different members of the family may have distinct effects on the barrier properties of the junction (Am J Physiol Lung Cell Mol Physiol 2003 August; 285:L1166-L1178.). In claudin 4, phosphorylation of a site similar to but distinct from Y217 has been shown to increase the permeability of tight junctions, and a need for specific antibodies to the various claudin family members was stated in the same report (J Biol Chem 2005 December; 280(51):42375-42382.). pY212 and pY217 of claudin 5 were observed in numerous lung cancer samples (Cell 2007 Dec. 14; 131(6):1190-1203.), and expression of the protein differentiates lung squamous cell carcinomas from adenocarcinomas (Mod Pathol 2007 September; 20(9):947-54.), suggesting that molecular probes to this protein would valuable in the diagnosis and/or treatment of lung cancers. In addition, polymorphism in the gene locus may be associated with schizophrenia (PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


Three members of the discs large homologue family (DLG3, PSD-93, PDS-95), as well as two of the proteins that are known to interact with them (SAPAP1, SAPAP3), are among the proteins listed in this patent. Discs large proteins participate in multi-protein complexes at areas of intercellular contact, such as synapses, where they contribute to cell proliferation, neuron adhesion and synaptic transmission (Genes Dev. 2004 Aug. 15; 18(16):1909-25); SAPAP proteins bind to DLG proteins at excitatory synapses with SAPAP1 found in cell bodies and SAPAP3 in dendrites (J Comp Neurol. 2004 Apr. 19; 472(1):24-39). Searches for safer and more effective anesthetics have shown that the inhaled anesthetic halothane disrupts interactions between DLG proteins and nNOS and NMDA receptors and that more targeted anesthetics may be developed by designing drugs to disrupt the interactions of specific DLG proteins. PSD-93 Y223 (SEQ ID NO: 32) and PSD95 Y233 (SEQ ID NO: 34) are adjacent to the halothane binding site, and molecular probes against these sites would provide insight into the molecular mechanisms of anesthetics (Mol. Interv. 2004 August; 4(4):215-21. J Biol. Chem. 2003 Sep. 19; 278(38):36669-75). Consistent with its role in neuronal function, mutation of DLG3 is associated with X-linked mental retardation, and its chromosomal position correlates with Parkinson disease (Human PSD™, Biobase Corporation, Beverly, Mass.); antibodies to Y808 (SEQ ID NO: 7) or other parts of the protein may serve as useful tools in dissecting the molecular mechanisms of these diseases. Similarly, SAPAP1 is upregulated in schizophrenia (Neuropsychopharmacology. 2003 October; 28(10): 1831-9) and its gene maps to a chromosomal region associated with psychiatric illness. (Psychiatry Genet. 2005 March; 15(1):37-44. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


Cbl (Cas-Br-M ecotropic retroviral transforming sequence), phosphorylated at Y141, is among the proteins listed in this patent. Cbl is an ubiquitin protein ligase that acts in cell migration, apoptosis, and microtubule polymerization. Its increased phosphorylation correlates with several neoplasms, and mouse Cbl is associated with AIDS. This protein has potential diagnostic and/or therapeutic implications based on association with myelocytic leukemia and lymphocytic leukemia (Am J Hum Genet 1996 November; 59(5):990-8. PhosphoSite®, Cell Signaling Technology, Danvers, Mass., Human PSD™, Biobase Corporation, Beverly, Mass.).


LSD1, phosphorylated at Y135, is among the proteins listed in this patent. Lysine-specific histone demethylase 1 is a transcription corepressor that plays a role in hydrogen peroxide and formaldehyde biosynthetic processes. It may also play a role in chromatin modification and silencing. LSD1 expression is upregulated in prostate carcinoma. (PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


PIK3R2, phosphorylated at Y460, is among the proteins listed in this patent. Phosphoinositide-3-kinase regulatory polypeptide 2 is a regulatory subunit of phosphatidylinositol 3-kinase that acts in signal transduction, cell motility and differentiation; tumorigenic fusion to USP8 gene may lead to chronic myeloproliferative disorder. (PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


CD82, phosphorylated at Y261, is among the proteins listed in this patent. It plays a role in T cell development, induces apoptosis, and inhibits cell migration, motility and adhesion. Its increased expression is associated with non small cell lung carcinoma; aberrant expression is associated with multiple neoplasms. CD82 has potential diagnostic and/or therapeutic implications based on association with colorectal neoplasms (Anticancer Res 2001 Septemer-October; 21(5):3581-7. PhosphoSite®, Cell Signaling Technology, Danvers, Mass., Human PSD™, Biobase Corporation, Beverly, Mass.).


KPNA3, phosphorylated at Y506, is among the proteins listed in this patent. Karyopherin alpha 3 binds to nuclear localization signals (NLS) and plays a role in nuclear import. Its gene expression is downregulated in B cell chronic lymphocytic leukemia. (PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


FLT3, phosphorylated at Y865, is among the proteins listed in this patent. Fms-related receptor tyrosine kinase 3 is a receptor for the hematopoietic growth factor FLT3LG that stimulates proliferation of stem and progenitor cells. Internal tandem duplication or mutations in the gene are associated with acute myeloid leukemia (AML). This protein has potential diagnostic and/or therapeutic implications based on association with myelocytic leukemia (Blood 2000 Dec. 1; 96(12):3907-14. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


Ets-1, phosphorylated at Y140, is among the proteins listed in this patent. V-ets avian erythroblastosis virus E26 oncogene homolog 1 is a transcriptional activator of genes involved in angiogenesis, immune response, apoptosis, cell proliferation, adhesion, and migration; it is also associated with cancer and rheumatoid arthritis. This protein has potential diagnostic and/or therapeutic implications based on association adenocarcinoma and colonic neoplasms (Anticancer Res 2002 May-June; 22(3):1581-4. PhosphoSite®, Cell Signaling Technology Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


CTNND1, phosphorylated at Y208, is among the proteins listed in this patent. Catenin (cadherin associated protein) delta 1, links cadherin E (CDH1) to cytoskeleton, regulates GTPase activation, apoptosis, cell proliferation and cell migration. Its altered expression is associated with several cancers. This protein has potential diagnostic and/or therapeutic implications based on association with colorectal neoplasms, stomach neoplasms, and pancreatic neoplasms (Anticancer Res 1999 Septemer-October; 19(5C):4401-5. PhosphoSite®, Cell Signaling Technology, Danvers, Mass. Human PSD™ Biobase Corporation, (Beverly, Mass.).


COPS3, phosphorylated at Y422, is among the proteins listed in this patent. It is a component of the COP9 signalosome complex (CSN), a complex involved in various cellular and developmental processes. The CSN complex regulates the ubiquitin (Ubl) conjugation pathway by mediating the deneddylation of the cullin subunits of SCF-type E3 ligase complexes, leading to a decrease in the Ubl ligase activity of SCF-type complexes such as SCF, CSA or DDB2. The complex is also involved in phosphorylation of p53/TP53, c-jun/JUN, IkappaBalpha/NFKBIA, ITPK1 and IRF8/ICSBP, possibly via its association with CK2 and PKD kinases. CSN-dependent phosphorylation of TP53 and JUN promotes and protects degradation by the Ubl system, respectively (PhosphoSite®, Cell Signaling Technology, Danvers, Mass.). Amplification and overexpression of COPS3 is associated with osteosarcoma, and molecular probes, such as antibodies against pY422, have potential diagnostic value for this disease (Cancer. 2007 May 1; 109(9):1870-6; Oncogene 2003 Aug. 14; 22(34):5358-61. Human PSD™, Biobase Corporation, Beverly, Mass.).


The invention identifies peptide sequences comprising phosphorylation sites useful to profile phosphotyrosine signaling in the analysis of oncogenesis and specifically in lung cancer (e.g., in non-small lung cancer, NSLC). For most solid tumors the tyrosine kinases that drive disease remain unknown, limiting the ability to identify drug targets and predict response. Tyrosine kinase signaling across 41 NSCLC cells lines and 150 NSCLC tumors have implicated a number of known oncogenic kinases such as EGFR and c-Met as well as novel ALK and ROS fusion proteins, along with others such as PDGFRα and DDR1. The compendium of phosphorylated sites provided herein constitutes a fundamental tool to profile a given sample across many possible target phosphorylation determinants offering a unique tool to characterize a given tumor to identify drug targets and predict response. The invention also provides peptides comprising a novel phosphorylation site of the invention. In one particular embodiment, the peptides comprise any one of the an amino acid sequences as set forth in column E of Table 1 and FIG. 2, which are trypsin-digested peptide fragments of the parent proteins. Alternatively, a parent signaling protein listed in Table 1 may be digested with another protease, and the sequence of a peptide fragment comprising a phosphorylation site can be obtained in a similar way. Suitable proteases include, but are not limited to, serine proteases (e.g. hepsin), metallo proteases (e.g. PUMP1), chymotrypsin, cathepsin, pepsin, thermolysin, carboxypeptidases, etc.


The invention also provides proteins and peptides that are mutated to eliminate a novel phosphorylation site of the invention. Such proteins and peptides are particular useful as research tools to understand complex signaling transduction pathways of cancer cells, for example, to identify new upstream kinase(s) or phosphatase(s) or other proteins that regulates the activity of a signaling protein; to identify downstream effector molecules that interact with a signaling protein, etc.


Various methods that are well known in the art can be used to eliminate a phosphorylation site. For example, the phosphorylatable tyrosine may be mutated into a non-phosphorylatable residue, such as phenylalanine. A “phosphorylatable” amino acid refers to an amino acid that is capable of being modified by addition of a phosphate group (any includes both phosphorylated form and unphosphorylated form). Alternatively, the tyrosine may be deleted. Residues other than the tyrosine may also be modified (e.g., delete or mutated) if such modification inhibits the phosphorylation of the tyrosine residue. For example, residues flanking the tyrosine may be deleted or mutated, so that a kinase can not recognize/phosphorylate the mutated protein or the peptide. Standard mutagenesis and molecular cloning techniques can be used to create amino acid substitutions or deletions.


2. Modulators of the Phosphorylation Sites


In another aspect, the invention provides a modulator that modulates tyrosine phosphorylation at a novel phosphorylation site of the invention, including small molecules, peptides comprising a novel phosphorylation site, and binding molecules that specifically bind at a novel phosphorylation site, including but not limited to antibodies or antigen-binding fragments thereof.


Modulators of a phosphorylation site include any molecules that directly or indirectly counteract, reduce, antagonize or inhibit tyrosine phosphorylation of the site. The modulators may compete or block the binding of the phosphorylation site to its upstream kinase(s) or phosphatase(s), or to its downstream signaling transduction molecule(s).


The modulators may directly interact with a phosphorylation site. The modulator may also be a molecule that does not directly interact with a phosphorylation site. For example, the modulators can be dominant negative mutants, i.e., proteins and peptides that are mutated to eliminate the phosphorylation site. Such mutated proteins or peptides could retain the binding ability to a downstream signaling molecule but lose the ability to trigger downstream signaling transduction of the wild type parent signaling protein.


The modulators include small molecules that modulate the tyrosine phosphorylation at a novel phosphorylation site of the invention. Chemical agents, referred to in the art as “small molecule” compounds are typically organic, non-peptide molecules, having a molecular weight less than 10,000, less than 5,000, less than 1,000, or less than 500 daltons. This class of modulators includes chemically synthesized molecules, for instance, compounds from combinatorial chemical libraries. Synthetic compounds may be rationally designed or identified based on known or inferred properties of a phosphorylation site of the invention or may be identified by screening compound libraries. Alternative appropriate modulators of this class are natural products, particularly secondary metabolites from organisms such as plants or fungi, which can also be identified by screening compound libraries. Methods for generating and obtaining compounds are well known in the art (Schreiber S L, Science 151: 1964-1969 (2000); Radmann J. and Gunther J., Science 151: 1947-1948 (2000)).


The modulators also include peptidomimetics, small protein-like chains designed to mimic peptides. Peptidomimetics may be analogues of a peptide comprising a phosphorylation site of the invention. Peptidomimetics may also be analogues of a modified peptide that are mutated to eliminate a phosphorylation site of the invention. Peptidomimetics (both peptide and non-peptidyl analogues) may have improved properties (e.g., decreased proteolysis, increased retention or increased bioavailability). Peptidomimetics generally have improved oral availability, which makes them especially suited to treatment of disorders in a human or animal.


In certain embodiments, the modulators are peptides comprising a novel phosphorylation site of the invention. In certain embodiments, the modulators are antibodies or antigen-binding fragments thereof that specifically bind at a novel phosphorylation site of the invention.


3. Heavy-Isotope Labeled Peptides (AQUA Peptides)


In another aspect, the invention provides peptides comprising a novel phosphorylation site of the invention. In a particular embodiment, the invention provides Heavy-Isotype Labeled Peptides (AQUA peptides) comprising a novel phosphorylation site. Such peptides are useful to generate phosphorylation site-specific antibodies for a novel phosphorylation site. Such peptides are also useful as potential diagnostic tools for screening carcinoma and/or leukemia, or as potential therapeutic agents for treating carcinoma and/or leukemia.


The peptides may be of any length, typically six to fifteen amino acids. The novel tyrosine phosphorylation site can occur at any position in the peptide; if the peptide will be used as an immunogen, it preferably is from seven to twenty amino acids in length. In some embodiments, the peptide is labeled with a detectable marker.


“Heavy-isotope labeled peptide” (used interchangeably with AQUA peptide) refers to a peptide comprising at least one heavy-isotope label, as described in WO/03016861, “Absolute Quantification of Proteins and Modified Forms Thereof by Multistage Mass Spectrometry” (Gygi et al.) (the teachings of which are hereby incorporated herein by reference, in their entirety). The amino acid sequence of an AQUA peptide is identical to the sequence of a proteolytic fragment of the parent protein in which the novel phosphorylation site occurs. AQUA peptides of the invention are highly useful for detecting, quantitating or modulating a phosphorylation site of the invention (both in phosphorylated and unphosphorylated forms) in a biological sample.


A peptide of the invention, including an AQUA peptides comprises any novel phosphorylation site. Preferably, the peptide or AQUA peptide comprises a novel phosphorylation site of a protein in Table 1 that is a adaptor/scaffold proteins; adhesion or extracellular matrix proteins; cytoskeletal proteins; enzyme proteins; G proteins or regulator proteins; non-protein kinase proteins; motor or contractile proteins; phosphatase proteins; protein kinases; receptor, channel, transporter or cell surface proteins; RNA processing proteins; and transcriptional regulator proteins.


Particularly preferred peptides and AQUA peptides are these comprising a novel tyrosine phosphorylation site (shown as a lower case “y” in a sequence listed in Table 1) selected from the group consisting of SEQ ID NOs: 320 (DDX1), 322 (DDX3), 323 (DDX3), 327 (DDX3Y), 154 (FBPase), 155 (G6PI), 200 (PIP5K), 222 (CD45), 223 (DARPP-32), 225 (FBP2), 238 (Cdc2), 239 (CDK9), 240 (CK2-alpha1), 247 (RSK2), 250 (RSK4), 251 (SRPK1), 266 (FGFR1), 268 (FGFR2), 275 (FLT3), 277 (Met), 35 (PSD-95), 97 (CAPZA2), 103 (cofilin 1), 286 (Cx43), 164 (Cdc42), 63 (SEMA4F), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 68 (CALB2), 73 (FREQ), 84 (DNAJB6), 93 (HMGB1), 97 (CAPZA2), 103 (cofilin 1), 164 (Cdc42), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 278 (PDGFRb), 286 (Cx43), 491 (CLTC), and 499 (NSF).


In some embodiments, the peptide or AQUA peptide comprises the amino acid sequence shown in any one of the above listed SEQ ID NOs. In some embodiments, the peptide or AQUA peptide consists of the amino acid sequence in said SEQ ID NOs. In some embodiments, the peptide or AQUA peptide comprises a fragment of the amino acid sequence in said SEQ ID NOs., wherein the fragment is six to twenty amino acid long and includes the phosphorylatable tyrosine. In some embodiments, the peptide or AQUA peptide consists of a fragment of the amino acid sequence in said SEQ ID NOs., wherein the fragment is six to twenty amino acid long and includes the phosphorylatable tyrosine.


In certain embodiments, the peptide or AQUA peptide comprises any one of the SEQ ID NOs listed in column H, which are trypsin-digested peptide fragments of the parent proteins.


It is understood that parent protein listed in Table 1 may be digested with any suitable protease (e.g., serine proteases (e.g. trypsin, hepsin), metallo proteases (e.g. PUMP1), chymotrypsin, cathepsin, pepsin, thermolysin, carboxypeptidases, etc), and the resulting peptide sequence comprising a phosphorylated site of the invention may differ from that of trypsin-digested fragments (as set forth in Column E), depending the cleavage site of a particular enzyme. An AQUA peptide for a particular a parent protein sequence should be chosen based on the amino acid sequence of the parent protein and the particular protease for digestion; that is, the AQUA peptide should match the amino acid sequence of a proteolytic fragment of the parent protein in which the novel phosphorylation site occurs.


An AQUA peptide is preferably at least about 6 amino acids long. The preferred ranged is about 7 to 15 amino acids.


The AQUA method detects and quantifies a target protein in a sample by introducing 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. By comparing to the peptide standard, one may readily determines the quantity of a peptide having the same sequence and protein modification(s) in the biological sample. Briefly, the AQUA methodology has two stages: (1) peptide internal standard selection and validation; method development; and (2) implementation using validated peptide internal standards to detect and quantify a target protein in a 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 used, e.g., to quantify change in protein phosphorylation as a result of drug treatment, or to quantify 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 a particular protease for digestion. 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 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 the modified form of the 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 may be developed for a known phosphorylation site previously identified by the IAP-LC-MS/MS method within a target protein. One AQUA peptide incorporating the phosphorylated form of the site, and a second AQUA peptide incorporating the unphosphorylated form of site may be developed. In this way, the two standards may be used to detect and quantify both the phosphorylated and unphosphorylated 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. Thus, peptides longer than about 20 amino acids are not preferred. The preferred ranged is about 7 to 15 amino acids. A peptide sequence is also selected that is not likely to be chemically reactive during mass spectrometry, thus sequences comprising cysteine, tryptophan, or methionine are avoided.


A peptide sequence that is outside a phosphorylation site may be selected as internal standard to determine the quantity of all forms of the target protein. Alternatively, a peptide encompassing a phosphorylated site may be selected as internal standard to detect and quantify only the phosphorylated form of the target protein. Peptide standards for both phosphorylated form and unphosphorylated form can be used together, to determine, the extent of phosphorylation in a particular 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 13C, 15N, 17O, 18O, or 34S, are among preferred labels. Pairs of peptide internal standards that incorporate a different isotope label may also be prepared. Preferred amino acid residues into which a heavy isotope label may be incorporated include leucine, proline, valine, and phenylalanine.


Peptide internal standards are characterized according to their mass-to-charge (m/z) ratio, and preferably, also according to their retention time on a chromatographic column (e.g. an HPLC column). Internal standards that co-elute with unlabeled peptides of identical sequence are selected as optimal internal standards. The internal standard is then analyzed by fragmenting the peptide by any suitable means, for example by collision-induced dissociation (CID) using, e.g., argon or helium as a collision gas. 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 are preferably used. 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 preferred method.


Each isolated peptide is then examined by monitoring of a selected reaction in the MS. This involves using the prior knowledge gained by the characterization of the peptide internal standard and then requiring the MS to continuously monitor a specific ion in the MS/MS or 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.


Accordingly, AQUA internal peptide standards (heavy-isotope labeled peptides) may be produced, as described above, for any of the 482 novel phosphorylation sites of the invention (see Table 1/FIG. 2). For example, peptide standards for a given phosphorylation site (e.g., an AQUA peptide having the sequence INMLTAGyAER (SEQ ID NO: 3), wherein “y” corresponds to phosphorylatable tyrosine 671 of CNKSR2) may be produced for both the phosphorylated and unphosphorylated forms of the sequence. Such standards may be used to detect and quantify both phosphorylated form and unphosphorylated form of the parent signaling protein (e.g., CNKSR2) in a biological sample.


Heavy-isotope labeled equivalents of a phosphorylation site of the invention, 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.


The novel phosphorylation sites of the invention are particularly well suited for development of corresponding AQUA peptides, since the IAP method by which they were identified (see Part A above and Example 1) inherently confirmed that such peptides are in fact produced by enzymatic digestion (e.g., 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) that may be used for detecting, quantitating, or modulating any of the phosphorylation sites of the invention (Table 1). For example, an AQUA peptide having the sequence FMDyVQLHSTDK (SEQ ID NO: 38), wherein y (Tyr 122) may be either phosphotyrosine or tyrosine, and wherein V=labeled valine (e.g., 14C)) is provided for the quantification of phosphorylated (or unphosphorylated) form of SHANK3 (an adaptor/scaffold protein) in a biological sample.


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, AQUA peptides corresponding to both the phosphorylated and unphosphorylated forms of SEQ ID NO:38 (a trypsin-digested fragment of SHANK3, with a tyrosine 122 phosphorylation site) may be used to quantify the amount of phosphorylated SHANK3 in a biological sample, e.g., a tumor cell sample or a sample before or after treatment with a therapeutic agent.


Peptides and AQUA peptides provided by the invention will be highly useful in the further study of signal transduction anomalies underlying cancer, including carcinoma and/or leukemias. Peptides and AQUA peptides of the invention may also be used for identifying diagnostic/bio-markers of carcinoma and/or leukemias, identifying new potential drug targets, and/or monitoring the effects of test therapeutic agents on signaling proteins and pathways.


4. Phosphorylation Site-Specific Antibodies


In another aspect, the invention discloses phosphorylation site-specific binding molecules that specifically bind at a novel tyrosine phosphorylation site of the invention, and that distinguish between the phosphorylated and unphosphorylated forms. In one embodiment, the binding molecule is an antibody or an antigen-binding fragment thereof. The antibody may specifically bind to an amino acid sequence comprising a phosphorylation site identified in Table 1.


In some embodiments, the antibody or antigen-binding fragment thereof specifically binds the phosphorylated site. In other embodiments, the antibody or antigen-binding fragment thereof specially binds the unphosphorylated site. An antibody or antigen-binding fragment thereof specially binds an amino acid sequence comprising a novel tyrosine phosphorylation site in Table 1 when it does not significantly bind any other site in the parent protein and does not significantly bind a protein other than the parent protein. An antibody of the invention is sometimes referred to herein as a “phospho-specific” antibody.


An antibody or antigen-binding fragment thereof specially binds an antigen when the dissociation constant is ≦1 mM, preferably ≦100 nM, and more preferably ≦10 nM.


In some embodiments, the antibody or antigen-binding fragment of the invention binds an amino acid sequence that comprises a novel phosphorylation site of a protein in Table 1 that is a adaptor/scaffold proteins; adhesion or extracellular matrix proteins; cytoskeletal proteins; enzyme proteins; G proteins or regulator proteins; non-protein kinase proteins; motor or contractile proteins; phosphatase proteins; protein kinases; receptor, channel, transporter or cell surface proteins; RNA processing proteins; and transcriptional regulator proteins.


In particularly preferred embodiments, an antibody or antigen-binding fragment thereof of the invention specially binds an amino acid sequence comprising a novel tyrosine phosphorylation site shown as a lower case “y” in a sequence listed in Table 1 selected from the group consisting of SEQ ID NOS: 320 (DDX1), 322 (DDX3), 323 (DDX3), 327 (DDX3Y), 154 (FBPase), 155 (G6PI), 200 (PIP5K), 222 (CD45), 223 (DARPP-32), 225 (FBP2), 238 (Cdc2), 239 (CDK9), 240 (CK2-alpha1), 247 (RSK2), 250 (RSK4), 251 (SRPK1), 266 (FGFR1), 268 (FGFR2), 275 (FLT3), 277 (Met), 35 (PSD-95), 97 (CAPZA2), 103 (cofilin 1), 286 (Cx43), 164 (Cdc42), 63 (SEMA4F), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 68 (CALB2), 73 (FREQ), 84 (DNAJB6), 93 (HMGB1), 97 (CAPZA2), 103 (cofilin 1), 164 (Cdc42), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 278 (PDGFRb), 286 (Cx43), 491 (CLTC), and 499 (NSF).


In some embodiments, an antibody or antigen-binding fragment thereof of the invention specifically binds an amino acid sequence comprising any one of the above listed SEQ ID NOs. In some embodiments, an antibody or antigen-binding fragment thereof of the invention especially binds an amino acid sequence comprises a fragment of one of said SEQ ID NOs., wherein the fragment is four to twenty amino acid long and includes the phosphorylatable tyrosine.


In certain embodiments, an antibody or antigen-binding fragment thereof of the invention specially binds an amino acid sequence that comprises a peptide produced by proteolysis of the parent protein with a protease wherein said peptide comprises a novel tyrosine phosphorylation site of the invention. In some embodiments, the peptides are produced from trypsin digestion of the parent protein. The parent protein comprising the novel tyrosine phosphorylation site can be from any species, preferably from a mammal including but not limited to non-human primates, rabbits, mice, rats, goats, cows, sheep, and guinea pigs. In some embodiments, the parent protein is a human protein and the antibody binds an epitope comprising the novel tyrosine phosphorylation site shown by a lower case “y” in Column E of Table 1. Such peptides include any one of the SEQ ID NOs.


An antibody of the invention can be an intact, four immunoglobulin chain antibody comprising two heavy chains and two light chains. The heavy chain of the antibody can be of any isotype including IgM, IgG, IgE, IgG, IgA or IgD or sub-isotype including IgG1, IgG2, IgG3, IgG4, IgE1, IgE2, etc. The light chain can be a kappa light chain or a lambda light chain.


Also within the invention are antibody molecules with fewer than 4 chains, including single chain antibodies, Camelid antibodies and the like and components of the antibody, including a heavy chain or a light chain. The term “antibody” (or “antibodies”) refers to all types of immunoglobulins. The term “an antigen-binding fragment of an antibody” refers to any portion of an antibody that retains specific binding of the intact antibody. An exemplary antigen-binding fragment of an antibody is the heavy chain and/or light chain CDR, or the heavy and/or light chain variable region. The term “does not bind,” when appeared in context of an antibody's binding to one phospho-form (e.g., phosphorylated form) of a sequence, means that the antibody does not substantially react with the other phospho-form (e.g., non-phosphorylated form) of the same sequence. 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 some embodiments an immunoglobulin chain may comprise in order from 5′ to 3′, a variable region and a constant region. The variable region may comprise three complementarity determining regions (CDRs), with interspersed framework (FR) regions for a structure FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. Also within the invention are heavy or light chain variable regions, framework regions and CDRs. An antibody of the invention may comprise a heavy chain constant region that comprises some or all of a CH1 region, hinge, CH2 and CH3 region.


An antibody of the invention may have an binding affinity (KD) of 1×10−7 M or less. In other embodiments, the antibody binds with a KD of 1×10−8 M, 1×10−9 M, 1×10−10 M, 1×10−11 M, 1×10−12M or less. In certain embodiments, the KD is 1 μM to 500 pM, between 500 pM to 1 μM, between 1 μM to 100 nM, or between 100 mM to 10 nM.


Antibodies of the invention can be derived from any species of animal, preferably a mammal. Non-limiting exemplary natural antibodies include antibodies derived from human, chicken, goats, and rodents (e.g., rats, mice, hamsters and rabbits), including transgenic rodents genetically engineered to produce human antibodies (see, e.g., Lonberg et al., WO93/12227; U.S. Pat. No. 5,545,806; and Kucherlapati, et al., WO91/10741; U.S. Pat. No. 6,150,584, which are herein incorporated by reference in their entirety). Natural antibodies are the antibodies produced by a host animal. “Genetically altered antibodies” refer to antibodies wherein the amino acid sequence has been varied from that of a native antibody. Because of the relevance of recombinant DNA techniques to this application, one need not be confined to the sequences of amino acids found in natural antibodies; antibodies can be redesigned to obtain desired characteristics. The possible variations are many and range from the changing of just one or a few amino acids to the complete redesign of, for example, the variable or constant region. Changes in the constant region will, in general, be made in order to improve or alter characteristics, such as complement fixation, interaction with membranes and other effector functions. Changes in the variable region will be made in order to improve the antigen binding characteristics.


The antibodies of the invention include antibodies of any isotype including IgM, IgG, IgD, IgA and IgE, and any sub-isotype, including IgG1, IgG2a, IgG2b, IgG3 and IgG4, IgE1, IgE2 etc. The light chains of the antibodies can either be kappa light chains or lambda light chains.


Antibodies disclosed in the invention may be polyclonal or monoclonal. As used herein, the term “epitope” refers to the smallest portion of a protein capable of selectively binding to the antigen binding site of an antibody. It is well accepted by those skilled in the art that the minimal size of a protein epitope capable of selectively binding to the antigen binding site of an antibody is about five or six to seven amino acids.


Other antibodies specifically contemplated are oligoclonal antibodies. As used herein, the phrase “oligoclonal antibodies” refers to a predetermined mixture of distinct monoclonal antibodies. See, e.g., PCT publication WO 95/20401; U.S. Pat. Nos. 5,789,208 and 6,335,163. In one embodiment, oligoclonal antibodies consisting of a predetermined mixture of antibodies against one or more epitopes are generated in a single cell. In other embodiments, oligoclonal antibodies comprise a plurality of heavy chains capable of pairing with a common light chain to generate antibodies with multiple specificities (e.g., PCT publication WO 04/009618). Oligoclonal antibodies are particularly useful when it is desired to target multiple epitopes on a single target molecule. In view of the assays and epitopes disclosed herein, those skilled in the art can generate or select antibodies or mixtures of antibodies that are applicable for an intended purpose and desired need.


Recombinant antibodies against the phosphorylation sites identified in the invention are also included in the present application. These recombinant antibodies have the same amino acid sequence as the natural antibodies or have altered amino acid sequences of the natural antibodies in the present application. They can be made in any expression systems including both prokaryotic and eukaryotic expression systems or using phage display methods (see, e.g., Dower et al., WO91/17271 and McCafferty et al., WO92/01047; U.S. Pat. No. 5,969,108, which are herein incorporated by reference in their entirety).


Antibodies can be engineered in numerous ways. They can be made as single-chain antibodies (including small modular immunopharmaceuticals or SMIPs™), Fab and F(ab′)2 fragments, etc. Antibodies can be humanized, chimerized, deimmunized, or fully human. Numerous publications set forth the many types of antibodies and the methods of engineering such antibodies. For example, see U.S. Pat. Nos. 6,355,245; 6,180,370; 5,693,762; 6,407,213; 6,548,640; 5,565,332; 5,225,539; 6,103,889; and 5,260,203.


The genetically altered antibodies should be functionally equivalent to the above-mentioned natural antibodies. In certain embodiments, modified antibodies provide improved stability or/and therapeutic efficacy. Examples of modified antibodies include those with conservative substitutions of amino acid residues, and one or more deletions or additions of amino acids that do not significantly deleteriously alter the antigen binding utility. Substitutions can range from changing or modifying one or more amino acid residues to complete redesign of a region as long as the therapeutic utility is maintained. Antibodies of this application can be modified post-translationally (e.g., acetylation, and/or phosphorylation) or can be modified synthetically (e.g., the attachment of a labeling group).


Antibodies with engineered or variant constant or Fc regions can be useful in modulating effector functions, such as, for example, antigen-dependent cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Such antibodies with engineered or variant constant or Fc regions may be useful in instances where a parent singling protein (Table 1) is expressed in normal tissue; variant antibodies without effector function in these instances may elicit the desired therapeutic response while not damaging normal tissue. Accordingly, certain aspects and methods of the present disclosure relate to antibodies with altered effector functions that comprise one or more amino acid substitutions, insertions, and/or deletions.


In certain embodiments, genetically altered antibodies are chimeric antibodies and humanized antibodies.


The chimeric antibody is an antibody having portions derived from different antibodies. For example, a chimeric antibody may have a variable region and a constant region derived from two different antibodies. The donor antibodies may be from different species. In certain embodiments, the variable region of a chimeric antibody is non-human, e.g., murine, and the constant region is human.


The genetically altered antibodies used in the invention include CDR grafted humanized antibodies. In one embodiment, the humanized antibody comprises heavy and/or light chain CDRs of a non-human donor immunoglobulin and heavy chain and light chain frameworks and constant regions of a human acceptor immunoglobulin. The method of making humanized antibody is disclosed in U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,761; 5,693,762; and 6,180,370 each of which is incorporated herein by reference in its entirety.


Antigen-binding fragments of the antibodies of the invention, which retain the binding specificity of the intact antibody, are also included in the invention. Examples of these antigen-binding fragments include, but are not limited to, partial or full heavy chains or light chains, variable regions, or CDR regions of any phosphorylation site-specific antibodies described herein.


In one embodiment of the application, the antibody fragments are truncated chains (truncated at the carboxyl end). In certain embodiments, these truncated chains possess one or more immunoglobulin activities (e.g., complement fixation activity). Examples of truncated chains include, but are not limited to, Fab fragments (consisting of the VL, VH, CL and CH1 domains); Fd fragments (consisting of the VH and CH1 domains); Fv fragments (consisting of VL and VH domains of a single chain of an antibody); dAb fragments (consisting of a VH domain); isolated CDR regions; (Fab′)2 fragments, bivalent fragments (comprising two Fab fragments linked by a disulphide bridge at the hinge region). The truncated chains can be produced by conventional biochemical techniques, such as enzyme cleavage, or recombinant DNA techniques, each of which is known in the art. These polypeptide fragments may be produced by proteolytic cleavage of intact antibodies by methods well known in the art, or by inserting stop codons at the desired locations in the vectors using site-directed mutagenesis, such as after CH1 to produce Fab fragments or after the hinge region to produce (Fab′)2 fragments. Single chain antibodies may be produced by joining VL- and VH-coding regions with a DNA that encodes a peptide linker connecting the VL and VH protein fragments


Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment of an antibody yields an F(ab′)2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen.


“Fv” usually refers to the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising three CDRs specific for an antigen) has the ability to recognize and bind antigen, although likely at a lower affinity than the entire binding site.


Thus, in certain embodiments, the antibodies of the application may comprise 1, 2, 3, 4, 5, 6, or more CDRs that recognize the phosphorylation sites identified in Column E of Table 1.


The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. Fab′ fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab′)2 antibody fragments originally were produced as pairs of Fab′ fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.


“Single-chain Fv” or “scFv” antibody fragments comprise the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain. In certain embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that enables the scFv to form the desired structure for antigen binding. For a review of scFv see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore, eds. (Springer-Verlag: New York, 1994), pp. 269-315.


SMIPs are a class of single-chain peptides engineered to include a target binding region and effector domain (CH2 and CH3 domains). See, e.g., U.S. Patent Application Publication No. 20050238646. The target binding region may be derived from the variable region or CDRs of an antibody, e.g., a phosphorylation site-specific antibody of the application. Alternatively, the target binding region is derived from a protein that binds a phosphorylation site.


Bispecific antibodies may be monoclonal, human or humanized antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for the phosphorylation site, the other one is for any other antigen, such as for example, a cell-surface protein or receptor or receptor subunit. Alternatively, a therapeutic agent may be placed on one arm. The therapeutic agent can be a drug, toxin, enzyme, DNA, radionuclide, etc.


In some embodiments, the antigen-binding fragment can be a diabody. The term “diabody” refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90: 6444-6448 (1993).


Camelid antibodies refer to a unique type of antibodies that are devoid of light chain, initially discovered from animals of the camelid family. The heavy chains of these so-called heavy-chain antibodies bind their antigen by one single domain, the variable domain of the heavy immunoglobulin chain, referred to as VHH. VHHs show homology with the variable domain of heavy chains of the human VHIII family. The VHHs obtained from an immunized camel, dromedary, or llama have a number of advantages, such as effective production in microorganisms such as Saccharomyces cerevisiae.


In certain embodiments, single chain antibodies, and chimeric, humanized or primatized (CDR-grafted) antibodies, as well as chimeric or CDR-grafted single chain antibodies, comprising portions derived from different species, are also encompassed by the present disclosure as antigen-binding fragments of an antibody. The various portions of these antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques. For example, nucleic acids encoding a chimeric or humanized chain can be expressed to produce a contiguous protein. See, e.g., U.S. Pat. Nos. 4,816,567 and 6,331,415; U.S. Pat. No. 4,816,397; European Patent No. 0,120,694; WO 86/01533; European Patent No. 0,194,276 B1; U.S. Pat. No. 5,225,539; and European Patent No. 0,239,400 B1. See also, Newman et al., BioTechnology, 10: 1455-1460 (1992), regarding primatized antibody. See, e.g., Ladner et al., U.S. Pat. No. 4,946,778; and Bird et al., Science, 242: 423-426 (1988)), regarding single chain antibodies.


In addition, functional fragments of antibodies, including fragments of chimeric, humanized, primatized or single chain antibodies, can also be produced. Functional fragments of the subject antibodies retain at least one binding function and/or modulation function of the full-length antibody from which they are derived.


Since the immunoglobulin-related genes contain separate functional regions, each having one or more distinct biological activities, the genes of the antibody fragments may be fused to functional regions from other genes (e.g., enzymes, U.S. Pat. No. 5,004,692, which is incorporated by reference in its entirety) to produce fusion proteins or conjugates having novel properties.


Non-immunoglobulin binding polypeptides are also contemplated. For example, CDRs from an antibody disclosed herein may be inserted into a suitable non-immunoglobulin scaffold to create a non-immunoglobulin binding polypeptide. Suitable candidate scaffold structures may be derived from, for example, members of fibronectin type III and cadherin superfamilies.


Also contemplated are other equivalent non-antibody molecules, such as protein binding domains or aptamers, which bind, in a phospho-specific manner, to an amino acid sequence comprising a novel phosphorylation site of the invention. See, e.g., Neuberger et al., Nature 312: 604 (1984). Aptamers are oligonucleic acid or peptide molecules that bind a specific target molecule. DNA or RNA aptamers are typically short oligonucleotides, engineered through repeated rounds of selection to bind to a molecular target. Peptide aptamers typically consist of a variable peptide loop attached at both ends to a protein scaffold. This double structural constraint generally increases the binding affinity of the peptide aptamer to levels comparable to an antibody (nanomolar range).


The invention also discloses the use of the phosphorylation site-specific antibodies with immunotoxins. Conjugates that are immunotoxins including antibodies have been widely described in the art. The toxins may be coupled to the antibodies by conventional coupling techniques or immunotoxins containing protein toxin portions can be produced as fusion proteins. In certain embodiments, antibody conjugates may comprise stable linkers and may release cytotoxic agents inside cells (see U.S. Pat. Nos. 6,867,007 and 6,884,869). The conjugates of the present application can be used in a corresponding way to obtain such immunotoxins. Illustrative of such immunotoxins are those described by Byers et al., Seminars Cell Biol 2:59-70 (1991) and by Fanger et al., Immunol Today 12:51-54 (1991). Exemplary immunotoxins include radiotherapeutic agents, ribosome-inactivating proteins (RIPs), chemotherapeutic agents, toxic peptides, or toxic proteins.


The phosphorylation site-specific antibodies disclosed in the invention may be used singly or in combination. The antibodies may also be used in an array format for high throughput uses. An antibody microarray is a collection of immobolized antibodies, typically spotted and fixed on a solid surface (such as glass, plastic and silicon chip).


In another aspect, the antibodies of the invention modulate at least one, or all, biological activities of a parent protein identified in Column A of Table 1. The biological activities of a parent protein identified in Column A of Table 1 include: 1) ligand binding activities (for instance, these neutralizing antibodies may be capable of competing with or completely blocking the binding of a parent signaling protein to at least one, or all, of its ligands; 2) signaling transduction activities, such as receptor dimerization, or tyrosine phosphorylation; and 3) cellular responses induced by a parent signaling protein, such as oncogenic activities (e.g., cancer cell proliferation mediated by a parent signaling protein), and/or angiogenic activities.


In certain embodiments, the antibodies of the invention may have at least one activity selected from the group consisting of: 1) inhibiting cancer cell growth or proliferation; 2) inhibiting cancer cell survival; 3) inhibiting angiogenesis; 4) inhibiting cancer cell metastasis, adhesion, migration or invasion; 5) inducing apoptosis of cancer cells; 6) incorporating a toxic conjugate; and 7) acting as a diagnostic marker.


In certain embodiments, the phosphorylation site specific antibodies disclosed in the invention are especially indicated for diagnostic and therapeutic applications as described herein. Accordingly, the antibodies may be used in therapies, including combination therapies, in the diagnosis and prognosis of disease, as well as in the monitoring of disease progression. The invention, thus, further includes compositions comprising one or more embodiments of an antibody or an antigen binding portion of the invention as described herein. The composition may further comprise a pharmaceutically acceptable carrier. The composition may comprise two or more antibodies or antigen-binding portions, each with specificity for a different novel tyrosine phosphorylation site of the invention or two or more different antibodies or antigen-binding portions all of which are specific for the same novel tyrosine phosphorylation site of the invention. A composition of the invention may comprise one or more antibodies or antigen-binding portions of the invention and one or more additional reagents, diagnostic agents or therapeutic agents.


The present application provides for the polynucleotide molecules encoding the antibodies and antibody fragments and their analogs described herein. Because of the degeneracy of the genetic code, a variety of nucleic acid sequences encode each antibody amino acid sequence. The desired nucleic acid sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the desired polynucleotide. In one embodiment, the codons that are used comprise those that are typical for human or mouse (see, e.g., Nakamura, Y., Nucleic Acids Res. 28: 292 (2000)).


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 targeted signaling protein phosphorylation sties 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.)


5. Methods of Making Phosphorylation Site-Specific Antibodies


In another aspect, the invention provides a method for making phosphorylation site-specific antibodies.


Polyclonal antibodies of the invention may be produced according to standard techniques by immunizing a suitable animal (e.g., rabbit, goat, etc.) with an antigen comprising a novel tyrosine phosphorylation site of the invention. (i.e. a phosphorylation site shown in Table 1) in either the phosphorylated or unphosphorylated state, depending upon the desired specificity of the antibody, collecting immune serum from the animal, and separating the polyclonal antibodies from the immune serum, in accordance with known procedures and screening and isolating a polyclonal antibody specific for the novel tyro sine phosphorylation site of interest as further described below. Methods for immunizing non-human animals such as mice, rats, sheep, goats, pigs, cattle and horses are well known in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Press, 1990.


The immunogen may be the full length protein or a peptide comprising the novel tyro sine phosphorylation site of interest. In some embodiments the immunogen is a peptide of from 7 to 20 amino acids in length, preferably about 8 to 17 amino acids in length. In some embodiments, the peptide antigen desirably will comprise about 3 to 8 amino acids on each side of the phosphorylatable tyrosine. In yet other embodiments, the peptide antigen desirably will comprise four or more amino acids flanking each side of the phosphorylatable amino acid and encompassing it. 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)).


Suitable peptide antigens may comprise all or partial sequence of a trypsin-digested fragment as set forth in Column E of Table 1/FIG. 2. Suitable peptide antigens may also comprise all or partial sequence of a peptide fragment produced by another protease digestion.


Preferred immunogens are those that comprise a novel phosphorylation site of a protein in Table 1 that is a adaptor/scaffold proteins; adhesion or extracellular matrix proteins; cytoskeletal proteins; enzyme proteins; G proteins or regulator proteins; non-protein kinase proteins; motor or contractile proteins; phosphatase proteins; protein kinases; receptor, channel, transporter or cell surface proteins; RNA processing proteins; and transcriptional regulator proteins. In some embodiments, the peptide immunogen is an AQUA peptide, for example, any one of SEQ ID NOS: 1-19, 21-22, 25-27, 29-136, 138-144, 147-204, 206-232, 234-257, 259-277, 279-301, 303-374, 376-393, 395-403, 405-435, 437-447, 449-461, 463-500.


Particularly preferred immunogens are peptides comprising any one of the novel tyrosine phosphorylation site shown as a lower case “y” in a sequence listed in Table 1 selected from the group consisting of SEQ ID NOS: 320 (DDX1), 322 (DDX3), 323 (DDX3), 327 (DDX3Y), 154 (FBPase), 155 (G6PI), 200 (PIP5K), 222 (CD45), 223 (DARPP-32), 225 (FBP2), 238 (Cdc2), 239 (CDK9), 240 (CK2-alpha1), 247 (RSK2), 250 (RSK4), 251 (SRPK1), 266 (FGFR1), 268 (FGFR2), 275 (FLT3), 277 (Met), 35 (PSD-95), 97 (CAPZA2), 103 (cofilin 1), 286 (Cx43), 164 (Cdc42), 63 (SEMA4F), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 68 (CALB2), 73 (FREQ), 84 (DNAJB6), 93 (HMGB1), 97 (CAPZA2), 103 (cofilin 1), 164 (Cdc42), 215 (MYH4), 217 (MYH7), 221 (MYO1F), 278 (PDGFRb), 286 (Cx43), 491 (CLTC), and 499 (NSF).


In some embodiments the immunogen is administered with an adjuvant. Suitable adjuvants will be well known to those of skill in the art. Exemplary adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes).


For example, a peptide antigen comprising the novel adaptor/scaffold protein phosphorylation site in SEQ ID NO: 4 shown by the lower case “y” in Table 1 may be used to produce antibodies that specifically bind the novel tyrosine phosphorylation site.


When the above-described methods are used for producing polyclonal antibodies, following immunization, the polyclonal antibodies which secreted into the bloodstream can be recovered using known techniques. Purified forms of these antibodies can, of course, be readily prepared by standard purification techniques, such as for example, affinity chromatography with Protein A, anti-immunoglobulin, or the antigen itself. In any case, in order to monitor the success of immunization, the antibody levels with respect to the antigen in serum will be monitored using standard techniques such as ELISA, RIA and the like.


Monoclonal antibodies of the invention may be produced by any of a number of means that are well-known in the art. In some embodiments, antibody-producing B cells are isolated from an animal immunized with a peptide antigen as described above. The B cells may be from the spleen, lymph nodes or peripheral blood. Individual B cells are isolated and screened as described below to identify cells producing an antibody specific for the novel tyrosine phosphorylation site of interest. Identified cells are then cultured to produce a monoclonal antibody of the invention.


Alternatively, a monoclonal phosphorylation site-specific antibody of the invention may be produced using standard hybridoma technology, 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 any of a number of standard means. Methods of immortalizing cells include, but are not limited to, transfecting them with oncogenes, infecting them with an oncogenic virus and cultivating them under conditions that select for immortalized cells, subjecting them to carcinogenic or mutating compounds, fusing them with an immortalized cell, e.g., a myeloma cell, and inactivating a tumor suppressor gene. See, e.g., Harlow and Lane, supra. If fusion with myeloma cells is used, the myeloma cells preferably do not secrete immunoglobulin polypeptides (a non-secretory cell line). Typically the antibody producing cell and the immortalized cell (such as but not limited to myeloma cells) with which it is fused are from the same species. Rabbit fusion hybridomas, for example, may be produced as described in U.S. Pat. No. 5,675,063, C. Knight, Issued Oct. 7, 1997. The immortalized antibody producing cells, such as 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.


The invention also encompasses antibody-producing cells and cell lines, such as hybridomas, as described above.


Polyclonal or monoclonal antibodies may also be obtained through in vitro immunization. For example, phage display techniques can be used to provide libraries containing a repertoire of antibodies with varying affinities for a particular antigen. Techniques for the identification of high affinity human antibodies from such libraries are described by Griffiths et al., (1994) EMBO J., 13:3245-3260; Nissim et al., ibid, pp. 692-698 and by Griffiths et al., ibid, 12:725-734, which are incorporated by reference.


The antibodies may be produced recombinantly using methods well known in the art for example, according to the methods disclosed in U.S. Pat. No. 4,349,893 (Reading) or U.S. Pat. No. 4,816,567 (Cabilly et al.) The antibodies may also be chemically constructed by specific antibodies made according to the method disclosed in U.S. Pat. No. 4,676,980 (Segel et al.)


Once a desired phosphorylation site-specific antibody is identified, polynucleotides encoding the antibody, such as heavy, light chains or both (or single chains in the case of a single chain antibody) or portions thereof such as those encoding the variable region, may be cloned and isolated from antibody-producing cells using means that are well known in the art. 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.)


Accordingly, in a further aspect, the invention provides such nucleic acids encoding the heavy chain, the light chain, a variable region, a framework region or a CDR of an antibody of the invention. In some embodiments, the nucleic acids are operably linked to expression control sequences. The invention, thus, also provides vectors and expression control sequences useful for the recombinant expression of an antibody or antigen-binding portion thereof of the invention. Those of skill in the art will be able to choose vectors and expression systems that are suitable for the host cell in which the antibody or antigen-binding portion is to be expressed.


Monoclonal antibodies of the invention may be produced recombinantly by expressing the encoding nucleic acids in a suitable host cell under suitable conditions. Accordingly, the invention further provides host cells comprising the nucleic acids and vectors described above.


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 a single desired isotype are preferred for a particular application, particular isotypes can be prepared directly, by selecting from the initial fusion, or prepared secondarily, from a parental hybridoma secreting a monoclonal antibody of different isotype by using the sib selection technique to isolate class-switch variants (Steplewski, et al., Proc. Nat'l. Acad. Sci., 82: 8653 (1985); Spira et al., J. Immunol. Methods, 74: 307 (1984)). Alternatively, the isotype of a monoclonal antibody with desirable propertied can be changed using antibody engineering techniques that are well-known in the art.


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 phosphorylated and/or unphosphosphorylated peptide library by ELISA to ensure specificity for both the desired antigen (i.e. that epitope including a phosphorylation site of the invention and for reactivity only with the phosphorylated (or unphosphorylated) form of the antigen. Peptide competition assays may be carried out to confirm lack of reactivity with other phospho-epitopes on the parent protein. The antibodies may also be tested by Western blotting against cell preparations containing the parent signaling protein, e.g., cell lines over-expressing the parent protein, to confirm reactivity with the desired phosphorylated epitope/target.


Specificity against the desired phosphorylated epitope may also be examined by constructing mutants lacking phosphorylatable residues at positions outside the desired epitope that are known to be phosphorylated, or by mutating the desired phospho-epitope and confirming lack of reactivity. Phosphorylation site-specific antibodies of the invention may exhibit some limited cross-reactivity to related epitopes in non-target proteins. This is not unexpected as most antibodies exhibit some degree of cross-reactivity, and anti-peptide antibodies will often cross-react with epitopes having high homology to the immunizing peptide. See, e.g., Czernik, supra. Cross-reactivity with non-target proteins is readily characterized by Western blotting alongside markers of known molecular weight. Amino acid sequences of cross-reacting proteins may be examined to identify phosphorylation sites with flanking sequences that are highly homologous to that of a phosphorylation site of the invention.


In certain cases, polyclonal antisera may exhibit some undesirable general cross-reactivity to phosphotyrosine itself, which may be removed by further purification of antisera, e.g., over a phosphotyramine column. Antibodies of the invention specifically bind their target protein (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 examine phosphorylation and activation state and level of a phosphorylation site 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 lysed erythrocytes and cell debris. Adherring cells may be scrapped off plates and washed with PBS. 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 parent signaling protein enumerated in Table 1), washed and labeled with a fluorescent-labeled secondary antibody. Additional fluorochrome-conjugated marker antibodies (e.g., CD45, CD34) may also be added at this time to aid in the subsequent identification of specific hematopoietic cell types. The cells would then be analyzed on a flow cytometer (e.g. a Beckman Coulter FC500) according to the specific protocols of the instrument used.


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 may specifically bind to a signaling protein or polypeptide listed in Table 1 only when phosphorylated at the specified tyrosine residue, but are not limited only to binding to the listed signaling proteins of human species, per se. The invention includes antibodies that also bind conserved and highly homologous or identical phosphorylation sites in respective signaling proteins from other species (e.g., mouse, rat, monkey, yeast), in addition to binding the phosphorylation site of the human homologue. The term “homologous” refers to two or more sequences or subsequences that have at least about 85%, at least 90%, at least 95%, or higher nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using sequence comparison method (e.g., BLAST) and/or by visual inspection. Highly homologous or identical sites conserved in other species can readily be identified by standard sequence comparisons (such as BLAST).


Methods for making bispecific antibodies are within the purview of those skilled in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature, 305:537-539 (1983)). Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. In certain embodiments, the fusion is with an immunoglobulin heavy-chain constant domain, including at least part of the hinge, CH2, and CH3 regions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of illustrative currently known methods for generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986); WO 96/27011; Brennan et al., Science 229:81 (1985); Shalaby et al., J. Exp. Med. 175:217-225 (1992); Kostelny et al., J. Immunol. 148(5):1547-1553 (1992); Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993); Gruber et al., J. Immunol. 152:5368 (1994); and Tutt et al., J. Immunol. 147:60 (1991). Bispecific antibodies also include cross-linked or heteroconjugate antibodies. Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.


Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol., 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins may be linked to the Fab′ portions of two different antibodies by gene fusion. The antibody homodimers may be reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. A strategy for making bispecific antibody fragments by the use of single-chain Fv (scFv) dimers has also been reported. See Gruber et al., J. Immunol., 152:5368 (1994). Alternatively, the antibodies can be “linear antibodies” as described in Zapata et al. Protein Eng. 8(10):1057-1062 (1995). Briefly, these antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-CH1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.


To produce the chimeric antibodies, the portions derived from two different species (e.g., human constant region and murine variable or binding region) can be joined together chemically by conventional techniques or can be prepared as single contiguous proteins using genetic engineering techniques. The DNA molecules encoding the proteins of both the light chain and heavy chain portions of the chimeric antibody can be expressed as contiguous proteins. The method of making chimeric antibodies is disclosed in U.S. Pat. No. 5,677,427; U.S. Pat. No. 6,120,767; and U.S. Pat. No. 6,329,508, each of which is incorporated by reference in its entirety.


Fully human antibodies may be produced by a variety of techniques. One example is trioma methodology. The basic approach and an exemplary cell fusion partner, SPAZ-4, for use in this approach have been described by Oestberg et al., Hybridoma 2:361-367 (1983); Oestberg, U.S. Pat. No. 4,634,664; and Engleman et al., U.S. Pat. No. 4,634,666 (each of which is incorporated by reference in its entirety).


Human antibodies can also be produced from non-human transgenic animals having transgenes encoding at least a segment of the human immunoglobulin locus. The production and properties of animals having these properties are described in detail by, see, e.g., Lonberg et al., WO93/12227; U.S. Pat. No. 5,545,806; and Kucherlapati, et al., WO91/10741; U.S. Pat. No. 6,150,584, which are herein incorporated by reference in their entirety.


Various recombinant antibody library technologies may also be utilized to produce fully human antibodies. For example, one approach is to screen a DNA library from human B cells according to the general protocol outlined by Huse et al., Science 246:1275-1281 (1989). The protocol described by Huse is rendered more efficient in combination with phage-display technology. See, e.g., Dower et al., WO 91/17271 and McCafferty et al., WO 92/01047; U.S. Pat. No. 5,969,108, (each of which is incorporated by reference in its entirety).


Eukaryotic ribosome can also be used as means to display a library of antibodies and isolate the binding human antibodies by screening against the target antigen, as described in Coia G, et al., J. Immunol. Methods 1: 254 (1-2):191-7 (2001); Hanes J. et al., Nat. Biotechnol. 18(12):1287-92 (2000); Proc. Natl. Acad. Sci. U.S.A. 95(24):14130-5 (1998); Proc. Natl. Acad. Sci. U.S.A. 94(10):4937-42 (1997), each which is incorporated by reference in its entirety.


The yeast system is also suitable for screening mammalian cell-surface or secreted proteins, such as antibodies. Antibody libraries may be displayed on the surface of yeast cells for the purpose of obtaining the human antibodies against a target antigen. This approach is described by Yeung, et al., Biotechnol. Prog. 18(2):212-20 (2002); Boeder, E. T., et al., Nat. Biotechnol. 15(6):553-7 (1997), each of which is herein incorporated by reference in its entirety. Alternatively, human antibody libraries may be expressed intracellularly and screened via the yeast two-hybrid system (WO0200729A2, which is incorporated by reference in its entirety).


Recombinant DNA techniques can be used to produce the recombinant phosphorylation site-specific antibodies described herein, as well as the chimeric or humanized phosphorylation site-specific antibodies, or any other genetically-altered antibodies and the fragments or conjugate thereof in any expression systems including both prokaryotic and eukaryotic expression systems, such as bacteria, yeast, insect cells, plant cells, mammalian cells (for example, NS0 cells).


Once produced, the whole antibodies, their dimers, individual light and heavy chains, or other immunoglobulin forms of the present application can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (see, generally, Scopes, R., Protein Purification (Springer-Verlag, N.Y., 1982)). Once purified, partially or to homogeneity as desired, the polypeptides may then be used therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent staining, and the like. (See, generally, Immunological Methods, Vols. I and II (Lefkovits and Pernis, eds., Academic Press, NY, 1979 and 1981).


6. Therapeutic Uses


In a further aspect, the invention provides methods and compositions for therapeutic uses of the peptides or proteins comprising a phosphorylation site of the invention, and phosphorylation site-specific antibodies of the invention.


In one embodiment, the invention provides for a method of treating or preventing carcinoma and/or leukemia in a subject, wherein the carcinoma and/or leukemia is associated with the phosphorylation state of a novel phosphorylation site in Table 1, whether phosphorylated or dephosphorylated, comprising: administering to a subject in need thereof a therapeutically effective amount of a peptide comprising a novel phosphorylation site (Table 1) and/or an antibody or antigen-binding fragment thereof that specifically bind a novel phosphorylation site of the invention (Table 1). The antibodies may be full-length antibodies, genetically engineered antibodies, antibody fragments, and antibody conjugates of the invention.


The term “subject” refers to a vertebrate, such as for example, a mammal, or a human. Although present application are primarily concerned with the treatment of human subjects, the disclosed methods may also be used for the treatment of other mammalian subjects such as dogs and cats for veterinary purposes.


In one aspect, the disclosure provides a method of treating carcinoma and/or leukemia in which a peptide or an antibody that reduces at least one biological activity of a targeted signaling protein is administered to a subject. For example, the peptide or the antibody administered may disrupt or modulate the interaction of the target signaling protein with its ligand. Alternatively, the peptide or the antibody may interfere with, thereby reducing, the down-stream signal transduction of the parent signaling protein. An antibody that specifically binds the novel tyrosine phosphorylation site only when the tyrosine is phosphorylated, and that does not substantially bind to the same sequence when the tyrosine is not phosphorylated, thereby prevents downstream signal transduction triggered by a phospho-tyrosine. Alternatively, an antibody that specifically binds the unphosphorylated target phosphorylation site reduces the phosphorylation at that site and thus reduces activation of the protein mediated by phosphorylation of that site. Similarly, an unphosphorylated peptide may compete with an endogenous phosphorylation site for same kinases, thereby preventing or reducing the phosphorylation of the endogenous target protein. Alternatively, a peptide comprising a phosphorylated novel tyrosine site of the invention but lacking the ability to trigger signal transduction may competitively inhibit interaction of the endogenous protein with the same down-stream ligand(s).


The antibodies of the invention may also be used to target cancer cells for effector-mediated cell death. The antibody disclosed herein may be administered as a fusion molecule that includes a phosphorylation site-targeting portion joined to a cytotoxic moiety to directly kill cancer cells. Alternatively, the antibody may directly kill the cancer cells through complement-mediated or antibody-dependent cellular cytotoxicity.


Accordingly in one embodiment, the antibodies of the present disclosure may be used to deliver a variety of cytotoxic compounds. Any cytotoxic compound can be fused to the present antibodies. The fusion can be achieved chemically or genetically (e.g., via expression as a single, fused molecule). The cytotoxic compound can be a biological, such as a polypeptide, or a small molecule. As those skilled in the art will appreciate, for small molecules, chemical fusion is used, while for biological compounds, either chemical or genetic fusion can be used.


Non-limiting examples of cytotoxic compounds include therapeutic drugs, radiotherapeutic agents, ribosome-inactivating proteins (RIPs), chemotherapeutic agents, toxic peptides, toxic proteins, and mixtures thereof. The cytotoxic drugs can be intracellularly acting cytotoxic drugs, such as short-range radiation emitters, including, for example, short-range, high-energy α-emitters. Enzymatically active toxins and fragments thereof, including ribosome-inactivating proteins, are exemplified by saporin, luffin, momordins, ricin, trichosanthin, gelonin, abrin, etc. Procedures for preparing enzymatically active polypeptides of the immunotoxins are described in WO84/03508 and WO85/03508, which are hereby incorporated by reference. Certain cytotoxic moieties are derived from adriamycin, chlorambucil, daunomycin, methotrexate, neocarzinostatin, and platinum, for example.


Exemplary chemotherapeutic agents that may be attached to an antibody or antigen-binding fragment thereof include taxol, doxorubicin, verapamil, podophyllotoxin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, bisulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, transplatinum, 5-fluorouracil, vincristin, vinblastin, or methotrexate.


Procedures for conjugating the antibodies with the cytotoxic agents have been previously described and are within the purview of one skilled in the art.


Alternatively, the antibody can be coupled to high energy radiation emitters, for example, a radioisotope, such as 131I, a γ-emitter, which, when localized at the tumor site, results in a killing of several cell diameters. See, e.g., S. E. Order, “Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody in Cancer Therapy”, Monoclonal Antibodies for Cancer Detection and Therapy, Baldwin et al. (eds.), pp. 303-316 (Academic Press 1985), which is hereby incorporated by reference. Other suitable radioisotopes include α-emitters, such as 212Bi, 213Bi, and 211At, and β-emitters, such as 186Re and 90Y.


Because many of the signaling proteins in which novel tyrosine phosphorylation sites of the invention occur also are expressed in normal cells and tissues, it may also be advantageous to administer a phosphorylation site-specific antibody with a constant region modified to reduce or eliminate ADCC or CDC to limit damage to normal cells. For example, effector function of an antibodies may be reduced or eliminated by utilizing an IgG1 constant domain instead of an IgG2/4 fusion domain. Other ways of eliminating effector function can be envisioned such as, e.g., mutation of the sites known to interact with FcR or insertion of a peptide in the hinge region, thereby eliminating critical sites required for FcR interaction. Variant antibodies with reduced or no effector function also include variants as described previously herein.


The peptides and antibodies of the invention may be used in combination with other therapies or with other agents. Other agents include but are not limited to polypeptides, small molecules, chemicals, metals, organometallic compounds, inorganic compounds, nucleic acid molecules, oligonucleotides, aptamers, spiegelmers, antisense nucleic acids, locked nucleic acid (LNA) inhibitors, peptide nucleic acid (PNA) inhibitors, immunomodulatory agents, antigen-binding fragments, prodrugs, and peptidomimetic compounds. In certain embodiments, the antibodies and peptides of the invention may be used in combination with cancer therapies known to one of skill in the art.


In certain aspects, the present disclosure relates to combination treatments comprising a phosphorylation site-specific antibody described herein and immunomodulatory compounds, vaccines or chemotherapy. Illustrative examples of suitable immunomodulatory agents that may be used in such combination therapies include agents that block negative regulation of T cells or antigen presenting cells (e.g., anti-CTLA4 antibodies, anti-PD-L1 antibodies, anti-PDL-2 antibodies, anti-PD-1 antibodies and the like) or agents that enhance positive co-stimulation of T cells (e.g., anti-CD40 antibodies or anti 4-1BB antibodies) or agents that increase NK cell number or T-cell activity (e.g., inhibitors such as IMiDs, thalidomide, or thalidomide analogs). Furthermore, immunomodulatory therapy could include cancer vaccines such as dendritic cells loaded with tumor cells, proteins, peptides, RNA, or DNA derived from such cells, patient derived heat-shock proteins (hsp's) or general adjuvants stimulating the immune system at various levels such as CpG, Luivac®, Biostim®, Ribomunyl®, Imudon®, Bronchovaxom® or any other compound or other adjuvant activating receptors of the innate immune system (e.g., toll like receptor agonist, anti-CTLA-4 antibodies, etc.). Also, immunomodulatory therapy could include treatment with cytokines such as IL-2, GM-CSF and IFN-gamma.


Furthermore, combination of antibody therapy with chemotherapeutics could be particularly useful to reduce overall tumor burden, to limit angiogenesis, to enhance tumor accessibility, to enhance susceptibility to ADCC, to result in increased immune function by providing more tumor antigen, or to increase the expression of the T cell attractant LIGHT.


Pharmaceutical compounds that may be used for combinatory anti-tumor therapy include, merely to illustrate: aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, camptothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gemcitabine, genistein, goserelin, hydroxyurea, idarubicin, ifosfamide, imatinib, interferon, irinotecan, letrozole, leucovorin, leuprolide, levamisole, lomustine, mechlorethamine, medroxyprogesterone, megestrol, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, nocodazole, octreotide, oxaliplatin, paclitaxel, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, suramin, tamoxifen, temozolomide, teniposide, testosterone, thioguanine, thiotepa, titanocene dichloride, topotecan, trastuzumab, tretinoin, vinblastine, vincristine, vindesine, and vinorelbine.


These chemotherapeutic anti-tumor compounds may be categorized by their mechanism of action into groups, including, for example, the following classes of agents: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate inhibitors and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristine, vinblastine, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecin, carboplatin, chlorambucil, cisplatin, cyclophosphamide, cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, hexamethylmelamineoxaliplatin, iphosphamide, melphalan, mechlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, taxol, taxotere, teniposide, triethylenethiophosphoramide and etoposide (VP16)); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmustine (BCNU) and analogs, streptozocin), trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel, abciximab; antimigratory agents; antisecretory agents (breveldin); immunosuppressives (cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine, mycophenolate mofetil); immunomodulatory agents (thalidomide and analogs thereof such as lenalidomide (Revlimid, CC-5013) and CC-4047 (Actimid)), cyclophosphamide; anti-angiogenic compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor (VEGF) inhibitors, fibroblast growth factor (FGF) inhibitors); angiotensin receptor blocker; nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab); cell cycle inhibitors and differentiation inducers (tretinoin); mTOR inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), amsacrine, camptothecin, daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin and mitoxantrone, topotecan, irinotecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers and caspase activators; and chromatin disruptors.


In certain embodiments, pharmaceutical compounds that may be used for combinatory anti-angiogenesis therapy include: (1) inhibitors of release of “angiogenic molecules,” such as bFGF (basic fibroblast growth factor); (2) neutralizers of angiogenic molecules, such as anti-βbFGF antibodies; and (3) inhibitors of endothelial cell response to angiogenic stimuli, including collagenase inhibitor, basement membrane turnover inhibitors, angiostatic steroids, fungal-derived angiogenesis inhibitors, platelet factor 4, thrombospondin, arthritis drugs such as D-penicillamine and gold thiomalate, vitamin D3 analogs, alpha-interferon, and the like. For additional proposed inhibitors of angiogenesis, see Blood et al., Biochim. Biophys. Acta, 1032:89-118 (1990), Moses et al., Science, 248:1408-1410 (1990), Ingber et al., Lab. Invest., 59:44-51 (1988), and U.S. Pat. Nos. 5,092,885, 5,112,946, 5,192,744, 5,202,352, and 6,573,256. In addition, there are a wide variety of compounds that can be used to inhibit angiogenesis, for example, peptides or agents that block the VEGF-mediated angiogenesis pathway, endostatin protein or derivatives, lysine binding fragments of angiostatin, melanin or melanin-promoting compounds, plasminogen fragments (e.g., Kringles 1-3 of plasminogen), troponin subunits, inhibitors of vitronectin αvβ3, peptides derived from Saposin B, antibiotics or analogs (e.g., tetracycline or neomycin), dienogest-containing compositions, compounds comprising a MetAP-2 inhibitory core coupled to a peptide, the compound EM-138, chalcone and its analogs, and naaladase inhibitors. See, for example, U.S. Pat. Nos. 6,395,718, 6,462,075, 6,465,431, 6,475,784, 6,482,802, 6,482,810, 6,500,431, 6,500,924, 6,518,298, 6,521,439, 6,525,019, 6,538,103, 6,544,758, 6,544,947, 6,548,477, 6,559,126, and 6,569,845.


7. Diagnostic Uses


In a further aspect, the invention provides methods for detecting and quantitating phosphorylation at a novel tyrosine phosphorylation site of the invention. For example, peptides, including AQUA peptides of the invention, and antibodies of the invention are useful in diagnostic and prognostic evaluation of carcinoma and/or leukemias, wherein the carcinoma and/or leukemia is associated with the phosphorylation state of a novel phosphorylation site in Table 1, whether phosphorylated or dephosphorylated.


Methods of diagnosis can be performed in vitro using a biological sample (e.g., blood sample, lymph node biopsy or tissue) from a subject, or in vivo. The phosphorylation state or level at the tyrosine residue identified in the corresponding row in Column D of Table 1 may be assessed. A change in the phosphorylation state or level at the phosphorylation site, as compared to a control, indicates that the subject is suffering from, or susceptible to, carcinoma and/or leukemia.


In one embodiment, the phosphorylation state or level at a novel phosphorylation site is determined by an AQUA peptide comprising the phosphorylation site. The AQUA peptide may be phosphorylated or unphosphorylated at the specified tyrosine position.


In another embodiment, the phosphorylation state or level at a phosphorylation site is determined by an antibody or antigen-binding fragment thereof, wherein the antibody specifically binds the phosphorylation site. The antibody may be one that only binds to the phosphorylation site when the tyrosine residue is phosphorylated, but does not bind to the same sequence when the tyrosine is not phosphorylated; or vice versa.


In particular embodiments, the antibodies of the present application are attached to labeling moieties, such as a detectable marker. One or more detectable labels can be attached to the antibodies. Exemplary labeling moieties include radiopaque dyes, radiocontrast agents, fluorescent molecules, spin-labeled molecules, enzymes, or other labeling moieties of diagnostic value, particularly in radiologic or magnetic resonance imaging techniques.


A radiolabeled antibody in accordance with this disclosure can be used for in vitro diagnostic tests. The specific activity of an antibody, binding portion thereof, probe, or ligand, depends upon the half-life, the isotopic purity of the radioactive label, and how the label is incorporated into the biological agent. In immunoassay tests, the higher the specific activity, in general, the better the sensitivity. Radioisotopes useful as labels, e.g., for use in diagnostics, include iodine (131I or 125I), indium (111In), technetium (99Tc), phosphorus (32P), carbon (14C), and tritium (3H), or one of the therapeutic isotopes listed above.


Fluorophore and chromophore labeled biological agents can be prepared from standard moieties known in the art. Since antibodies and other proteins absorb light having wavelengths up to about 310 nm, the fluorescent moieties may be selected to have substantial absorption at wavelengths above 310 nm, such as for example, above 400 nm. A variety of suitable fluorescers and chromophores are described by Stryer, Science, 162:526 (1968) and Brand et al., Annual Review of Biochemistry, 41:843-868 (1972), which are hereby incorporated by reference. The antibodies can be labeled with fluorescent chromophore groups by conventional procedures such as those disclosed in U.S. Pat. Nos. 3,940,475, 4,289,747, and 4,376,110, which are hereby incorporated by reference.


The control may be parallel samples providing a basis for comparison, for example, biological samples drawn from a healthy subject, or biological samples drawn from healthy tissues of the same subject. Alternatively, the control may be a pre-determined reference or threshold amount. If the subject is being treated with a therapeutic agent, and the progress of the treatment is monitored by detecting the tyrosine phosphorylation state level at a phosphorylation site of the invention, a control may be derived from biological samples drawn from the subject prior to, or during the course of the treatment.


In certain embodiments, antibody conjugates for diagnostic use in the present application are intended for use in vitro, where the antibody is linked to a secondary binding ligand or to an enzyme (an enzyme tag) that will generate a colored product upon contact with a chromogenic substrate. Examples of suitable enzymes include urease, alkaline phosphatase, (horseradish) hydrogen peroxidase and glucose oxidase. In certain embodiments, secondary binding ligands are biotin and avidin or streptavidin compounds.


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 in subjects before, during, and after treatment with a therapeutic agent targeted at inhibiting tyrosine phosphorylation 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 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).


Alternatively, antibodies of the invention may be used 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.


Peptides and 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 arrays 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 the phosphorylation state or level at two or more phosphorylation sites of the invention (Table 1) in a biological sample, the method comprising utilizing two or more antibodies or AQUA peptides of the invention. In one preferred embodiment, two to five antibodies or AQUA peptides of the invention are used. In another preferred embodiment, six to ten antibodies or AQUA peptides of the invention are used, while in another preferred embodiment eleven to twenty antibodies or AQUA peptides of the invention are used.


In certain embodiments the diagnostic methods of the application may be used in combination with other cancer diagnostic tests.


The biological sample analyzed may be any sample that is suspected of having abnormal tyrosine phosphorylation at a novel phosphorylation site of the invention, such as a homogenized neoplastic tissue sample.


8. Screening Assays


In another aspect, the invention provides a method for identifying an agent that modulates tyrosine phosphorylation at a novel phosphorylation site of the invention, comprising: a) contacting a candidate agent with a peptide or protein comprising a novel phosphorylation site of the invention; and b) determining the phosphorylation state or level at the novel phosphorylation site. A change in the phosphorylation level of the specified tyrosine in the presence of the test agent, as compared to a control, indicates that the candidate agent potentially modulates tyrosine phosphorylation at a novel phosphorylation site of the invention.


In one embodiment, the phosphorylation state or level at a novel phosphorylation site is determined by an AQUA peptide comprising the phosphorylation site. The AQUA peptide may be phosphorylated or unphosphorylated at the specified tyrosine position.


In another embodiment, the phosphorylation state or level at a phosphorylation site is determined by an antibody or antigen-binding fragment thereof, wherein the antibody specifically binds the phosphorylation site. The antibody may be one that only binds to the phosphorylation site when the tyrosine residue is phosphorylated, but does not bind to the same sequence when the tyrosine is not phosphorylated; or vice versa.


In particular embodiments, the antibodies of the present application are attached to labeling moieties, such as a detectable marker.


The control may be parallel samples providing a basis for comparison, for example, the phosphorylation level of the target protein or peptide in absence of the testing agent. Alternatively, the control may be a pre-determined reference or threshold amount.


9. Immunoassays


In another aspect, the present application concerns immunoassays for binding, purifying, quantifying and otherwise generally detecting the phosphorylation state or level at a novel phosphorylation site of the invention.


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 used 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 using 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.


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.


In certain embodiments, immunoassays are the various types of enzyme linked immunoadsorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. However, it will be readily appreciated that detection is not limited to such techniques, and Western blotting, dot and slot blotting, FACS analyses, and the like may also be used. The steps of various useful immunoassays have been described in the scientific literature, such as, e.g., Nakamura et al., in Enzyme Immunoassays: Heterogeneous and Homogeneous Systems, Chapter 27 (1987), incorporated herein by reference.


In general, the detection of immunocomplex formation is well known in the art and may be achieved through the application of numerous approaches. These methods are based upon the detection of radioactive, fluorescent, biological or enzymatic tags. Of course, one may find additional advantages through the use of a secondary binding ligand such as a second antibody or a biotin/avidin ligand binding arrangement, as is known in the art.


The antibody used in the detection may itself be conjugated to a detectable label, wherein one would then simply detect this label. The amount of the primary immune complexes in the composition would, thereby, be determined.


Alternatively, the first antibody that becomes bound within the primary immune complexes may be detected by means of a second binding ligand that has binding affinity for the antibody. In these cases, the second binding ligand may be linked to a detectable label. The second binding ligand is itself often an antibody, which may thus be termed a “secondary” antibody. The primary immune complexes are contacted with the labeled, secondary binding ligand, or antibody, under conditions effective and for a period of time sufficient to allow the formation of secondary immune complexes. The secondary immune complexes are washed extensively to remove any non-specifically bound labeled secondary antibodies or ligands, and the remaining label in the secondary immune complex is detected.


An enzyme linked immunoadsorbent assay (ELISA) is a type of binding assay. In one type of ELISA, phosphorylation site-specific antibodies disclosed herein are immobilized onto a selected surface exhibiting protein affinity, such as a well in a polystyrene microtiter plate. Then, a suspected neoplastic tissue sample is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound target signaling protein may be detected.


In another type of ELISA, the neoplastic tissue samples are immobilized onto the well surface and then contacted with the phosphorylation site-specific antibodies disclosed herein. After binding and washing to remove non-specifically bound immune complexes, the bound phosphorylation site-specific antibodies are detected.


Irrespective of the format used, ELISAs have certain features in common, such as coating, incubating or binding, washing to remove non-specifically bound species, and detecting the bound immune complexes.


The radioimmunoassay (RIA) is an analytical technique which depends on the competition (affinity) of an antigen for antigen-binding sites on antibody molecules. Standard curves are constructed from data gathered from a series of samples each containing the same known concentration of labeled antigen, and various, but known, concentrations of unlabeled antigen. Antigens are labeled with a radioactive isotope tracer. The mixture is incubated in contact with an antibody. Then the free antigen is separated from the antibody and the antigen bound thereto. Then, by use of a suitable detector, such as a gamma or beta radiation detector, the percent of either the bound or free labeled antigen or both is determined. This procedure is repeated for a number of samples containing various known concentrations of unlabeled antigens and the results are plotted as a standard graph. The percent of bound tracer antigens is plotted as a function of the antigen concentration. Typically, as the total antigen concentration increases the relative amount of the tracer antigen bound to the antibody decreases. After the standard graph is prepared, it is thereafter used to determine the concentration of antigen in samples undergoing analysis.


In an analysis, the sample in which the concentration of antigen is to be determined is mixed with a known amount of tracer antigen. Tracer antigen is the same antigen known to be in the sample but which has been labeled with a suitable radioactive isotope. The sample with tracer is then incubated in contact with the antibody. Then it can be counted in a suitable detector which counts the free antigen remaining in the sample. The antigen bound to the antibody or immunoadsorbent may also be similarly counted. Then, from the standard curve, the concentration of antigen in the original sample is determined.


10. Pharmaceutical Formulations and Methods of Administration


Methods of administration of therapeutic agents, particularly peptide and antibody therapeutics, are well-known to those of skill in the art.


Peptides of the invention can be administered in the same manner as conventional peptide type pharmaceuticals. Preferably, peptides are administered parenterally, for example, intravenously, intramuscularly, intraperitoneally, or subcutaneously. When administered orally, peptides may be proteolytically hydrolyzed. Therefore, oral application may not be usually effective. However, peptides can be administered orally as a formulation wherein peptides are not easily hydrolyzed in a digestive tract, such as liposome-microcapsules. Peptides may be also administered in suppositories, sublingual tablets, or intranasal spray.


If administered parenterally, a preferred pharmaceutical composition is an aqueous solution that, in addition to a peptide of the invention as an active ingredient, may contain for example, buffers such as phosphate, acetate, etc., osmotic pressure-adjusting agents such as sodium chloride, sucrose, and sorbitol, etc., antioxidative or antioxygenic agents, such as ascorbic acid or tocopherol and preservatives, such as antibiotics. The parenterally administered composition also may be a solution readily usable or in a lyophilized form which is dissolved in sterile water before administration.


The pharmaceutical formulations, dosage forms, and uses described below generally apply to antibody-based therapeutic agents, but are also useful and can be modified, where necessary, for making and using therapeutic agents of the disclosure that are not antibodies.


To achieve the desired therapeutic effect, the phosphorylation site-specific antibodies or antigen-binding fragments thereof can be administered in a variety of unit dosage forms. The dose will vary according to the particular antibody. For example, different antibodies may have different masses and/or affinities, and thus require different dosage levels. Antibodies prepared as Fab or other fragments will also require differing dosages than the equivalent intact immunoglobulins, as they are of considerably smaller mass than intact immunoglobulins, and thus require lower dosages to reach the same molar levels in the patient's blood. The dose will also vary depending on the manner of administration, the particular symptoms of the patient being treated, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician. Dosage levels of the antibodies for human subjects are generally between about 1 mg per kg and about 100 mg per kg per patient per treatment, such as for example, between about 5 mg per kg and about 50 mg per kg per patient per treatment. In terms of plasma concentrations, the antibody concentrations may be in the range from about 25 μg/mL to about 500 μg/mL. However, greater amounts may be required for extreme cases and smaller amounts may be sufficient for milder cases.


Administration of an antibody will generally be performed by a parenteral route, typically via injection such as intra-articular or intravascular injection (e.g., intravenous infusion) or intramuscular injection. Other routes of administration, e.g., oral (p.o.), may be used if desired and practicable for the particular antibody to be administered. An antibody can also be administered in a variety of unit dosage forms and their dosages will also vary with the size, potency, and in vivo half-life of the particular antibody being administered. Doses of a phosphorylation site-specific antibody will also vary depending on the manner of administration, the particular symptoms of the patient being treated, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician.


The frequency of administration may also be adjusted according to various parameters. These include the clinical response, the plasma half-life of the antibody, and the levels of the antibody in a body fluid, such as, blood, plasma, serum, or synovial fluid. To guide adjustment of the frequency of administration, levels of the antibody in the body fluid may be monitored during the course of treatment.


Formulations particularly useful for antibody-based therapeutic agents are also described in U.S. Patent App. Publication Nos. 20030202972, 20040091490 and 20050158316. In certain embodiments, the liquid formulations of the application are substantially free of surfactant and/or inorganic salts. In another specific embodiment, the liquid formulations have a pH ranging from about 5.0 to about 7.0. In yet another specific embodiment, the liquid formulations comprise histidine at a concentration ranging from about 1 mM to about 100 mM. In still another specific embodiment, the liquid formulations comprise histidine at a concentration ranging from 1 mM to 100 mM. It is also contemplated that the liquid formulations may further comprise one or more excipients such as a saccharide, an amino acid (e.g., arginine, lysine, and methionine) and a polyol. Additional descriptions and methods of preparing and analyzing liquid formulations can be found, for example, in PCT publications WO 03/106644, WO 04/066957, and WO 04/091658.


Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the pharmaceutical compositions of the application.


In certain embodiments, formulations of the subject antibodies are pyrogen-free formulations which are substantially free of endotoxins and/or related pyrogenic substances. Endotoxins include toxins that are confined inside microorganisms and are released when the microorganisms are broken down or die. Pyrogenic substances also include fever-inducing, thermostable substances (glycoproteins) from the outer membrane of bacteria and other microorganisms. Both of these substances can cause fever, hypotension and shock if administered to humans. Due to the potential harmful effects, it is advantageous to remove even low amounts of endotoxins from intravenously administered pharmaceutical drug solutions. The Food & Drug Administration (“FDA”) has set an upper limit of 5 endotoxin units (EU) per dose per kilogram body weight in a single one hour period for intravenous drug applications (The United States Pharmacopeial Convention, Pharmacopeial Forum 26 (1):223 (2000)). When therapeutic proteins are administered in amounts of several hundred or thousand milligrams per kilogram body weight, as can be the case with monoclonal antibodies, it is advantageous to remove even trace amounts of endotoxin.


The amount of the formulation which will be therapeutically effective can be determined by standard clinical techniques. In addition, in vitro assays may optionally be used to help identify optimal dosage ranges. The precise dose to be used in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. The dosage of the compositions to be administered can be determined by the skilled artisan without undue experimentation in conjunction with standard dose-response studies. Relevant circumstances to be considered in making those determinations include the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms. For example, the actual patient body weight may be used to calculate the dose of the formulations in milliliters (mL) to be administered. There may be no downward adjustment to “ideal” weight. In such a situation, an appropriate dose may be calculated by the following formula:

Dose(mL)=[patient weight(kg)×dose level(mg/kg)/drug concentration(mg/mL)]


For the purpose of treatment of disease, the appropriate dosage of the compounds (for example, antibodies) will depend on the severity and course of disease, the patient's clinical history and response, the toxicity of the antibodies, and the discretion of the attending physician. The initial candidate dosage may be administered to a patient. The proper dosage and treatment regimen can be established by monitoring the progress of therapy using conventional techniques known to those of skill in the art.


The formulations of the application can be distributed as articles of manufacture comprising packaging material and a pharmaceutical agent which comprises, e.g., the antibody and a pharmaceutically acceptable carrier as appropriate to the mode of administration. The packaging material will include a label which indicates that the formulation is for use in the treatment of prostate cancer.


11. Kits


Antibodies and peptides (including AQUA peptides) of the invention may also be used within a kit for detecting the phosphorylation state or level at a novel phosphorylation site of the invention, comprising at least one of the following: an AQUA peptide comprising the phosphorylation site, or an antibody or an antigen-binding fragment thereof that binds to an amino acid sequence comprising the phosphorylation site. Such a kit may further comprise a packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay. Where the antibody is labeled with an enzyme, the kit will include substrates and co-factors required by the enzyme. In addition, other additives may be included such as stabilizers, buffers and the like. The relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents that substantially optimize the sensitivity of the assay. Particularly, the reagents may be provided as dry powders, usually lyophilized, including excipients that, on dissolution, will provide a reagent solution having the appropriate concentration.


The following Examples are provided only to further illustrate the invention, and are not intended to limit its scope, except as provided in the claims appended hereto. The invention encompasses modifications and variations of the methods taught herein which would be obvious to one of ordinary skill in the art.


Example 1
Isolation of Phosphotyrosine-Containing Peptides from Extracts of Carcinoma and/or Leukemia Cell Lines and Identification of Novel Phosphorylation Sites

In order to discover novel tyrosine phosphorylation sites in leukemia, IAP isolation techniques were used to identify phosphotyrosine-containing peptides in cell extracts from human leukemia cell lines and patient cell lines identified in Column G of Table 1 including: 101206; 143.98.2; 23132/87; 23132/87: 10% serum; 3T3(EGFR|deletion∥EGF); 42-MG-BA; 5637; 639L; 8-MG-BA; A172; A498; A549; A704; AML-30410; B13_AML; B18_AML; BC-3C; BC004; BC005; BC007; BJ629; BJ630; BJ631; BJ635; BJ665; BT1; BT2; Baf3(FGFR1|truncation: 10ZF); Baf3(FLT31D835Y); Baf3(FLT3|K663Q); Baf3(Jak2|Jak2|V617F); Baf3(TEL-FGFR3); CAKI-2; CAL-29; CAL-51; CAL-85-1; CCF-STTG1; CHP-212; CHP126; CHRF; CMK; CML-06/164; COLO-699; CTV-1; Caki-2; Cal-148; Calu-3; CaoV4; Colo680N; DK-MG; DMS 53; DMS 79; DU.528; DV-90; Detroit562; EFM-19; EFO-21; ENT01; ENT02; ENT03; ENT05; ENT10; ENT12; ENT14; ENT15; ENT19; ENT7; EOL-1; ES2; EVSA-T; FUOV1; GAMG; GI-CA-N; GI-L1-N; GMS-10; H1435; H1437; H1568; H1650; H1651; H1703; H1734; H1781; H1838; H1915; H1975; H2023; H2052; H2066; H2085; H2135; H2172; H2342; H2452; H28; H3255; H358; H4; H446; H4; H520; H524; H596; H810; HCC1143; HCC1395; HCC1428; HCC15; HCC1806; HCC1937; HCC827; HCT 116; HCT15; HCT8; HD-MyZ; HDLM-2; HEL; HL130A; HL131B; HL145A; HL146A; HL152A; HL183A; HL183B; HL184A; HL184B; HL1881; HL213A; HL226A; HL226B; HL233B; HL234A; HL25A; HL53A; HL53B; HL55A; HL59A; HL66A; HL68A; HL83A; HL84A; HL98A; HP28; HT29; Hs746T; IMR32; J82; Jurkat; K562; KATO III; KELLY; KG-1; KMS-11; KPL-1; Kyse140; Kyse270; Kyse410; Kyse450; Kyse510; Kyse70; L428; L540; LAN-1; LAN-5; LCLC-103H; LN-405; LN18; LXF-289; M059J; M059K; MDAH2774; MHH-NB-11; MKN-45; MKPL-1; ML-1; MT-3; MV4-11; Me-F2; MiaPaca; Molm 14; N06BJ505(2); N06BJ573(9); N06BJ591(11); N06BJ593(13); N06BJ606(19); N06CS02; N06CS06; N06CS103; N06CS106; N06CS107; N06CS16; N06CS17; N06CS22(2)-R; N06CS22-1; N06CS22-2; N06CS23; N06CS39; N06CS40; N06CS75; N06CS77; N06CS87; N06CS90; N06CS91; N06CS93-2; N06CS94; N06CS97; N06CS98; N06CS98-2; N06CS98-R; N06N101; N06N102; N06N103; N06N106; N06N121; N06N127; N06N128; N06N131; N06N80; N06N90; N06N93; N06bj523(3); N06bj567(7); N06bj594(14); N06bj595(15); N06bj638(26); N06bj639(27); N06bj667(29); N06c144; N06c78; N06cs108; N06cs109; N06cs110; N06cs110-R; N06cs112; N06cs113; N06cs117; N06cs121; N06cs122; N06cs123; N06cs123(2); N06cs126; N06cs128; N06cs132; N06cs132-1; N06cs59; N06cs63; N06cs72; N06cs76; N06cs88; N06cs92; NALM-19; NCI-H716; OPM-1; OV90; PA-1; RSK2-3; RSK2-4; S 2; SCLC T1; SCLC T4; SEM; SK-ES-1; SK-N-AS; SK-N-BE(2); SK-N-DZ; SK-N-FI; SK-OV-3; SNB-19; SNU-1; SNU-16; SNU-5; SNU-C2B; SUP-T13; SW1088; SW1710; SW480; SW620; Scaber; T17; T98G; TOV112D; TOV21G; U118 MG; UM-UC-1; UT-7; ZR-75-30; brain; cs002; cs005; cs018; cs009; cs024; cs025; cs026; cs037; cs041; cs042; cs057; cs068; cs069; cs070; cs103; cs104; cs105; cs106; cs107; cs110; cs114; cs133; cs136; csC44; csC45; csC50; csC52; csC56; csC60; csC66; csC71; gz21; gz30; gz33; gz41; gz42; gz7; gz73; gz74; gz75; gzB1; h2073; sw48.


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.


Adherent cells at about 70-80% confluency were starved in medium without serum overnight and stimulated, with ligand depending on the cell type or not stimulated. After complete aspiration of medium from the plates, cells were scraped off the plate in 10 ml lysis buffer per 2×108 cells (20 mM HEPES pH 8.0, 9 M urea, 1 mM sodium vanadate, supplemented with 2.5 mM sodium pyrophosphate, 1 mM β-glycerol-phosphate) and sonicated.


Frozen tissue samples were cut to small pieces, homogenize in lysis buffer (20 mM HEPES pH 8.0, 9 M Urea, 1 mM sodium vanadate, supplemented with 2.5 mM sodium pyrophosphate, 1 mM β-glycerol-phosphate, 1 ml lysis buffer for 100 mg of frozen tissue) using a polytron for 2 times of 20 sec. each time. Homogenate is then briefly 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) was added at 10-20 μg/mL. Digestion was performed for 1 day 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 (Waters) 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 matter (mainly in peptide fractions III) was removed by centrifugation. IAP was performed on each peptide fraction separately. The phosphotyrosine monoclonal antibody P-Tyr-100 (Cell Signaling Technology, Inc., catalog number 9411) was coupled at 4 mg/ml beads to protein G (Roche), respectively. Immobilized antibody (15 μl, 60 μg) was added as 1:1 slurry in IAP buffer to 1 ml of each peptide fraction, and the mixture was incubated overnight at 4° C. with gentle rotation. The immobilized antibody beads were washed three times with 1 ml IAP buffer and twice with 1 ml water, all at 4° C. Peptides were eluted from beads by incubation with 75 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% acetonitrile 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 matter 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 55 μl of 0.15% TFA at room temperature for 10 min (eluate 1), followed by a wash of the beads (eluate 2) with 45 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 C18 microtips (StageTips or ZipTips). 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 7.6-9.0 μl of 0.4% acetic acid/0.005% heptafluorobutyric acid. For single fraction analysis, 1 μl of 60% MeCN, 0.1% TFA, was used for elution from the microcolumns. This sample was loaded onto a 10 cm×75 μm PicoFrit capillary column (New Objective) packed with Magic C18 AQ reversed-phase resin (Michrom Bioresources) using a Famos autosampler with an inert sample injection valve (Dionex). The column was then developed with a 45-min linear gradient of acetonitrile delivered at 200 nl/min (Ultimate, Dionex), and tandem mass spectra were collected in a data-dependent manner with an LTQ 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 Browser package (v. 27, rev. 12) supplied as part of BioWorks 3.0 (ThermoFinnigan). Individual MS/MS spectra were extracted from the raw data file using the Sequest Browser program CreateDta, with the following settings: bottom MW, 700; top MW, 4,500; minimum number of ions, 40; minimum TIC, 2×103; and precursor charge state, unspecified. Spectra were extracted from the beginning of the raw data file before sample injection to the end of the eluting gradient. The IonQuest and VuDta programs were not used to further select MS/MS spectra for Sequest analysis. MS/MS spectra were evaluated with the following TurboSequest parameters: peptide mass tolerance, 2.5; fragment ion tolerance, 1.0; 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 then current NCBI human protein database. 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.


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 phosphopeptide sequence is assigned to co-eluting ions with different charge states, since the MS/MS spectrum changes markedly with charge state; (ii) the phosphorylation 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 phosphorylation site is found in more than one peptide sequence context due to homologous but not identical protein isoforms; (iv) the phosphorylation site is found in more than one peptide sequence context due to homologous but not identical proteins among species; and (v) phosphorylation 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 used 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 sequence assignments 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 are 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 Phosphorylation Site-Specific Polyclonal Antibodies

Polyclonal antibodies that specifically bind a novel phosphorylation site of the invention (Table 1/FIG. 2) only when the tyrosine residue is phosphorylated (and does not bind to the same sequence when the tyrosine is not phosphorylated), and vice versa, are produced according to standard methods by first constructing a synthetic peptide antigen comprising the phosphorylation site and then immunizing an animal to raise antibodies against the antigen, as further described below. Production of exemplary polyclonal antibodies is provided below.


A. PSD-95 (tyrosine 576).


A 28 amino acid phospho-peptide antigen, EDSVLSYETVTQMEVHy*ARPIIILGPTK (SEQ NO: 35; y*=phosphotyrosine), which comprises the phosphorylation site derived from human PSD-95 (an adaptor/scaffold protein, Tyr 576 being the phosphorylatable residue), 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) phosphorylation site-specific polyclonal antibodies as described in Immunization/Screening below.


B. PIP5K (Tyrosine 154).


An 20 amino acid phospho-peptide antigen, GKSQDSDLKQy*WMPDSQCKE (SEQ ID NO: 200; y*=phosphotyrosine), which comprises the phosphorylation site derived from human PIP5K (a cytoskeletal protein, Tyr 154 being the phosphorylatable residue), 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) phosphorylation site-specific polyclonal antibodies as described in Immunization/Screening below.


C. FGFR1 (Tyrosine 522).


A 9 amino acid phospho-peptide antigen, GMEy*LASKK (SEQ ID NO: 266; y*=phosphotyrosine, which comprises the phosphorylation site derived from human FGFR1 (a protein kinase, Tyr 522 being the phosphorylatable residue), 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) phosphorylation site-specific polyclonal 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 (500 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 an unphosphorylated synthetic peptide antigen-resin Knotes column to pull out antibodies that bind the unphosphorylated form of the phosphorylation sites. 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 phosphorylation sites. After washing the column extensively, the bound antibodies (i.e. antibodies that bind the phosphorylated peptides described in A-C above, but do not bind the unphosphorylated form of the peptides) 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 PSD-95, PIP5K or FGFR1), for example, brain tissue, jurkat cells or colorectal cancer tissue. 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 phosphorylation site-specific antibody is used at dilution 1:1000. Phospho-specificity of the antibody will be shown by binding of only the phosphorylated form of the target amino acid sequence. Isolated phosphorylation site-specific polyclonal antibody does not (substantially) recognize the same target sequence when not phosphorylated at the specified tyrosine position (e.g., the antibody does not bind to FGFR1 in the non-stimulated cells, when tyrosine 522 is not phosphorylated).


In order to confirm the specificity of the isolated antibody, different cell lysates containing various phosphorylated signaling proteins other than the target protein are prepared. The Western blot assay is performed again using these cell lysates. The phosphorylation site-specific polyclonal antibody isolated as described above is used (1:1000 dilution) to test reactivity with the different phosphorylated non-target proteins. The phosphorylation site-specific antibody does not significantly cross-react with other phosphorylated signaling proteins that do not have the described phosphorylation site, although occasionally slight binding to a highly homologous sequence 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 Phosphorylation Site-Specific Monoclonal Antibodies

Monoclonal antibodies that specifically bind a novel phosphorylation site of the invention (Table 1) only when the tyrosine residue is phosphorylated (and does not bind to the same sequence when the tyrosine is not phosphorylated) are produced according to standard methods by first constructing a synthetic peptide antigen comprising the phosphorylation site 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. PSD-95 (Tyrosine 576).


A 28 amino acid phospho-peptide antigen, EDSVLSYETVTQMEVHy*ARPIIILGPTK (SEQ ID NO: 35; y*=phosphotyrosine), which comprises the phosphorylation site derived from human PSD-95 (an adaptor/scaffold protein, Tyr 576 being the phosphorylatable residue), 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 phosphorylation site-specific monoclonal antibodies as described in Immunization/Fusion/Screening below.


B. PIP5K (Tyrosine 154).


A 20 amino acid phospho-peptide antigen, GKSQDSDLKQy*WMPDSQCKE (SEQ ID NO: 200; y*=phosphotyrosine), which comprises the phosphorylation site derived from human PIP5K (a non-protein kinase, Tyr 154 being the phosphorylatable residue), 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 phosphorylation site-specific monoclonal antibodies as described in Immunization/Fusion/Screening below.


C. FGFR1 (Tyrosine 522).


A 12 amino acid phospho-peptide antigen, GMEy*LASKK (SEQ ID NO: 266; y*=phosphotyrosines), which comprises the phosphorylation site derived from human FGFR1 (a protein kinase, Tyr 522 being the phosphorylatable residue), 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 phosphorylation site-specific monoclonal 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 PSD-95, PIP5K and FGFR1) 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.


Example 4
Production and Use of AQUA Peptides for Detecting and Quantitating Phosphorylation at a Novel Phosphorylation Site

Heavy-isotope labeled peptides (AQUA peptides (internal standards)) for the detecting and quantitating a novel phosphorylation site of the invention (Table 1) only when the tyrosine residue is phosphorylated 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. SKB1 (Tyrosine 283).


An AQUA peptide comprising the sequence, EFCSYLQy*LEYLSQNR (SEQ ID NO: 79; y*=phosphotyrosine; Leucine being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from SKB1 (a cell cycle regulation protein, Tyr 283 being the phosphorylatable residue), 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 SKB1 (tyr 283) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated SKB1 (tyr 283) in the sample, as further described below in Analysis & Quantification.


B. Cofilin 1 (Tyrosine 82).


An AQUA peptide comprising the sequence MLPDKDCRy*ALYDATYETKESK (SEQ ID NO: 103 y*=phosphotyrosine; Proline being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from human Cofilin1 (Tyr 82) being the phosphorylatable residue), 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 Cofilin 1 (Tyr 82) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated Cofilin 1 (Tyr 82) in the sample, as further described below in Analysis & Quantification.


C. CDK9 (Tyrosine 287).


An AQUA peptide comprising the sequence LKAYVRDPy*ALDLIDKLLVLDPAQR (SEQ ID NO: 239; y*=phosphotyrosine; Leucine being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from human CDK9 (Tyr 287 being the phosphorylatable residue), 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 CDK9 (Tyr 287) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated CDK9 (Tyr 287) in the sample, as further described below in Analysis & Quantification.


D. DNAJB6 (Tyrosine 53).


An AQUA peptide comprising the sequence QVAEAy*EVLSDAK (SEQ ID NO: 53; y*=phosphotyrosine; valine being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from human DNAJB6 (Tyr 53 being the phosphorylatable residue), 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 DNAJB6 (Tyr 53) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated DNAJB6 (Tyr 53) 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 or LTQ) MS.


MS/MS spectra for each AQUA peptide should exhibit a strong y-type ion peak as the most intense fragment ion that is suitable for use in an SRM monitoring/analysis. Reverse-phase microcapillary columns (0.1 Ř150-220 mm) are prepared according to standard methods. An Agilent 1100 liquid chromatograph may be used to develop and deliver a solvent gradient [0.4% acetic acid/0.005% heptafluorobutyric acid (HFBA)/7% methanol and 0.4% acetic acid/0.005% HFBA/65% methanol/35% acetonitrile] to the microcapillary column by means of a flow splitter. 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 proteins 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 (LCQ DecaXP ion trap or TSQ Quantum triple quadrupole or LTQ). On the DecaXP, parent ions are isolated at 1.6 m/z width, the ion injection time being limited to 150 ms per microscan, with two microscans per peptide averaged, and with an AGC setting of 1×108; 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., 500 fmol).

Claims
  • 1. An isolated phosphorylation site-specific antibody that specifically binds a human DDEF2 only when phosphorylated at a tyrosine at position 724 comprised within the phosphorylatable peptide sequence set forth in SEQ ID NO: 172, wherein said antibody does not bind said protein when not phosphorylated at said tyrosine at position 724.
  • 2. An isolated phosphorylation site-specific antibody that specifically binds a human DDEF2 only when not phosphorylated at a tyrosine at position 724 comprised within the phosphorylatable peptide sequence set forth in SEQ ID NO: 172, wherein said antibody does not bind said protein when phosphorylated at said tyrosine at position 724.
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. 60/925,253, filed Apr. 19, 2007, the disclosure of which is incorporated herein, in its entirety, by reference.

US Referenced Citations (66)
Number Name Date Kind
3940475 Gross et al. Feb 1976 A
4289747 Chu et al. Sep 1981 A
4349893 Wiegman et al. Sep 1982 A
4376110 David et al. Mar 1983 A
4474893 Reading et al. Oct 1984 A
4634664 Oestberg et al. Jan 1987 A
4634666 Engleman et al. Jan 1987 A
4659678 Forrest et al. Apr 1987 A
4676980 Segal et al. Jun 1987 A
4727022 Skold et al. Feb 1988 A
4816397 Boss et al. Mar 1989 A
4816567 Cabilly et al. Mar 1989 A
4946778 Ladner et al. Aug 1990 A
5004692 Tso et al. Apr 1991 A
5092885 Yamada et al. Mar 1992 A
5112946 Maione et al. May 1992 A
5192744 Bouck et al. Mar 1993 A
5202352 Okada et al. Apr 1993 A
5225539 Winter et al. Jul 1993 A
5260203 Ladner et al. Nov 1993 A
5530101 Queen et al. Jun 1996 A
5545806 Lonberg et al. Aug 1996 A
5565332 Hoogenboom et al. Oct 1996 A
5585089 Queen et al. Dec 1996 A
5675063 Knight et al. Oct 1997 A
5677427 Goldenberg et al. Oct 1997 A
5693761 Queen et al. Dec 1997 A
5693762 Queen et al. Dec 1997 A
5789208 Sharon et al. Aug 1998 A
5969108 McCafferty et al. Oct 1999 A
6103889 Whitlow et al. Aug 2000 A
6120767 Robinson et al. Sep 2000 A
6150584 Kucherlapati et al. Nov 2000 A
6180370 Queen et al. Jan 2001 B1
6329508 Friden et al. Dec 2001 B1
6331415 Cabilly et al. Dec 2001 B1
6335163 Sharon et al. Jan 2002 B1
6355245 Evans et al. Mar 2002 B1
6395718 Slusher et al. May 2002 B1
6407213 Carter et al. Jun 2002 B1
6441140 Comb et al. Aug 2002 B1
6462075 Bowen et al. Oct 2002 B1
6465431 Thorn et al. Oct 2002 B1
6475784 Papkoff et al. Nov 2002 B1
6482802 Hu et al. Nov 2002 B1
6482810 Brem et al. Nov 2002 B1
6500431 Gill et al. Dec 2002 B1
6500924 Brooks et al. Dec 2002 B1
6518298 Green et al. Feb 2003 B2
6521439 Folkman et al. Feb 2003 B2
6525019 D'Amato et al. Feb 2003 B2
6538103 Ji et al. Mar 2003 B1
6544758 O'Reilly et al. Apr 2003 B2
6544947 Holaday et al. Apr 2003 B2
6548477 Olson et al. Apr 2003 B1
6548640 Winter et al. Apr 2003 B1
6559126 Tournaire et al. May 2003 B2
6569845 Futamura et al. May 2003 B1
6573256 Bishop et al. Jun 2003 B2
6783961 Edwards et al. Aug 2004 B1
6867007 Kauvar et al. Mar 2005 B2
6884869 Senter et al. Apr 2005 B2
6979557 Isogai et al. Dec 2005 B2
7109000 Edinger et al. Sep 2006 B2
7198896 Rush et al. Apr 2007 B2
7300753 Rush et al. Nov 2007 B2
Foreign Referenced Citations (25)
Number Date Country
0120694 Mar 1984 EP
1184665 Sep 1986 EP
0239400 Sep 1987 EP
0404097 Dec 1990 EP
WO 8403508 Sep 1984 WO
WO 8503508 Aug 1985 WO
WO 8601533 Mar 1986 WO
WO 9110741 Jul 1991 WO
WO 9117271 Nov 1991 WO
WO 9201047 Jan 1992 WO
WO 9311161 Jun 1993 WO
WO 9312227 Jun 1993 WO
WO 9520401 Aug 1995 WO
WO 9627011 Jun 1996 WO
WO 0049144 Aug 2000 WO
WO 0200729 Mar 2002 WO
WO 03016861 Feb 2003 WO
WO 03089474 Oct 2003 WO
WO 03106644 Dec 2003 WO
WO 2004009618 Jan 2004 WO
WO 2004039963 May 2004 WO
WO 2004066957 Aug 2004 WO
WO 2004091658 Oct 2004 WO
WO 2005056825 Jun 2005 WO
WO 2005083444 Sep 2005 WO
Related Publications (1)
Number Date Country
20090098581 A1 Apr 2009 US
Provisional Applications (1)
Number Date Country
60925253 Apr 2007 US