Protein Phosphorylation By Basophillic Serine/Threonine Kinases

Abstract
The invention discloses 461 novel phosphorylation sites identified in basophilic Ser/Thr kinase signaling pathways, 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

This invention relates to novel Serine/Threonine (S/T) protein phosphorylation sites in basophilic S/T kinase signaling pathways as well as 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 diabetes, cancer, developmental disorders, and autoimmune diseases. 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 investigate it.


The AGC protein kinase group contains 50 different kinases that share similar kinase domain structures and substrate preferences. The group includes PDK1, a master regulator of many other AGC kinases, and the Akt, protein kinase A (PKA), protein kinase C(PKC), ribosomal S6 kinase (RSK), serum- and glucocorticoid-induced kinase (SGK), and NDR/LATS kinase families (Mora et al, Semin Cell Dev Biol. 2004 15:161-70). AGC kinases and other basophilic kinases play critical roles in regulating growth, metabolism, proliferation and survival.


All of the AGC kinases studied to date are basophilic, i.e. they prefer basic amino acids flanking the serines/threonines that they phosphorylate. Some members of the AGC group have stringent requirements for basic residues at specific locations relative to the phosphorylated serine/threonine. For instance, the three Akt isoforms (Akt1-3) appear to have a nearly exclusive preference for arginine (R) at positions −5 and −3 relative to the phospho-acceptor residue at position 0. p70S6K and p90RSK can apparently tolerate lysine (K) or arginine (R) at position −5 better than the Akt kinases (Manning and Cantley, Cell. 2007 129:1261-74). Other kinases have more relaxed requirements for arginine on either side of the phospho-acceptor. PKA prefers at least one arginine/lysine at the −1, −2 or −3 positions. PKCs can phosphorylate sequences with arginines or lysines either C-terminal or N-terminal to the phosphoacceptor site (see FIG. 6).


A crucial early event in receptor mediated signaling is the activation of phosphatidylinositol 3-kinase (PI3K) and generation of phosphatidylinositol 3,4,5-trisphosphate (PIP3), a second messenger on the inner surface of the plasma membrane. PI3K phosphorylates phosphatidylinositol-4,5-bisphosphate (PIP2) to generate PIP3, in a reaction that can be reversed by the PIP3 phosphatase PTEN. PIP3 then recruits the AGC kinases PDK1 and Akt to the plasma membrane, where PDK1 is rapidly phosphorylated and activated (Cohen et al., FEBS Lett. 1997 Jun. 23; 410(1):3-10; Riojas et al, J Biol. Chem. 2006 281:21588-93).


mTOR, another crucial substrate of PDK, is an atypical protein kinase that is required for cell survival and regulates cell growth through the regulation of protein synthesis. When sufficient nutrients are available, mTOR is activated and regulates protein synthesis by phosphorylating and activating p70S6K, an AGC kinase with a specificity nearly identical to that of Akt, and phosphorylating and inactivating eukaryotic initiation factor 4E-binding protein (4E-BP1), a repressor of mRNA translation (Hay and Sonenberg, Genes Dev. 2004 18:1926-45).


Much of this control exerted by PDK1 and mTOR is mediated by their ability to phosphorylate key AGC kinases, which in turn regulate many downstream effector networks. PDK1 activates Akt and other members of the AGC group including PKC-delta, PKC-epsilon, PKC-zeta, PKN1, PKN2, SGK, SGK2, and SGK3. Many of these basophilic kinases in turn regulate other ser/thr kinases networks. For example, Akt1 or Akt2 phosphorylates ASK1, IKK-alpha, MLK3, SEK1, mTOR, QIK, Raf1, and WNK1; PKC-delta phosphorylates LIMK2, and p38-alpha.


Signals from AGC Kinases and other Basophillic Kinases set in motion a concerted response that touches virtually every compartment of cellular dynamics: metabolic regulation, DNA transcription, RNA processing, protein synthesis, vesicular transport, endocytosis, adhesion, molecular transport, and protein degradation, but very little of these processes are understood at the molecular level.


Despite the identification of a few key-signaling molecules involved in basophilic serine/threonine kinase related pathways and related disease progression are known, the vast majority of signaling protein changes and signaling pathways underlying the various associated disease types remain unknown. Therefore, there is presently an incomplete and inaccurate understanding of how protein activation within basophilic serine/threonine kinase related pathways drives various diseases including, among many others, various types of cancer and diabetes. Accordingly, there is a continuing and pressing need to unravel the molecular mechanisms of disease progression by identifying the downstream signaling proteins mediating cellular transformation in these diseases.


Presently, diagnosis of many basophilic serine/threonine kinase related diseases and cancer may made by tissue biopsy and detection of different cell surface markers. However, misdiagnosis can occur since some disease types 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 a disease including cancer can be sometimes detected, it is clear that other downstream effectors of constitutively active signaling molecules 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, cancer, 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 serine and threonine phosphorylation sites (Table 1) identified in basophilic S/T kinase signaling pathways. The novel sites occur in proteins such as: Adaptor/Scaffold proteins, apoptosis proteins, enzyme proteins, non-protein kinases, phosphatases, proteases, protein kinases Ser/Thr (non-receptor), vesicle proteins, g proteins or regulator proteins, chromatin or DNA binding/repair/replication proteins, cytoskeletal proteins, receptor/channel/transporter/cell surface proteins, RNA processing proteins, translation proteins, activator proteins, chaperone proteins, calcium binding proteins, transcriptional regulator proteins, tumor suppressor proteins, lipid binding proteins, secreted proteins, adhesion or extracellular matrix proteins, inhibitor proteins, mitochondrial proteins, endoplasmic reticulum or golgi apparatus proteins, cell cycle regulation proteins, transcriptional regulator proteins, ubiquitin conjugating proteins, 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 serine and/or threonine phosphorylation at a novel phosphorylation sites 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-Isotope 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 serine or threonine identified in Column D is phosphorylated, and do not significantly bind when the serine or threonine is not phosphorylated. In another embodiment, the antibodies specifically bind to an amino acid sequence comprising a phosphorylation site when the serine or threonine is not phosphorylated, and do not significantly bind when the serine or threonine 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 basophilic serine/threonine kinase signaling pathway related disease in a subject, wherein the disease 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 serine or threonine phosphorylation site of the invention.


In another aspect, the invention provides a method for identifying an agent that modulates a serine and/or threonine 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 serine and/or threonine in the presence of the test agent, as compared to a control, indicates that the candidate agent potentially modulates serine and/or threonine 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 461 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 serine and/or threonine residue at which phosphorylation occurs (each number refers to the amino acid residue position of the serine and/or threonine in the parent human protein, according to the published sequence retrieved by the SwissProt accession number); Column E=flanking sequences of the phosphorylatable serine and/or threonine residues; sequences (SEQ ID NOs: 1-461) were identified using Trypsin digestion of the parent proteins; in each sequence, the serine and/or threonine (see corresponding rows in Column D) appears in lowercase; Column F=the diseases with which the phosphorylation site has been associated; Column G=the cell type(s)/Tissues 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 serine 159 in ARHGEF2, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); lowercase “s” indicates the phosphorylated serine (corresponds to lowercase “s” in Column E of Table 1; SEQ ID NO: 27).



FIG. 4 is an exemplary mass spectrograph depicting the detection of the phosphorylation of threonine 1825 in afadin iso3, as further described in Example 1 (red and blue indicate ions detected in MS/MS spectrum); lowercase “t” indicates the phosphorylated Threonine (corresponds to lowercase “t” in Column E of Table 1; SEQ ID NO: 10).



FIG. 5 is a Western blot analysis of extracts from Jurkat and THP-1 cell lines using Phospho-SATB1 (Ser47) Antibody (SEQ. ID NO. 353) (upper) or SATB1 (L745) Antibody #3650 (lower). Antibody phospho-specificity was determined by treating cell extracts with λ phosphatase.





Additional Description for FIGS. 3 and 4


FIGS. 3 and 4 consists of two panes, a labeled mass spectrograph (MS/MS spectrum) in the top pane and a table of expected and observed ion mass-to-charge ratios in the bottom pane.


Top Pane, Miss./MS Spectrum

The x-axis is mass-to-charge ratio, and the y-axis is intensity. When spectra are shown in two panels so the peaks are more clearly labeled, the panels can have different y-axes maxima.


Spectrum peaks are labeled with ion assignments, usually b and y ions for CID spectra and c and z ions for ETD spectra, followed by the number of amino acid residues and the ion charge state. Peak labels enclosed in brackets indicate ions that have undergone neutral loss of one water group or one ammonia group. For example, “y11++” is they ion with 11 amino acid residues (spanning the first 11 peptide sequence residues) and with a charge of +2; “<b6+>” is the b ion with 6 residues (spanning the last 6 peptide sequence residues) and with a charge of +1 that has lost one water group or one ammonia group. A peak that appears to have more than one label is usually several nearby peaks with similar mass-to-charge ratios. An unlabeled peak usually corresponds to an ion that has undergone neutral loss of more than one water or ammonia group, or that has undergone neutral loss of a phosphate group, or that has a charge state greater than 2.


Bottom Pane, Table of Expected and Observed Ion Mass-to-charge Ratios

The peptide sequence is given in column AA, with b (or c) ion numbering in the left # column and y (or z) ion numbering in the right # column. Modified amino acid residues have a light shade in column AA and are listed in the table footer. For example, the footer “S (5):+167.00” means residue 5 of the peptide sequence in modified serine (S) with a residue mass of 167, corresponding to phosphoserine. The remainder of the table gives calculated mass-to-charge ratios for each ion and charge state. Shaded mass-to-charge ratios were observed in the spectrum.


DETAILED DESCRIPTION OF THE INVENTION

The inventors have discovered and disclosed herein novel serine and threonine phosphorylation sites in signaling proteins extracted from the cell line/tissue/patient sample listed in column G of FIG. 2. The newly discovered phosphorylation sites significantly extend our knowledge of basophillic Ser/Thr kinases, 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 basophilic Ser/Thr related signaling pathways cells provides and focuses further elucidation of many disease processes. And, the novel sites provide additional diagnostic and therapeutic targets.


1. Novel Phosphorylation Sites in Basophillic S/T Kinase Signaling Pathways

In one aspect, the invention provides 461 novel serine and/or threonine phosphorylation sites in signaling proteins from cellular extracts and tissue samples, 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 basophilic Ser/Thr Kinase related signaling pathways. 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, enzyme/non-protein kinase/phosphatase proteins, Ser/Thr (non-receptor) protein kinases, vesicle proteins, g proteins or regulator proteins, chromatin or DNA binding/repair/replication proteins, receptor/channel/transporter/cell surface proteins, RNA processing proteins, cytoskeletal proteins, transcriptional regulators and translation 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 is 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 cellular extracts and tissue samples: A 431; Adult mouse brain; Embryo mouse brain; H1373; H1703; H3255; H441; HCC1937; HCT116; HeLa; Jurkat; K562; MKN-45; N06cs95; TH-HY2; XY3-130T; XY3-52-T; XY3-68-T; XY3-95N; mouse brain; mouse liver; xy380T. In addition to the newly discovered phosphorylation sites (all having a phosphorylatable serine or threonine), 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 motif-specific, context-independent 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, the following motif-specific antibodies (commercially available from Cell Signaling Technology, Inc., Beverly, Mass.) may be used in the immunoaffinity step to isolate the widest possible number of phospho-serine and/or phospho-threonine containing peptides from the cell extracts: Akt Substrate; (s/t)F; (s/t)XXX(s/t); ATM/ATR Substrate; Multiplex-1; PKA Substrate; PKC Substrate; SsP; p-Thr; t(D/E)X(D/E); tPP; tXR


As described in more detail in the Examples, lysates may be prepared from various carcinoma 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 the following motif-specific antibodies: Akt Substrate; (s/t)F; (s/t)XXX(s/t); ATM/ATR Substrate; Multiplex-1; PKA Substrate; PKC Substrate; SsP; p-Thr; t(D/E)X(D/E); tPP; and tXR (commercially available from Cell Signaling Technology, Inc., Beverly, Mass.) 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 serine and/or threonine residue at which phosphorylation occurs (each number refers to the amino acid residue position of the serine and/or threonine in the parent human protein, according to the published sequence retrieved by the SwissProt accession number). Column E shows flanking sequences of the identified serine and/or threonine residues (which are the sequences of trypsin-digested peptides










TABLE 1







Novel Serine and Threonine Phosphorylation Sites.

















B
C
D
E
H




A
Accession
Protein
Phospho
Phosphorylation
SEQ. ID.


  1
Name
Number
Type
Residue
Site Sequence
NO:
















  2
AADACL3
NP_001096640.1
Unassigned
T51
LYQSKAStCTLKPGI
  1





  3
ABCA2
NP_997698.1
Receptor, channel,
T2132
TSTFKMLtGDESTTG
  2





transporter or cell





surface protein





  4
Abi-1
NP_005461.2
Adaptor/scaffold
S319
GTMTRQIsRHNSTTS
  3





  5
Abi-1
NP_005461.2
Adaptor/scaffold
S338
GGYRRTPsVTAQFSA
  4





  6
Abi-1 iso3
NP_005461.2
Adaptor/scaffold
S240
NQRPRTHsGSSGGSG
  5





  7
ADAM19
NP_075525.2
Unassigned
S831
SQIERTEsSRRPPPS
  6





  8
ADCY7
NP_001105.1
Receptor, channel,
T365
IKQVREAtGVDINMR
  7





transporter or cell





surface protein





  9
ADD1
NP_001110.2
Cytoskeletal protein
T724
KKKKKFRtPSFLKKS
  8





 10
AF-4
NP_005926.1
Transcriptional
S837
KEIKSQsSSSSSSHK
  9





regulator





 11
afadin iso3
NP_001035090.1
Adhesion or
T1825
VKASRKLtELENELN
 10





extracellular matrix





protein





 12
AFAP1L2
NP_115939.1
Cell cycle regulation
T345
NLGRKKStSLEPVER
 11





 13
ANK1
NP_000028.3
Adaptor/scaffold
T1380
EDRRRTPtPLALRYS
 12





 14
ANK2
NP_001139.3
Adaptor/scaffold
T2393
EASVKTDtGTESKPQ
 13





 15
ANKRD25
NP_056308.2
Transcriptional
S540
EPRERVPsVAEAPQL
 14





regulator





 16
ANKRD3
NP_065690.2
Protein kinase,
S364
SKLPsSGSGKRLSGV
 15





Ser/Thr (non-





receptor)





 17
ANKRD3
NP_065690.2
Protein kinase,
S372
SGSGKRLsGVSSVDS
 16





Ser/Thr (non-





receptor)





 18
ANKRD34
NP_001034977.1
Unassigned
S291
PRLSRRHsTEGPEDP
 17



A





 19
ANKRD34
NP_001034977.1
Unassigned
T316
GPLSRRNtAPEAQES
 18



A





 20
APBA1
NP_001154.2
Adaptor/scaffold
S80
ECLARSAsTESGFHN
 19





 21
APC
NP_001120983.1
Tumor suppressor
T2442
PVLVRQStFIKEAPS
 20





 22
AREGEF1
NP_006412.2
G protein or
S1079
TVRGREGsLTGTKDQ
 21





regulator





 23
ARHGAP1
NP_060757.4
G protein or
T639
KFLTRRPtLQAVREK
 22



2

regulator





 24
ARHGAP1
NP_060524.4
G protein or
S484
LERKRPAsMAVMEGD
 23



7

regulator





 25
ARHGAP2
NP_065875.3
G protein or
T980
YKDKREQtTPSEEEQ
 24



1

regulator





 26
ARHGAP6
NP_038286.2
G protein or
S436
VGIFRVGsSKKRVRQ
 25





regulator





 27
ARHGEF1
NP_055601.2
G protein or
S395
RGSSRYSsTETLKDD
 26



7

regulator





 28
ARHGEF2
NP_004714.2
G protein or
S159
NMRNRTLsVESLIDE
 27





regulator





 29
ARHGEF2
NP_004714.2
G protein or
S95
TIRERPSsAIYPSDS
 28





regulator





 30
ARHGEF2
NP_004714.2
G protein or
T157
SLNMRNRtLSVESLI
 29





regulator





 31
ARHGEF5
NP_005426.2
G protein or
S1044
RQGLRRPsILPEGSS
 30





regulator





 32
ARHGEF7
NP_001106984.1
G protein or
S531
QKQTKVTsVGNPTIK
 31





regulator





 33
ARVCF
NP_001661.1
Cytoskeletal protein
S916
ERRPRGAsSAGEASE
 32





 34
AS250
NP_066076.2
G protein or
S713
TEPMRFRsATTSGAP
 33





regulator





 35
AS250
NP_065076.2
G protein or
S844
KCRERQKsESTNSDT
 34





regulator





 36
AS250
NP_065076.2
G protein or
S846
RERQKSEsTNSDTTL
 35





regulator





 37
Asxl3
NP_085135.1
Unknown function
S929
NKTHKQGsTQSRLET
 36





 38
Asxl3
NP_085135.1
Unknown function
T936
STQSRLEtSHTSKSS
 37





 39
ATAD3A
NP_060658.2
Mitochondrial
S337
PSLVRETsRITVLEA
 38





protein





 40
ATF7
NP_001123532.1
Transcriptional
S44
FGPARTDsVIIADQT
 39





regulator





 41
ATPAF1
NP_073582.2
Unassigned
S145
FTKDKTLsSIFNIEM
 40





 42
AUTS2
NP_001120703.1
Unknown function
S1209
TLGGRPVsPRRTTPL
 41





 43
AVPR1A
NP_000697.1
Receptor, channel,
S283
VSSVKSIsRAKIRTV
 42





transporter or cell





surface protein





 44
B-Raf
NP_004324.2
Protein kinase,
S465
TVGQRIGsGSFGTVY
 43





Ser/Thr (non-





receptor)





 45
BAZ2B
NP_038478.2
Unknown function
S465
SNPKATsSSPAHPKQ
 44





 46
BC065369
CAI22612.3
Unassigned
S107
AREEKRVsGPSASKE
 45





 47
Bcl-6
NP_001697.2
Transcriptional
T653
KSHLRIHtGEKPYHC
 46





regulator





 48
BIN2
NP_057377.2
Adaptor/scaffold
S357
PTTERAKsQEEVLPS
 47





 49
BLNK
NP_001107566.1
Adaptor/scaffold
T231
RAGKKPTtPLKTTPV
 48





 50
BOP1
NP_056016.1
RNA processing
T277
PRRPRDPtPSFYDLW
 49





 51
BRD1
NP_055392.1
Cell
S415
NGVCRKEsSVKTVRS
 50





development/





differentiation





 52
BRD1
NP_055392.1
Cell
S416
GVCRKESsVKTVRST
 51





development/





differentiation





 53
BTBD3
NP_055777.1
Unknown function
S30
KNRSKKSsKKANTSS
 52





 54
C14orf149
NP_653182.1
Unassigned
T273
EQVDRSPtGSGVTAR
 53





 55
C16orf68
NP_077014.2
Unassigned
S132
VRRPRAAsDSNPAGP
 54





 56
C17orf100
NP_001098990.1
Unassigned
S11
ARGAKQSsPRVGTTR
 55





 57
C17orf100
NP_001098990.1
Unassigned
T16
QSSPRVGtTRYTETS
 56





 58
C17orf28
NP_085133.1
Chromatin, DNA-
S670
WREQRRPsTSSASGQ
 57





binding, DNA repair





or DNA replication





protein





 59
C18orf28
NP_001008240.1
Unknown function
S145
RSRsESETSTMAAKK
 58





 60
C6orf146
NP_775834.1
Unassigned
S323
AVTERPSsSKATPKV
 59





 61
C9orf140
NP_848543.2
Unknown function
T219
RGERRRHtIASGVDC
 60





 62
calponin 3
NP_001830.1
Cytoskeletal protein
S254
MGTNKVAsQKGMSVY
 61





 63
capicua
NP_055940.3
Chromatin, DNA-
S299
HKETRERsMSETGTA
 62





binding, DNA repair





or DNA replication





protein





 64
capicua
NP_055940.3
Chromatin, DNA-
S605
FRRKRPEsVGGLEPP
 63





binding, DNA repair





or DNA replication





protein





 65
CAPS2
NP_115995.2
Calcium-binding
S284
ETKIRTHsTLTENVL
 64





protein





 66
CAPS2
NP_115995.2
Calcium-binding
T282
LHETKIRtHSTLTEN
 65





protein





 67
CASZ1
NP_001073312.1
Transcriptional
S437
STFSKTDsITTGTVS
 66





regulator





 68
CCDC138
NP_659415.1
Unassigned
T48
KYKRRTLtSPGDLDI
 67





 69
CCDC27
NP_689705.2
Unassigned
S206
YLRKRRKsQTLSPVT
 68





 70
CCDC27
NP_689705.2
Unassigned
T208
RKRRKSQtLSPVTSS
 69





 71
CCM2
NP_113631.1
Adaptor/scaffold
S393
GRHRRALsTTSSSTT
 70





 72
CD2AP
NP_036252.1
Adhesion or
T229
EGSVKLRtRTSSSET
 71





extracellular matrix





protein





 73
Cdc42EP3
NP_006440.2
G protein or
S89
NEFFRANsTSDSVFT
 72





regulator





 74
CDCA7L
NP_061189.2
Transcriptional
S139
RSRSRRSsIGLRVAF
 73





regulator





 75
CDKAL1
NP_060244.2
Unknown function
T43
PKVRRRNtQKYLQEE
 74





 76
CEP164
NP_055771.4
Unknown function
S1013
ARKLKLEsQVDLLQA
 75





 77
CEP350
NP_055625.4
Cell cycle regulation
S103
TKSRKEKsRSPLRAT
 76





 78
CEP350
NP_055625.4
Cell cycle regulation
S105
SRKEKSRsPLRATTL
 77





 79
CEP350
NP_055625.4
Cell cycle regulation
T2000
HSLPKSCtSVSKQES
 78





 80
CEP72
NP_060610.2
Cytoskeletal protein
S532
EENSRLKsLLLSMKK
 79





 81
CGN
NP_065821.1
Adaptor/scaffold
S1036
DLKTRLAsSEGFQKP
 80





 82
CHCHD3
NP_060282.1
Mitochondrial
S113
ILRERICsEEERAKA
 81





protein





 83
CHD-6
NP_115597.3
Enzyme, misc.
S2680
PSCEREPsGDENCAE
 82





 84
CHKB
NP_689466.1
Unassigned
S39
TPKRRRAsSLSRDAE
 83





 85
CHKB
NP_689466.1
Unassigned
S40
PKRRRASsLSRDAER
 84





 86
CHMP4C
NP_689497.1
Vesicle protein
S214
ARRSRAAsSQRAEEE
 85





 87
CHORDC1
NP_036256.1
Calcium-binding
S200
SCCRRKTsDFNTFLA
 86





protein





 88
CHORDC1
NP_036256.1
Calcium-binding
T199
WSCCRRKtSDFNTFL
 87





protein





 89
CLASP1
NP_056097.1
Cell cycle regulation
T642
ASLGRIRtRRQSSGS
 88





 90
CLIC6
NP_444507.1
Receptor, channel,
S293
AGRARRVsGEPQQSG
 89





transporter or cell





surface protein





 91
CLPX
NP_006651.2
Mitochondrial
S220
AEVEKQTsLTPRELE
 90





protein





 92
CMTM8
NP_849199.2
Receptor, channel,
T11
PQRARSHtVTTTASS
 91





transporter or cell





surface protein





 93
coilin
NP_004636.1
Unknown function
T39
LNRCRVVtDLISLIR
 92





 94
CPXM2
NP_937791.1
Unassigned
S114
KKVMRTKsSEKAAND
 93





 95
CROCCL2
EAW51787.1
Unassigned
S42
RLRDKTDsTMQAHED
 94





 96
CTGLF4
NP_001071153.1
G protein or
S323
KVPGKWPsLATLACT
 95





regulator





 97
CYLD
NP_001035877.1
Ubiquitin
T522
FRGTRYFtCALKKAL
 96





conjugating system





 98
DAAM1
NP_055807.1
Adaptor/scaffold
S34
TYRLRNDsNFALQTM
 97





 99
DACT1
NP_001072988.1
Adaptor/scaffold
S618
GGGHRAGsRAHGHGR
 98





100
DACT1
NP_001072988.1
Adaptor/scaffold
T323
VRTNKPRtSVNADPT
 99





101
DATF1
NP_149072.1
Transcriptional
S1018
SILAKPSsSPDPRYL
100





regulator





102
Dbf4
NP_006707.1
Protein kinase,
T273
QVKLRIQtDGDKYGG
101





regulatory subunit





103
DCUN1D3
NP_775746.1
Unassigned
S155
FDGCKAIsADSIDGI
102





104
DDX19A
NP_060802.1
Unknown function
T467
KKIERLDtDDLDEIE
103





105
DDX43
NP_061135.1
Unassigned
S21
WASRRSsTVSRAPE
104





106
DDX43
NP_061135.1
Unassigned
T203
KNFYKEStATSAMSK
105





107
desmo-
NP_001008844.1
Cytoskeletal protein
T13
GSHPRINtLGRMIRA
106



plakin





108
desmo-
NP_068831.1
Cytoskeletal protein
T54
EACGRQYtLKKTTTY
107



plakin 3





109
DGAT2
NP_115953.2
Unassigned
S37
PALSREGsGRWGTGS
108





110
DGK-D
NP_690618.2
Kinase (non-
S44
QKLIRKVsTSGQIRQ
109





protein)





111
DISP1
NP_116279.2
Receptor, channel,
S1168
SPSDKGQsKTHTINA
110





transporter or cell





surface protein





112
DISP2
NP_277045.1
Receptor, channel,
S1393
DVWLRRPsTHTSGYS
111





transporter or cell





surface protein





113
DLG3
NP_065781.1
Adaptor/scaffold
T454
MEMNRRQtYEQANKI
112





114
DMPK2
NP_059995.2
Protein kinase,
T222
GSCLRLNtNGMVDSS
113





Ser/Thr (non-





receptor)





115
DNCH1
NP_001367.2
Motor or contractile
T4366
ETEKKTRtDSTSDGR
114





protein





116
DNMT3B
NP_787046.1
Enzyme, misc.
S110
FRETRTRsESPAVRT
115





117
DOCK4
NP_055520.3
G protein or
S1724
SPRERPCsAIYPTPV
116





regulator





118
DOT1L
NP_115871.1
Enzyme, misc.
S1093
RGRRKRAsAGTPSLS
117





119
DPH4
NP_859057.4
Enzyme, misc.
T45
YHPDKQStDVPAGTV
118





120
DSPP
NP_055023.2
Adhesion or
T389
SSGNRNItKEVGKGN
119





extracellular matrix





protein





121
DTNBP1
NP_898861.1
Vesicle protein
S11
TLRERLLsVQQDFTS
120





122
DYN1
NP_001005336.1
Vesicle protein
T443
ISTVRQCtKKLQQYP
121





123
E4F1
NP_004415.2
Transcriptional
T743
VLAARAGtSGTEQAT
122





regulator





124
ECT2
NP_060568.3
G protein or
S849
HVMSRLSsTSSLAGI
123





regulator





125
ELYS
NP_056261.3
Transcriptional
T2239
KSKPRKTtEVTGTGL
124





regulator





126
EMILIN1
NP_008977.1
Adhesion or
T385
VLSGRRGtELGGAAG
125





extracellular matrix





protein





127
EMSY
NP_064578.2
Chromatin, DNA-
T207
KPRKRRRtNSSSSSP
126





binding, DNA repair





or DNA replication





protein





128
ENaC
NP_001030.2
Receptor, channel,
S637
LRLERAFsNQLTDTQ
127



gamma

transporter or cell





surface protein





129
ENaC
NP_001030.2
Receptor, channel,
T629
TPPPKYNtLRLERAF
128



gamma

transporter or cell





surface protein





130
Epb4.2
NP_000110.2
Cytoskeletal protein
S278
LLNKRRGsVPILRQW
129





131
eplin
NP_057441.1
Cytoskeletal protein
S55
TNMEKKRsNTENLSQ
130





132
EPS8L1
NP_573441.2
Adaptor/scaffold
S580
WDRPRWDsCDSLNGL
131





133
exophilin
NP_055880.1
G protein or
S1853
SRRFRSFsELPSCDG
132



5

regulator





134
F25965
NP_061977.1
Unassigned
T94
GGPQRSNtYVIKLFD
133





135
FA82C
NP_060615.1
Mitochondrial
S149
FPFVRERsDSTGSSS
134





protein





136
FA82C
NP_060615.1
Mitochondrial
S151
FVRERSDsTGSSSVY
135





protein





137
FALZ
NP_004450.3
Transcriptional
T2481
RIRPStPSQLSPGQQ
136





regulator





138
FAM117A
NP_110429.1
Unknown function
S145
CAHKRSAsWGSTDHR
137





139
FAM125A
NP_612410.1
Adaptor/scaffold
T196
RLGSRAStLRRNDSI
138





140
FAM65B
NP_055537.2
Cytoskeletal protein
S21
NGIIRSQsFAGFSGL
139





141
FBXL13
NP_001104508.1
Ubiquitin
T440
MADCKGItDSSLRSL
140





conjugating system





142
FBXO41
NP_001073879.1
Unassigned
T540
RRPRRHStEGEEGDV
141





143
FDXR
NP_004101.2
Unassigned
T283
PRPRKRLtELLLRTA
142





144
FGFR1
NP_056934.2
Protein kinase, Tyr
S449
VRPSRLSsSGTPMLA
143





(receptor)





145
FGFR2
NP_000132.2
Protein kinase, Tyr
S472
RITTRLSsTADTPML
144





(receptor)





146
FGFR2
NP_000132.2
Protein kinase, Tyr
T468
TPLVRITtRLSSTAD
145





(receptor)





147
FGFR4
NP_998812.1
Protein kinase, Tyr
S440
VRGVRLSsSGPALLA
146





(receptor)





148
FLJ12994
NP_073762.5
Unknown function
S661
PRKRLsSTLQETQVP
147





149
FLJ12994
NP_073752.5
Unknown function
S662
PRKRLSsTLQETQVP
148





150
FLJ13213
NP_001013865.1
Transcriptional
S707
YEQEKRNsLKRPRDV
149





regulator





151
FLJ21438
NP_075055.1
Unknown function
S831
RPARRRQsAGPWPRP
150





152
FLJ21901
NP_078898.3
Unknown function
T83
WKLQKQKtSLLKNAE
151





153
FLJ32810
XP_001127597.2
G protein or
S751
AEGNKSYsGSIQSLT
152





regulator





154
FLJ44003
NP_660327.2
Unknown function
S24
GGTLRRSsSAPLIHG
153





155
FLJ44003
NP_660327.2
Unknown function
T51
LRTRRNStTIMSRHS
154





156
FNBP3
NP_060362.3
RNA processing
T921
ELEKRRRtLLEQLDD
155





157
FOXP4
NP_001012426.1
Unassigned
S292
LTSRRDSsSHEETPG
156





158
FRG2
NP_001005217.1
Unassigned
S149
DAHHRGHsRACTGHS
157





159
FRG2
NP_001005217.1
Unassigned
T153
RGHSRACtGHSKRHR
158





160
FRYL
NP_055845.1
Transcriptional
S1978
SSLARTRsLSSLREK
159





regulator





161
FXR1
NP_001013456.1
RNA processing
T511
RRSRRRRtDEDAVLM
160





162
GALNT8
NP_059113.1
Unassigned
T202
SIIQRAItSIINRTP
161





163
GAPVD1
NP_056450.2
Vesicle protein
S941
SSVRRPMsDPSWNRR
162





164
GATA-1
NP_002040.1
Transcriptional
S319
KGKKKRGsSLGGTGA
163





regulator





165
GATA-1
NP_002040.1
Transcriptional
S320
GKKKRGSsLGGTGAA
164





regulator





166
GRAMD1C
NP_060047.3
Receptor, channel,
T137
TFMTKEKtARLIPNA
165





transporter or cell





surface protein





167
GRID2IP
EAL23724.1
Adaptor/scaffold
T575
SFKGKMGtVSKSRAS
166





168
GRIN1
NP_443131.2
Cell
S895
EVRVRPGsALAAAVA
167





development/





differentiation





169
HACL1
NP_036392.2
Enzyme, misc.
T135
VEACRLYtKFSARPS
168





170
HARSL
NP_036340.1
Enzyme, misc.
T407
TKGEKVRtTETQVFV
169





171
HBP2
NP_064620.2
Transcriptional
T513
TPRKKVRtSSSGKGS
170





regulator





172
HDGF2
NP_116020.1
Secreted protein
S174
SKRARKAsSDLDQAS
171





173
HDGF2
NP_116020.1
Secreted protein
S175
KRARKASsDLDQASV
172





174
HECTD1
NP_056197.2
Ubiquitin
T1760
TKGGRRRtWDDDYVL
173





conjugating system





175
HER2
NP_004439.2
Protein kinase, Tyr
S1049
VHHRHRsSSTRSGGG
174





(receptor)





176
HER2
NP_004439.2
Protein kinase, Tyr
S1051
HHRHRSSsTRSGGGD
175





(receptor)





177
HGK
NP_004825.2
Protein kinase,
T187
RTVGRRNtFIGTPYW
176





Ser/Thr (non-





receptor)





178
HIPK1
NP_852003.1
Protein kinase,
S484
SPLRTTsSYNSLVPV
177





Ser/Thr (non-





receptor)





179
HIVEP1
NP_002105.2
Chromatin, DNA-
S779
TSLSRRGsIDSPKSY
178





binding, DNA repair





or DNA replication





protein





180
HMG2L1
NP_001003681.1
Chromatin, DNA-
S480
TTVKRKAsSSEGSMK
179





binding, DNA repair





or DNA replication





protein





181
HMGN4
NP_006344.1
Chromatin, DNA-
S76
PAKNRDAsTLQSQKA
180





binding, DNA repair





or DNA replication





protein





182
HP1BP3
NP_057371.2
Chromatin, DNA-
S520
STVIKKPsGGSSKKP
181





binding, DNA repair





or DNA replication





protein





183
HS3ST1
NP_005105.1
Enzyme, misc.
S135
KVPERVYsMNPSIRL
182





184
HS3ST1
NP_005105.1
Enzyme, misc.
S150
LLILRDPsERVLSDY
183





185
Hsn2
NP_998820.1
Unassigned
S61
AISQRRKsTSFLEAQ
184





186
Huntingtin
NP_002102.4
Cytoskeletal protein
S642
MSHCRQPsDSSVDKF
185





187
HYDIN
NP_116210.2
Unassigned
S2841
KSRDKYKsSLFPGNM
186





188
IKK-
NP_001093326.1
Protein kinase,
S178
EGRARAAsEQARQLE
187



gamma

regulatory subunit





189
IL 1RN
NP_000568.1
Vesicle protein
S15
RPSGRKSsKMQAFRI
188





190
ILK
NP_001014795.1
Protein kinase,
T181
RTRPRNGtLNKHSGI
189





Ser/Thr (non-





receptor)





191
IMPDH1
NP_000874.2
Enzyme, misc.
S85
MDRLRRAsMADYLIS
190



iso4





192
ITGB4
NP_000204.3
Receptor, channel,
S1483
HVPHRVLsTSSTLTR
191





transporter or cell





surface protein





193
ITGB4
NP_000204.3
Receptor, channel,
S1604
VFRVRAQsQEGWGRE
192





transporter or cell





surface protein





194
ITGB4
NP_000204.3
Receptor, channel,
T1797
GSLTRHVtQEFVSRT
193





transporter or cell





surface protein





195
ITGB4
NP_000204.3
Receptor, channel,
T1806
EFVSRTLtTSGTLST
194





transporter or cell





surface protein





196
ITIH3
NP_002208.3
Inhibitor protein
S388
RIPERSTsIVIMLTD
195





197
JMJD2B
NP_055830.1
Enzyme, misc.
T1065
AKRPRVGtPLATEDS
196





198
K14
NP_000517.2
Cytoskeletal protein
S44
GGSCRAPsTYGGGLS
197





199
K15
NP_002266.2
Cytoskeletal protein
S118
NLNDRLAsYLDKVRA
198





200
K19
NP_002267.2
Cytoskeletal protein
S93
NLNDRLAsYLDKVRA
199





201
K8
NP_002264.1
Cytoskeletal protein
S457
SSFSRTSsSRAVVVK
200





202
K8
NP_002264.1
Cytoskeletal protein
T26
AFSSRSYtSGPGSRI
201





203
KAB1
NP_001035864.1
Cell cycle regulation
S1107
SPRIRANsISRLSDS
202





204
KIAA0284
AAI12929.1
Cytoskeletal protein
S1220
TQTPRAGsSSRARSR
203





205
KIAA0284
AAI12929.1
Cytoskeletal protein
T1209
PKHTRSHtSTATQTP
204





206
KIAA0556
NP_056017.2
Unknown function
S1163
EEAMRRPsTADGEGD
205





207
KIAA0556
NP_056017.2
Unknown function
T1164
EAMRRPStADGEGDE
206





208
KIAA1107
NP_056052.2
Unknown function
T198
NVSGKPKtVTKSKTE
207





209
KIAA1107
NP_056052.2
Unknown function
T200
SGKPKTVtKSKTENG
208





210
KIAA1107
NP_056052.2
Unknown function
T204
KTVTKSKtENGDKAR
209





211
KIAA1219
NP_065069.1
Unassigned
S357
LGISRPRsDSAPPTP
210





212
KIAA1328
NP_065827.1
Cell cycle regulation
S533
PKPQRYPsREAGAWN
211





213
KIAA1468
NP_065905.2
Unassigned
S167
GAGGREPsTASGGGQ
212





214
KIAA1522
NP_065939.2
Unknown function
S410
QPRSRHPsSSSDTWS
213





215
KIAA1671
CAI17930.1
Unknown function
S244
RLKRRPVsAIFTESI
214





216
KIAA1671
CAI17930.1
Unknown function
S448
ISLFREDsTLALAVG
215





217
KIAA1706
NP_085139.2
Unassigned
S204
AERSRPPsTHTNGGL
216





218
KIAA1732
NP_054878.5
Unknown function
S1196
TVKAKIPsRQQEELP
217





219
KIAA1732
NP_054878.5
Unknown function
S2079
KEKRKRRsSLSPPSS
218





220
KIF11
NP_004514.2
Motor or contractile
T223
KGAAKRTtMTLMNA
219





protein





221
KIF13B
NP_056069.2
Motor or contractile
T1793
PEARRSAtLSGSATN
220





protein





222
KIF1B iso3
NP_904325.2
Motor or contractile
S1051
PPQLRWRsNSLNNGQ
221





protein





223
KIF1B iso3
NP_904325.2
Motor or contractile
S1053
QLRWRSNsLNNGQPK
222





protein





224
KIF5B
NP_004512.1
Motor or contractile
T451
QLVEKLKtQMLDQEE
223





protein





225
KNDC1
NP_689856.6
G protein or
S192
CRVCRSLsAVGRRVL
224





regulator





226
KNDC1
NP_689856.6
G protein or
S200
AVGRRVLsIESFGAL
225





regulator





227
LAD1
NP_005549.2
Adhesion or
T19
SSLARQRtLEDEEEQ
226





extracellular matrix





protein





228
lamin A/C
NP_733821.1
Cytoskeletal protein
T548
RKLVRSVtVVEDDED
227





229
LARP5
NP_055970.1
RNA processing
S731
KRLSREQsTPPKSPQ
228





230
LATS1
NP_004681.1
Protein kinase,
T17
YRQMRPKtFPASNYT
229





Ser/Thr (non-





receptor)





231
LGR5
NP_003658.1
Unassigned
S848
WTRSKHPsLMSINSD
230





232
LIN7B
NP_071448.1
Unassigned
S190
ARFEKMRsARRRQQH
231





233
LNP1
NP_001078920.1
Unassigned
S128
FRTKRSAsLGPESRK
232





234
LOC10012
XP_001722582.1
Unassigned
S89
DFLLKLSsVSICRKK
233



7983





235
LOC10013
XP_001725602.1
Unknown function
T143
RWQFRPTtDTLAVGT
234



0053





236
LOC10013
XP_001720876.1
Unknown function
T98
SPSPRVTtRAQDSEG
235



0745





237
LOC10013
XP_001717801.1
Unassigned
S2791
QLQDRKLsMLTPGIH
236



0981





238
LOC10013
XP_001714157.1
Unassigned
S123
RSPGRRYsHRLPAAT
237



2424





239
LOC10013
XP_001714157.1
Unassigned
S135
AATGRPLsAAAAAAA
238



2424





240
LOC10013
XP_001714216.1
Unassigned
S76
GGLSRLSsWPSDDIC
239



3182





241
LOC10013
XP_001715238.1
Unassigned
S236
RSYPRVFsLVPASPE
240



3676





242
LOC10013
XP_001714165.1
Unassigned
S157
HRKPRGRsRRAPQMP
241



3885





243
LOC10013
XP_001716491.1
Unassigned
S159
VAARAGsPPGPEYR
242



3930





244
LOC10013
XP_001714308.1
Unassigned
T25
PRPVRANtRPPQLPL
243



4195





245
LOC14410
NP_778228.2
Unknown function
T1013
GPESRYQtLPGRGLS
244



0





246
LOC64489
XP_943795.2
Unassigned
S277
RSTLKGPsTTEVPNT
245



3





247
L0C64879
XP_001718424.1
Unassigned
S84
RRRCRARsFSLPADP
246



1





248
L0C64879
XP_001718424.1
Unassigned
S86
RCRARSFsLPADPIL
247



1





249
L0C72884
XP_001715065.1
Unassigned
S25
SSTYRLSsSRTQPAW
248



6





250
L0C72884
XP_001715065.1
Unassigned
T20
MMKSRSStYRLSSSR
249



6





251
LOC81691
NP_112203.1
Unassigned
S649
YCFLKFKsFGSAQQA
250





252
LRRC58
NP_001093148.1
Unassigned
T256
VRFVRDLtYDPPTLL
251





253
LSM1
NP_055277.1
RNA processing
T129
LSIPRADtLDEY
252





254
LUC7L
NP_958815.1
Unassigned
S262
RLSRRSGsRTRDRRR
253





255
MAD2L1B
NP_001003690.1
Cell cycle regulation
S134
KHFYRKPsPQAEEML
254



P





256
MAGI3
EAW56562.1
Receptor, channel,
S1255
KRRPRDQsLSPSKGE
255





transporter or cell





surface protein





257
MAGI3
NP_690864.2
Receptor, channel,
T224
ESQRKRTtSVSKMER
256





transporter or cell





surface protein





258
MAP7
NP_003971.1
Adhesion or
S183
DPDRRSVsTMNLSKY
257





extracellular matrix





protein





259
MARCH7
NP_073737.1
Ubiquitin
S284
RTTRRLLsRIASSMS
258





conjugating system





260
MARCH7
NP_073737.1
Ubiquitin
S288
RLLSRIAsSMSSTFF
259





conjugating system





261
MARK1
NP_061120.3
Protein kinase,
T504
GSMARRNtYVCERTT
260





Ser/Thr (non-





receptor)





262
MARK3
NP_001122392.1
Protein kinase,
S474
IPERKKSsTVPSSNT
261





Ser/Thr (non-





receptor)





263
MAST2
NP_055927.2
Protein kinase,
S299
AMRPRSRsLSPGRSP
262





Ser/Thr (non-





receptor)





264
MAST2
NP_055927.2
Protein kinase,
S301
RPRSRSLsPGRSPVS
263





Ser/Thr (non-





receptor)





265
MAST3
NP_055831.1
Protein kinase,
S153
SPRLRPRsRSLSPGR
264





Ser/Thr (non-





receptor)





266
MAST3
NP_055831.1
Protein kinase,
S155
RLRPRSRsLSPGRAT
265





Ser/Thr (non-





receptor)





267
MAST3
NP_055831.1
Protein kinase
S157
RPRSRSLsPGRATGT
266





Ser/Thr (non-





receptor)





268
MATN3
NP_002372.1
Unassigned
T104
LEFTKVKtFVSRIID
267





269
matrin 3
NP_061322.2
RNA processing
T150
LQLKRRRtEEGPTLS
268





270
MBOAT2
NP_620154.2
Unassigned
T474
KTQRRKNtHENIQLS
269





271
MBTD1
NP_060113.2
Transcriptional
T460
LTPPRGYtKLPFKWF
270





regulator





272
MCRS1
NP_006328.2
RNA processing
S98
SSEKKKVsKAPSTPV
271





273
ME3
NP_006671.2
Unassigned
S545
LSTIRDVsLRIAIKV
272





274
MED4
NP_054885.1
Unassigned
S32
STRERLLsALEDLEV
273





275
MEF2D
NP_005911.1
Transcriptional
T517
VKRMRLDtWTLK
274





regulator





276
MICB
NP_005922.2
Receptor, channel,
T101
QDLRRTLtHIKDQKG
275





transporter or cell





surface protein





277
MLL2
NP_003473.3
Transcriptional
S1331
RGRARLKsTASSIET
276





regulator





278
MLL2
NP_003473.3
Transcriptional
S4534
KASDRLVsSRKKLRK
277





regulator





279
MLL3
NP_733751.2
Enzyme, misc.
S3788
LKNKKSSsLLNQKPE
278





280
MRPS33
NP_444263.1
Translation
S38
MKVVKLFsELPLAKK
279





281
MTBP
NP_071328.2
Ubiquitin
T687
SRLIRYEtQTTCTRE
280





conjugating system





282
MVP
NP_005106.2
RNA processing
S864
PLGRRVAsGPSPGEG
281





283
MYO18A
NP_510880.2
Cytoskeletal protein
S1068
ASSRRVSsSSELDLP
282





284
MYO6A
NP_001135967.1
Motor or contractile
S1504
NDDQKVRsLLTSTIN
283





protein





285
MYO9A
NP_008832.2
Motor or contractile
S889
HKKKKPPsISAQFQA
284





protein





286
MYPN
NP_115967.2
Cytoskeletal protein
S1047
PIRSRLTsAGQSHRG
285





287
NAP1L1
NP_631946.1
Chromatin, DNA-





binding, DNA repair
T39
KLKARQLtVQMMQNP
286





or DNA replication





protein





288
NAP5
NP_997246.2
Unassigned
S1085
MTSSKSVsPGRKGQL
287





289
nav1
NP_065176.2
Adhesion or





extracellular matrix
S594
SDAKKPPsGIARPST
288





protein





290
NAV2
NP_660093.2
Unknown function
S1681
NMTIRLQsLTMTAEQ
289





291
NDRG4
NP_075061.1
Unassigned
S336
LARSRTAsLTSASSV
290





292
NEB
NP_004534.2
Cytoskeletal protein
S3314
LHIAKVQsDREYKKD
291





293
NEDD4L
AAM76729.1
Ubiquitin
S367
APAGRARsSTVTGGE
292





conjugating system





294
NEO1
NP_002490.2
Receptor, channel,
S1299
SLSDRANsTESVRNT
293





transporter or cell





surface protein





295
NHS
NP_938011.1
Adhesion or
S1173
KSVSRQYsTEDTILS
294





extracellular matrix





protein





296
NIPBL
NP_597677.2
Chromatin, DNA-
S874
ERKHRHEsGDSRERP
295





binding, DNA repair





or DNA replication





protein





297
NLRP7
NP_996611.2
Apoptosis
T734
FIGKKTLtHLTLAGH
296





298
NOT1
NP_057368.3
Transcriptional
T1053
TTVAKTVtVTRPTGV
297





regulator





299
NPAT
NP_002510.2
Cell cycle regulation
T1213
EMTKKQGtSSNNKNV
298





300
NUDT7
NP_001099133.1
Enzyme, misc.
S236
KVHKKATsRL
299





301
NuMA-1
NP_006176.2
Cell cycle regulation
T1812
RSARRRTtQIINITM
300





302
NUP210L
NP_997191.2
Unknown function
T1054
NYILRATtIGQTTLV
301





303
ODZ3
NP_001073946.1
Receptor, channel,
T1660
GDMDKAItVDIESSS
302





transporter or cell





surface protein





304
OIP5
NP_009211.1
Unassigned
T14
RHRSRCAtPPRGDFC
303





305
OXNAD1
NP_612390.1
Enzyme, misc.
T48
IMKSKRKtDHMERTA
304





306
P15RS
NP_060640.2
Unassigned
S37
LIHHRKHsRPIVTVW
305





307
PAC3-1
NP_060496.2
Adaptor/scaffold
S320
KTRRKLTsTSAITRQ
306





308
PAP-alpha
NP_116021.2
RNA processing
S617
PTVSRVVsSTRLVNP
307





309
PCDH10
NP_116586.1
Calcium-binding
S897
DRPRRVNsSAFQEAD
308





protein





310
PCDHGA4
NP_114442.1
Unassigned
S470
ENNPRGAsILSMTAQ
309





311
PDE1B
NP_000915.1
Enzyme, misc.
T144
RMFRRTYtSVGPTYS
310





312
PDE1C
NP_005011.1
Enzyme, misc.
S46
KTSQRLRsLVKQLER
311





313
periplakin
NP_002696.3
Cytoskeletal protein
S887
LQRNRPDsGVEEAWK
312





314
PERK
NP_004827.4
Protein kinase,
S555
PHRQRKEsETQCQTE
313





Ser/Thr (non-





receptor)





315
PIK3CA
NP_006209.2
Kinase (non-
T313
SYSRRIStATPYMNG
314





protein)





316
PIP5K
AAR19397.1
Kinase (non-
S1177
FAHSKDAsSTSSGKS
315





protein)





317
PIP5K
NP_055855.2
Kinase (non-
S1474
KMQARLMsSSVDTPQ
316





protein)





318
PIP5K
NP_055855.2
Kinase (non-
S845
TIKLRGGsDYELARV
317





protein)





319
PKD1
NP_002733.2
Protein kinase,
S421
KHTKRKSsTVMKEGW
318





Ser/Thr (non-





receptor)





320
plakophilin
NP_001005242.2
Adhesion or
T248
LTYPRPGtSRSMGNL
319



2

extracellular matrix





protein





321
plakophilin
NP_009114.1
Adhesion or
S134
LSCSRRLsSAHNGGS
320



3

extracellular matrix





protein





322
POLQ
NP_955452.3
Enzyme, misc.
S15
GKRRRSEsGSDSFSG
321





323
POM121
NP_742017.1
Receptor, channel,
S126
HLNKRSRsSSMSSLT
322





transporter or cell





surface protein





324
PPP5C
NP_006238.1
Phosphatase
S105
GYYRRAAsNMALGKF
323





325
PROM2
NP_653308.1
Receptor, channel,
S814
RPIRKRLsSTSSEET
324





transporter or cell





surface protein





326
PROM2
NP_653308.1
Receptor, channel,
S815
PIRKRLSsTSSEETQ
325





transporter or cell





surface protein





327
PRR14
NP_076936.1
Unknown function
T363
RPRPRRHtVGGGEMA
326





328
PRSS15
NP_004784.2
Protease
T899
VGGIKEKtIAAKRAG
327





329
PTMS
NP_002815.3
Cell cycle regulation
T96
ADPKRQKtENGASA
328





330
PTOV1
NP_059128.2
Unassigned
S53
PPRIRARsAPPMEGA
329





331
PWP2H
NP_005040.2
Unassigned
T256
DQEGDRETtIRGKAT
330





332
PWWP2
NP_001092107.1
Unknown function
S206
RARRRLGsGPDRELR
331





333
PXN iso3
AAC05175.1
Adhesion or
S313
AARHRTPsLRSPDQP
332





extracellular matrix





protein





334
Q6EEV4
NP_001018113.1
Unassigned
S50
APRLRAPsSRGLGAA
333





335
R3HCC1
CAB43396.1
Unknown function
S56
FLLQKQLsKVLLFPP
334





336
Rab1A
NP_004152.1
G protein or
T75
AGQERFRtITSSYYR
335





regulator





337
Rap1GAP
NP_002876.1
G protein or
S439
KRVIRSRsQSMDAMG
336





regulator





338
Rap1GAP
NP_002876.1
G protein or
S441
VIRSRSQsMDAMGLS
337





regulator





339
RapGEF6
NP_057424.2
G protein or
S1094
KKRARRSsLLNAKKL
338





regulator





340
RbBP6
NP_008841.2
Cell cycle regulation
T1256
VRRKVtGTEGSSSTL
339





341
RBMX2
NP_057108.2
Unassigned
S314
RSRERESsNPSDRWR
340





342
REPS1
NP_114128.3
Vesicle protein
S536
VTRQRSHsGTSPDNT
341





343
REPS2
NP_004717.2
G protein or
S461
KARPRSRsYSSTSIE
342





regulator





344
RGS3
NP_570613.2
G protein or
T660
QNSLRRRtHSEGSLL
343





regulator





345
Rin1
NP_004283.2
G protein or
S291
CQLLRREsSVGYRVP
344





regulator





346
RINL
NP_940847.1
G protein or
S407
DRAPRGLsSEARASL
345





regulator





347
RIP3
NP_055949.2
G protein or
S265
GRKVRVEsGYFSLEK
346





regulator





348
RLTPR
NP_001013860.1
Unassigned
S1268
SIKSRTHsVSADPSC
347





349
ROS
NP_002935.2
Protein kinase, Tyr
T1274
KIHNRNStIISFSVY
348





(receptor)





350
RP5-
NP_068752.2
Unassigned
S113
LGRPRPHsAPSLGTS
349



1077B9.4





351
RPA40
NP_976035.1
Transcriptional
T282
VANPRLDtFSREIFR
350





regulator





352
SAMD4
NP_056404.2
RNA processing
S167
QNRGRSDsVDYGQTH
351





353
SAPAP4
NP_892118.1
Receptor, channel,
S127
APKRKLSsIGIQVDC
352



iso2

transporter or cell





surface protein





354
SATB1
NP_001124482.1
Transcriptional
S47
LGRGRLGsTGAKMQG
353





regulator





355
sciellin
NP_659001.1
Adaptor/scaffold
S68
WLNRHNsHDALDRK
354





356
SCYL1
NP_001041683.1
Protein kinase,
S737
STQPRPDsWGEDNWE
355





Ser/Thr (non-





receptor)





357
SEMA4B
NP_064595.2
Receptor, channel,
S809
ESEKRPLsIQDSFVE
356





transporter or cell





surface protein





358
SEMA6A
NP_065847.1
Receptor, channel,
S988
VTVSRQPsLNAYNSL
357





transporter or cell





surface protein





359
SENP7
NP_001070671.1
Unassigned
S11
RKLGRRPsSSEIITE
358





360
SEPT14
NP_997249.2
Unassigned
T366
RVKEKEAtFKEAEKE
359





361
SERPINB8
NP_942130.1
Inhibitor protein
T50
FMGAKGStAAQMSQA
360





362
SGSM2
NP_001091979.1
G protein or
S231
GIRKRHSsGSASEDR
361





regulator





363
SH2BP1
NP_055448.1
Transcriptional
S1097
RKRRPsGSEQSDNES
362





regulator





364
SH3MD2
NP_065921.2
Adaptor/scaffold
T684
EPSGRIVtVLPGLPT
363





365
SHRM
NP_065910.3
Cytoskeletal protein
S403
RHRERPSsWSSLDQK
364





366
SHROOM1
NP_597713.1
Cytoskeletal protein
S308
ASRSRSAsGEVLGSW
365





367
SHROOM4
NP_065768.2
Cytoskeletal protein
S664
SMMLRARsSECLSQA
366





368
Sin1
NP_077022.1
Adaptor/scaffold
S84
DFGIRRRsNTAQRLE
367





369
SIPA1L1
NP_056371.1
G protein or
S1632
SLPLRRPsYTLGMKS
368





regulator





370
SIPA1L1
NP_056371.1
G protein or
S176
RIRQRSNsDITISEL
369





regulator





371
SIPA1L3
NP_055888.1
G protein or
S170
LPLRHRsSSEITLSE
370





regulator





372
SIX2
NP_058628.2
Unassigned
T158
PREKRELtEATGLTT
371





373
SKI
NP_003027.1
Unassigned
T103
RSTERCEtVLEGETI
372





374
skMLCK
NP_149109.1
Protein kinase,
S586
ANRFKKIsSSGALMA
373





Ser/Thr (non-





receptor)





375
SLC12A7
NP_006589.2
Receptor, channel,
S997
KYRSRDTsLSGFKDL
374





transporter or cell





surface protein





376
SLC19A1
NP_919231.1
Receptor, channel,
T222
DDRGRCEtSASELER
375





transporter or cell





surface protein





377
SLC4A2
NP_003031.3
Receptor, channel,
T257
ALLPRVPtDEIEAQT
376





transporter or cell





surface protein





378
SLC4A4
NP_003750.1
Receptor, channel,
S1034
TFLERHTsC
377





transporter or cell





surface protein





379
SNX3
NP_003786.1
Vesicle protein
T48
VGRGRFTtYEIRVKT
378





380
SNX3
NP_003786.1
Vesicle protein
T55
TYEIRVKtNLPIFKL
379





381
S0X4
NP_003098.1
Transcriptional
S136
RPRKKVKsGNANSSS
380





regulator





382
SPATA13
NP_694568.1
Unknown function
S26
ARRRRPIsVIGGVSL
381





383
SPATS2
NP_075559.2
Unknown function
S386
STRSRCSsVTSVSLS
382





384
SPECC1
NP_001028727.1
Unknown function
S70
TKHLRTPsTKPKQEN
383





385
SPECC1L
NP_056145.1
Cell cycle regulation
T833
GLSRRSStSSEPTPT
384





386
SPG20
NP_001135766.1
Unknown function
S358
PGRTRPSsDQLKEAS
385





387
SPTA1
NP_003117.2
Cytoskeletal protein
S1912
ALNEKTPsLAKAIAA
386





388
Src
NP_005408.1
Protein kinase, Tyr
T246
GLCHRLTtVCPTSKP
387





(non-receptor)





389
SRm300
NP_057417.3
RNA processing
S1852
PPTSRKRsRSRTSPA
388





390
SRm300
NP_057417.3
RNA processing
S827
SPPPKQKsKTPSRQS
389





391
SRm300
NP_057417.3
RNA processing
S831
KQKSKTPsRQSHSSS
390





392
ST0X2
NP_064610.1
Unknown function
T8
MKKTRSTtLRRAWPS
391





393
STXBP5L
NP_055795.1
Unassigned
S818
NSYNRSRsSSISSID
392





394
STXBP5L
NP_055795.1
Unassigned
S820
YNRSRSSsISSIDKD
393





395
SUSD2
NP_062547.1
Receptor, channel,
S405
RTPPRVPsMSHWLYD
394





transporter or cell





surface protein





396
synergin,
NP_009178.3
Adaptor/scaffold
S1098
EQPFRDRsNTLNEKP
395



gamma





397
SYT17
NP_057608.2
Unassigned
S110
YSLTRRIsSLESRRP
396





398
SYTL5
NP_620135.1
Unknown function
T195
LSKFRSAtRGEIITP
397





399
TACC2
NP_996744.2
Cell cycle regulation
S2177
ASETKTEsAKTEGPS
398





400
TACC2
NP_996744.2
Cell cycle regulation
S712
EGLGRMEsFLTLESE
399





401
TAF11
NP_005634.1
Translation
S74
TTVEREDsSLLNPAA
400





402
TAZ
NP_056287.1
Transcriptional
S311
PYHSREQsTDSGLGL
401





regulator





403
TAZ
NP_056287.1
Transcriptional
S66
GSHSRQSsTDSSGGH
402





regulator





404
TBC1D15
NP_073608.2
G protein or
S274
STHQRPPsEMADFLS
403





regulator





405
TBC1D4
NP_055647.2
G protein or
T749
GRKRtSSTCSNESLS
404





regulator





406
TCEB1P3
XP_001717253.1
Unknown function
S71
EIPSRVLsKVCTYFM
405





407
TCF20
NP_005641.1
Transcriptional
S825
GPWERKSsSTAPEMK
406





regulator





408
TCF20
NP_005641.1
Transcriptional
S826
PWERKSSsTAPEMKQ
407





regulator





409
tensin 3
NP_073585.8
Adaptor/scaffold
S941
GPGQRREsSSSAERQ
408





410
TFII-I
NP_127494.1
Transcriptional
T764
VPFRRPStFGIPRLE
409





regulator





411
TFIIF-
NP_002087.2
Transcriptional
S415
LEQGKRVsEMPAAKR
410



alpha

regulator





412
TFIIIC-
NP_001511.2
Transcriptional
S1063
PRVRKNSsTDQGSDE
411



alpha

regulator





413
TIMELESS
NP_003911.1
Transcriptional
T804
TAVVREMtEGYGSLD
412





regulator





414
TIP20
NP_001002836.2
Unknown function
T89
TRHQRTHtGERPNAC
413





415
TMCC1
NP_001017395.2
Unassigned
S156
MQSGRPRsSSTTDAP
414





416
TMCC1
AAH39859.1
Unassigned
S158
SGRPRSSsTTDAPTG
415





417
TMCC1
NP_001017395.2
Unassigned
S63
FQHQRRRsSVSPHDV
416





418
TNFRSF21
NP_055267.1
Receptor, channel,
S562
EPLLRCDsTSSGSSA
417





transporter or cell





surface protein





419
TNFSF8
NP_001235.1
Secreted protein
S67
LVVQRTDsIPNSPDN
418





420
TOMM70A
NP_055635.3
Receptor, channel,
S434
CIRLRPEsALAQAQK
419





transporter or cell





surface protein





421
TRIM3
NP_006449.2
Cytoskeletal protein
S455
KAVRRPSsMYSTGGK
420





422
Trio
NP_009049.2
Protein kinase,
S1785
TSPVRRLsSGKADGH
421





Ser/Thr (non-





receptor)





423
Trio
NP_009049.2
Protein kinase,
S1786
SPVRRLSsGKADGHV
422





Ser/Thr (non-





receptor)





424
TRIP12
NP_004229.1
Ubiquitin
T1377
AEDEREStDDESNPL
423





conjugating system





425
Trpc1
NP_003295.1
Receptor, channel,
S718
SMRQKMQsTDQATVE
424





transporter or cell





surface protein





426
TRPC6
NP_004612.2
Receptor, channel,
S13
AFGPRRGsSPRGAAG
425





transporter or cell





surface protein





427
TRPC6
NP_004612.2
Receptor, channel,
S14
FGPRRGSsPRGAAGA
426





transporter or cell





surface protein





428
TSP50
NP_037402.1
Unassigned
T16
ARGQRPRtSAPSRAG
427





429
TTC39B
NP_689787.1
Unknown function
S54
PRQRGAsTVSSSSST
428





430
UNKL
EAW85660.1
Unassigned
S737
GRGERDSsQRPLRPQ
429





431
UNKL
EAW85660.1
Unassigned
T751
QTTHRQDtRPVPS
430





432
USP24
NP_056121.1
Protease
S1212
GIRNRLSsSGSNCSS
431





433
USP34
NP_055524.3
Protease
S1458
NRRIRREsTGSYSDL
432





434
USP43
NP_694942.3
Unassigned
S844
ESRRRPRsTSQSIVS
433





435
VPRBP
NP_055518.1
Ubiquitin
S255
KTSSRVNsTTKPEDG
434





conjugating system





436
VPS26A
NP_004887.2
Vesicle protein
S269
RDVNKKFsVRYFLNL
435





437
WDHD1
NP_001008397.1
Chromatin, DNA-
S251
SGRPRQRsHILEDDE
436





binding, DNA repair





or DNA replication





protein





438
WDR20
NP_851819.1
Unknown function
S223
FGRDRANsTQSRLSK
437





439
WDR37
NP_054742.2
Unknown function
S30
LSIRRTNsSEQERTG
438





440
WDR43
NP_055946.1
Unknown function
T312
GYCKKPLtSNCTIQI
439





441
WFDC9
NP_671731.1
Unassigned
T59
KRCTKIMtCVRPNHT
440





442
WNT1
NP_005421.1
Adhesion or
S144
HSVARSCsEGSIESC
441





extracellular matrix





protein





443
ZAP
NP_078901.3
Chromatin, DNA-
S310
QDRARPPsGSSKATD
442





binding, DNA repair





or DNA replication





protein





444
ZFP64
NP_060667.2
Transcriptional
S334
KATLRKHsRVHQSEH
443





regulator





445
ZHX2
NP_055758.1
Transcriptional
T220
EGTARLVtDTAEILS
444





regulator





446
ZNF185
NP_009081.2
Chromatin, DNA-
S153
PYNIRRSsTSGDTEE
445





binding, DNA repair





or DNA replication





protein





447
ZNF185
NP_009081.2
Chromatin, DNA-
S64
SGRSRATsFSSAGEV
446





binding, DNA repair





or DNA replication





protein





448
ZNF185
NP_009081.2
Chromatin, DNA-
T154
YNIRRSStSGDTEEE
447





binding, DNA repair





or DNA replication





protein





449
ZNF197
NP_008922.1
Transcriptional
S284
ADSHKGTsKRLQGSV
448





regulator





450
ZNF24
NP_008896.2
Transcriptional
T330
INHQRIHtGEKPYEC
449





regulator





451
ZNF395
NP_061130.1
Transcriptional
S449
PVRSRSLsFSEPQQP
450





regulator





452
ZNF436
NP_085137.1
Unassigned
S456
TECEKSFsRSSALIK
451





453
ZNF511
NP_665805.2
Chromatin, DNA-
S230
IYRHRIPsTICFGQG
452





binding, DNA repair





or DNA replication





protein





454
ZNF514
NP_116177.1
Transcriptional
S78
HSDWKRRsKSKESMP
453





regulator





455
ZNF514
NP_116177.1
Transcriptional
S80
DWKRRSKsKESMPSW
454





regulator





456
ZNF585A
NP_954577.1
Transcriptional
T238
IAHRRIHtGEKPYEC
455





regulator





457
ZNF609
NP_055857.1
Unknown function
S100
SKSGKDTsKPTPGTS
456





458
ZNF609
NP_055857.1
Unknown function
S95
SKSKRSKsGKDTSKP
457





459
ZNF66
XP_001719810.1
Unassigned
T595
TTHKRIHtADKPYKC
458





460
ZO1
NP_003248.3
Adaptor/scaffold
S978
ADSLRTPsTEAAHIM
459





461
ZSCAN21
NP_666019.1
Unassigned
T467
SKHQRVHtGEGEAP
460





462
ZZEF1
NP_055928.3
Cell cycle regulation
S1537
RGRLRLLsFRSMEEA
461









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.


ANKRD25, phosphorylated at S540, is among the proteins listed in this patent. ANKRD25 has strong similarity to ankyrin repeat domain 25 (mouse Ankrd25), is mainly involved in regulating cell proliferation and cell cycle activities, contains an ankyrin repeat, and may mediate protein-protein interactions. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


C17orf28, phosphorylated at S670, is among the proteins listed in this patent. C17orf28, Chromosome 17 open reading frame 28 (downregulated in multiple cancer 1), member of a class of inside out membrane proteins, a putative integral membrane protein that is downregulated in many cancer cell lines. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


C9orf140, phosphorylated at T219, is among the proteins listed in this patent. C9orf140, Protein of unknown function, has high similarity to uncharacterized mouse 2010317E24Rik. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


capicua, phosphorylated at S299, is among the proteins listed in this patent. capicua, Capicua homolog, a member of a subfamily of the HMG box superfamily that complexes with ATXN1, may play a role in central nervous system development; fusion of the corresponding gene with DUX4 gene is associated with Ewing-like sarcomas. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


CDKAL1, phosphorylated at T43, is among the proteins listed in this patent. CDKAL1, Member of the UPF0004 uncharacterized protein family, contains a radical SAM superfamily domain and a TRAM domain, has weak similarity to rat Cdk5rap1, which binds the rat Cdk5 activator (rat Nck5a) and inhibits rat Cdk5. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


Dbf4, phosphorylated at T273, is among the proteins listed in this patent. Dbf4, Activator of S phase kinase, binds to and activates kinase activity of CDC7 (CDC7L1), required for the initiation of DNA replication at the G1 to S transition. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


FAM117A, phosphorylated at S145, is among the proteins listed in this patent. FAM117A, Protein of unknown function, has strong similarity to uncharacterized mouse 5730593F17Rik. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.). FXR1, phosphorylated at T511, is among the proteins listed in this patent. FXR1, Fragile X mental retardation autosomal homolog 1, binds FMR1 and associates with mRNPs and with 60S ribosomal subunits and may play a role in neuronal ribosome and RNA metabolism; autoantigen to FXR1 is expressed in a systemic scleroderma patient. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Systemic Scleroderma (J Biol Chem 1998 Jul. 3; 273(27):17122-7). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


GRIN1, phosphorylated at S895, is among the proteins listed in this patent. GRIN1, Protein with high similarity to mouse Gprin1, which binds to activated G(z)alpha, G(O)alpha, and G(i) alpha subunits, induces neurite-like extensions and activates CDC42, may act as G alpha subunit effector in regulating neurite outgrowth in growth cones. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


HDGF2, phosphorylated at S174, is among the proteins listed in this patent. HDGF2, Protein containing a PWWP domain, has low similarity to PC4 and SFRS1 interacting protein 2 (human PSIP1), which is a transcriptional co-activator that functions in the stress response, mRNA splicing, positive regulation of cell growth, and transcription. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


HGK, phosphorylated at T187, is among the proteins listed in this patent. HGK, Mitogen-activated protein kinase kinase kinase kinase 4, a serine-threonine kinase that binds RAP2A and hGBP3 (SPG3A), activates JNK activity, involved in T cell activation. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


ILK, phosphorylated at T181, is among the proteins listed in this patent. ILK, Integrin linked kinase, regulates integrin-mediated cell adhesion, migration, and motility, involved in antiapoptosis and actin cytoskeleton reorganization, acts as a tumor suppressor, aberrant expression correlates with multiple neoplasms and diabetes. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Prostatic Neoplasms, Ewing's Sarcoma, Colonic Neoplasms, Primitive Neuroectodermal Tumors (Br J Cancer 2003 Jun. 2; 88(11):1755-62.). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


KAB1, phosphorylated at S1107, is among the proteins listed in this patent. It is a potential marker for mature centrioles. (Mol Biol Cell. 2005 March; 16(3):1095-107. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, Mass. Human PSD™, Biobase Corporation, Beverly, Mass.).


lamin A/C, phosphorylated at T548, is among the proteins listed in this patent. lamin A/C, Lamin A, nuclear lamina structural protein, maintains chromatin and nuclear structure, cleaved during apoptosis; mutations cause progeria, Emery-Dreifuss muscular dystrophy, familial partial lipodystrophy, metabolic syndrome and dilated cardiomyopathy. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Emery-Dreifuss Muscular Dystrophy (Am J Hum Genet. 2000 April; 66(4):1407-12.). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


LSM1, phosphorylated at T129, is among the proteins listed in this patent. LSM1, LSM1 homolog U6 small nuclear RNA associated (S. cerevisiae), regulates cell growth and cell cycle, may maintain transformed status of cancer cells, increased expression correlates with numerous neoplasms and decreased expression with prostatic neoplasms. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Prostatic Neoplasms (Br J Cancer 2002 Mar. 18; 86(6):940-6.). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


MVP, phosphorylated at 5864, is among the proteins listed in this patent. MVP, Major vault protein, a subunit of the ribonucleoprotein particle vault, acts as a PTPN11 and MAPK1 scaffold protein, mediates multidrug resistance via nucleocytoplasmic transport, overexpressed in several types of tumors. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Stomach Neoplasms, Lung Neoplasms (Anticancer Res 1998 Jul-Aug; 18(4C):3077-80.). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


MY018A, phosphorylated at S1068, is among the proteins listed in this patent. MY018A, Myosin XVIIIA, a putative myosin, contains a PDZ domain. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


Abi-1, phosphorylated at S319, is among the proteins listed in this patent. Abi-1, Ab1-interactor 1, binds SH3 domains of EPS8, ABL1, and SPTA1, acts in pinocytosis, regulates actin nucleator protein WASF2, inhibits cell proliferation; fusion of gene to SSH3BP1 or MLL is linked to acute myelogenous leukemia. (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


CD2AP, phosphorylated at T229, is among the proteins listed in this patent. CD2AP, CD2-associated protein, a putative adaptor protein that associates with ASAP1 (DDEF1) and functions in organization of the actin cytoskeleton and may also act in cell migration; gene mutation is associated with focal segmental glomerulosclerosis. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Nephrotic Syndrome (Kidney Int 2004 September; 66(3):945-54.). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


FGFR2, phosphorylated at 5472, is among the proteins listed in this patent. FGFR2, Fibroblast growth factor receptor 2, acts in induction of apoptosis, skeletal development, cell migration and differentiation, aberrantly expressed in several types of cancer; mutations cause acrocephalosyndactylia, craniosynostoses. This protein has potential diagnostic and/or therapeutic implications based on association with the following diseases: Craniofacial Dysostosis, Acrocephalosyndactylia (Am J Hum Genet. 1996 March; 58(3):491-8). (PhosphoSitePlus®, Cell Signaling Technology, Danvers, MA. Human PSD™, Biobase Corporation, Beverly, Mass.).


The invention also provides peptides comprising a novel phosphorylation site of the invention. In one particular embodiment, the peptides comprise any one of the amino acid sequences as set forth in SEQ ID NOs: 1-461, 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.


Various methods that are well known in the art can be used to eliminate a phosphorylation site. For example, the phosphorylatable serine and/or threonine 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 serine and/or threonine may be deleted. Residues other than the serine and/or threonine may also be modified (e.g., delete or mutated) if such modification inhibits the phosphorylation of the serine and/or threonine residue. For example, residues flanking the serine and/or threonine may be deleted or mutated, so that a kinase cannot 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 serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine 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 SL, 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-Isotope 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 for basophilic Ser/Thr kinase signaling related diseases, or as potential therapeutic agents for treating basophilic Ser/Thr kinase related diseases.


The peptides may be of any length, typically six to fifteen amino acids. The novel serine and/or threonine 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 an adaptor/scaffold proteins, enzyme/non-protein kinase/phoshpatase proteins, Ser/Thr (non-receptor) protein kinases, vesicle proteins, g proteins or regulator proteins, chromatin or DNA binding/repair/replication proteins, receptor/channel/transporter/cell surface proteins, RNA processing proteins, cytoskeletal proteins, transcriptional regulators and translation proteins.


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 includes the phosphorylatable serine and/or threonine. 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 includes the phosphorylatable serine and/or threonine.


In certain embodiments, the peptide or AQUA peptide comprises any one of SEQ ID NOs: 1-461, 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, 15 N, 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 MS2 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 461 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 ECLARSAsTESGFHN (SEQ ID NO: 19), wherein “s” corresponds to phosphorylatable serine 80 of APBA1) 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., APBA1) 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 NQRPRTHsGSSGGSG (SEQ ID NO: 5), wherein s (Ser 240) is phosphoserine, and wherein P=labeled proline (e.g., 14C)) is provided for the quantification of phosphorylated (or unphosphorylated) form of Abi-1 iso3 (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: 5 (a trypsin-digested fragment of Abi-1 iso3, with a Ser 240 phosphorylation site) may be used to quantify the amount of phosphorylated Abi-1 iso3 in a biological sample, e.g., 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 basophilic Ser/Thr kinase signaling related disease (including, among many others, cancer and diabetes) and pathways. Peptides and AQUA peptides of the invention may also be used for identifying diagnostic/bio-markers of basophilic Ser/Thr kinase signaling related disease (including, among many others, diabetes and cancer), 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 serine and/or threonine 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 serine and/or threonine 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 adaptor/scaffold proteins, enzyme/non-protein kinase/phoshpatase proteins, Ser/Thr (non-receptor) protein kinases, vesicle proteins, g proteins or regulator proteins, chromatin or DNA binding/repair/replication proteins, receptor/channel/transporter/cell surface proteins, RNA processing proteins, cytoskeletal proteins, transcriptional regulators and translation proteins.


It shall be understood that if a given sequence disclosed herein comprises more than one amino acid that can be modified, this invention includes sequences comprising modifications at one or more of the amino acids. In one non-limiting example, where the sequence is: VCYTVINHIPHQRSSLSSNDDGYE, and the *symbol indicates the preceding amino acid is modified (e.g., a Y* indicates a modified (e.g., phosphorylated) tyrosine residues, the invention includes, without limitation,











VCY*TVINHIPHQRSSLSSNDDGYE,







VCYT*VINHIPHQRSSLSSNDDGYE,







VCYTVINHIPHQRS*SLSSNDDGYE,







VCYTVINHIPHQRSS*LSSNDDGYE,







VCYTVINHIPHQRSSLS*SNDDGYE,







VCYTVINHIPHQRSSLSS*NDDGYE,






VCYTVINHIPHQRSSLSSNDDGY*E, as well as sequences comprising more than one modified amino acid including











VCY*T*VINHIPHQRSSLSSNDDGYE,







VCY*TVINHIPHQRS*SLSSNDDGYE,







VCY*TVINHIPHQRSSLSSNDDGY*E,






VCY*T*VINHIPHQRS*S*LS*S*NDDGY*E, etc. Thus, an antibody of the invention may specifically bind to VCY*TVINHIPHQRSSLSSNDDGYE, or may specifically bind to VCYT*VINHIPHQRSSLSSNDDGYE, or may specifically bind to VCYTVINHIPHQRS*SLSSNDDGYE, and so forth. In some embodiments, an antibody of the invention specifically binds the sequence comprising a modification at one amino acid residues in the sequence. In some embodiments, an antibody of the invention specifically binds the sequence comprising modifications at two or more amino acid residues in the sequence.


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 includes the phosphorylatable serine and/or threonine.


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 serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine phosphorylation site shown by a lower case “s” or “t” in Column E of Table 1. Such peptides include any one of SEQ ID NOs: 1-461.


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, 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 1×10−7M or less. In other embodiments, the antibody binds with a KD of 1×10−8 M, 1×10−9 M, 1×10−1° M, 1×10−11M, 1×10−12M or less. In certain embodiments, the KD is 1 pM 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 VHIIII 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 serine and/or threonine 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) stimulating metabolic processes in cellular responses to basophilic Ser/Thr kinase signaling 2) mimicking the cellular responses to basophilic Ser/Thr kinase signaling, 3) providing co-stimulatory signals that are capable of reversing or relieving basophilic Ser/Thr kinase signaling hypo-responsiveness 4) regulating cellular responses to basophilic Ser/Thr kinase signaling 5) discovering markers for normal and abnormal basophilic Ser/Thr kinase signaling responsiveness 6) 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 serine and/or threonine phosphorylation site of the invention or two or more different antibodies or antigen-binding portions all of which are specific for the same novel serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine. 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 an adaptor/scaffold proteins, enzyme/non-protein kinase/phoshpatase proteins, Ser/Thr (non-receptor) protein kinases, vesicle proteins, g proteins or regulator proteins, chromatin or DNA binding/repair/replication proteins, receptor/channel/transporter/cell surface proteins, RNA processing proteins, cytoskeletal proteins, transcriptional regulators and translation proteins. In some embodiments, the peptide immunogen is an AQUA peptide, for example, any one of SEQ ID NOS: 1-461.


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 adhesion or extracellular matrix protein protein phosphorylation site in SEQ ID NO: 10 shown by the lower case “s” in Table 1 may be used to produce antibodies that specifically bind the novel serine phosphorylation site.


When the above-described methods are used for producing polyclonal antibodies, following immunization, the polyclonal antibodies that are 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 serine and/or threonine 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 phosphoserine and/or threonine 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. Adhering 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 serine and/or threonine 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-Cu1-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 Perris, 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 a particular basophilic Ser/Thr kinase signaling related disease in a subject, wherein the disease 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 maybe 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 basophilic Ser/Thr kinase signaling related disease (including, among many others, diabetes and cancer) 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 serine and/or threonine phosphorylation site only when the serine and/or threonine is phosphorylated, and that does not substantially bind to the same sequence when the serine and/or threonine is not phosphorylated, thereby prevents downstream signal transduction triggered by a phospho-serine and/or threonine. 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 the same target (e.g., kinases), thereby preventing or reducing the phosphorylation of the endogenous target protein. Alternatively, a peptide comprising a phosphorylated novel serine and/or threonine 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 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 cells. Alternatively, the antibody may directly kill the 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 serine and/or threonine 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, porflmer, 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 serine and/or threonine 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 basophilic Ser/Thr kinase signaling related disease including (among many others) cancer and diabetes, wherein the disease 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 serine and/or threonine 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 a particular basophilic Ser/Thr kinase signaling related disease.


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 serine and/or threonine 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 serine and/or threonine residue is phosphorylated, but does not bind to the same sequence when the serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine 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 diagnostic tests.


The biological sample analyzed may be any sample that is suspected of having abnormal serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine in the presence of the test agent, as compared to a control, indicates that the candidate agent potentially modulates serine and/or threonine 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 serine and/or threonine 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 serine and/or threonine residue is phosphorylated, but does not bind to the same sequence when the serine and/or threonine 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 that 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.


EXAMPLES
Example 1
Isolation of Phospho-serine and Phospho-threonine Containing Peptides Related To Basophilic Ser/Thr Kinase Signaling Pathways and Identification of Novel Phosphorylation Sites

In order to discover novel serine and/or threonine phosphorylation sites in basophilic Ser/Thr kinase signaling pathways, IAP isolation techniques were used to identify phosphoserine and/or threonine-containing peptides in cell extracts from cellular extracts from basophilic Ser/Thr kinase signaling related tissue samples including: A 431; Adult mouse brain; Embryo mouse brain; H1373; H1703; H3255; H441; HCC1937; HCT 116; HeLa; Jurkat; K562; MKN-45; N06cs95; TH-HY2; XY3-130T; XY3-52-T; XY3-68-T; XY3-95N; mouse brain; mouse liver; xy380T. Tryptic phosphoserine and/or threonine-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 gyro-phosphate, 1 mM β-glycerol-phosphate) and sonicated.


Adherent cells at about 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 mN sodium vanadate, supplemented with 2.5 mM sodium pyrophosphate, 1 mM b-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 1-2 days at room temperature.


Trifluoroacetic acid (TFA) was added to protein digests to a final concentration of 1%, precipitate was removed by centrifugation, and digests were loaded onto Sep-Pak C18 columns (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 was obtained by eluting columns with 2 volumes each of 8, 12, and 15% MeCN in 0.1% TFA and combining the eluates with eluates obtained 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 was removed by centrifugation. IAP was performed on each peptide fraction separately. The phosphoserine and/or threonine monoclonal antibody phospho-Akt substrate motif antibody (Cell Signaling Technology, Inc., catalog number 9614) 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% acetonitirile in 0.1% TFA and combination of all eluates. IAP on this peptide fraction was performed as follows: After lyophilization, peptide was dissolved in 1.4 ml IAP buffer (MOPS pH 7.2, 10 mM sodium phosphate, 50 mM NaCl) and insoluble 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 μA of 0.15% TFA. Both eluates were combined.


Analysis by LC-MS/MS Mass Spectrometry.

40 μl or more of IAP eluate were purified by 0.2 μl StageTips 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 Miss./MS spectra were extracted from the raw data file using the Sequest Browser program CreateDta, with the following settings: bottom MW, 700; top MW, 4,500; minimum number of ions, 20 (40 for LTQ); minimum TIC, 4×105(2×103 for LTQ); and precursor charge state, unspecified. Spectra were extracted from the beginning of the raw data file before sample injection to the end of the eluting gradient. The IonQuest and VuDta programs were not used to further select MS/MS spectra for Sequest analysis. MS/MS spectra were evaluated with the following TurboSequest parameters: peptide mass tolerance, 2.5; fragment ion tolerance, 0.0 (1.0 for LTQ); 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 and/or threonine residues. It was determined that restricting phosphorylation to serine and/or threonine 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 serine and/or threonine-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 serine and/or threonine residue is phosphorylated (and does not bind to the same sequence when the serine and/or threonine 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. MAST3 (serine 157).


A 15 amino acid phospho-peptide antigen, RPRSRSLs*PGRATGT (SEQ NO: 266; s*=phosphoserine), which comprises the phosphorylation site derived from afadin iso2 (a protein kinase, ser 157 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. LSM1 (threonine 129).


A 15 amino acid phospho-peptide antigen, LSIPRADt*LDEY (SEQ NO:252; t*=phosphothreonine), which comprises the phosphorylation site derived from LSM1 (an RNA processing protein, Thr 129 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. KIAA0284 (serine 1220).


A 15 amino acid phospho-peptide antigen, TQTPRAGs*SSRARSR (SEQ NO 203; s*=phosphoserine), which comprises the phosphorylation site derived from KIAA0284 (a cytoskeletal protein, Ser 1220 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 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 KIAA0284), found in, for example, 3T3-L1 or mouse liver cells. 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 serine and/or threonine position (e.g., the antibody does not bind to afadin iso3 in the non-stimulated cells, when threonine 1825 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 serine and/or threonine residue is phosphorylated (and does not bind to the same sequence when the serine and/or threonine 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. ARHGEF2 (serine 159).


A 15 amino acid phospho-peptide antigen, NMRNRTLs*VESLIDE (SEQ ID NO: 27; s*=phosphoserine), which comprises the phosphorylation site derived from ARHGEF2 (a G protein or regulator protein, Ser 159 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. BRD1 (serine 415).


A 15 amino acid phospho-peptide antigen, NGVCRKEs*SVKTVRS (SEQ ID NO: 50; s*=phosphoserine), which comprises the phosphorylation site derived from BRD1 (Ser 415 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. ATAD3A (serine 337).


A 15 amino acid phospho-peptide antigen, PSLVRETs*RITVLEA (SEQ ID NO: 38; s*=phosphoserine), which comprises the phosphorylation site derived from ATAD3A (a mitochondrial protein, Ser 337 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 Tks5 phospho-peptide antigenon 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 serine and/or threonine 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. afadin iso 3 (threonine 1825)


An AQUA peptide comprising the sequence, VKASRKLt*ELENELN (SEQ ID NO: 10; t*=phosphothreonine; Leucine being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from afadin iso 3 (Thr 1825 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 afadin iso 3 (Thr 1825) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated afadin iso 3 (Thr 1825) in the sample, as further described below in Analysis & Quantification.


B. ADD1 (threonine 724)


An AQUA peptide comprising the sequence, Rt*PSFLKKS (SEQ ID NO: 8; t*=phosphothreonine; Proline being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from ADD1 (Thr 724 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 ADD1 (Thr 724) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated ADD1 (Thr 724) in the sample, as further described below in Analysis & Quantification.


C. APBA1 (serine 80)


An AQUA peptide comprising the sequence, ECLARSAs*TESGFHN (SEQ ID NO: 19; s*=phosphoserine; Leucine being 14C/15N-labeled, as indicated in bold), which comprises the phosphorylation site derived from APBA1 (ser 80 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 APBA1 (ser 80) AQUA peptide is then spiked into a biological sample to quantify the amount of phosphorylated APBA1 (ser 80) 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).


Example 5
Development of the Phospho-SATB1 (Ser 47) Polyclonal Antibody

A 15 amino acid phospho-peptide antigen, LGRGRLGs*TGAKMQG (where s=phosphoserine) (SEQ ID NO: 353), corresponding to residues 40-54 of human SATB1 encompassing the serine 47 plus cysteine on the N-terminus for coupling, was constructed according to standard synthesis techniques using a Rainin/Protein Technologies, Inc., Symphony peptide synthesizer. See ANTIBODIES: A LABORATORY MANUAL, supra.; Merrifield, supra.


These peptides were coupled to KLH, and rabbits are then injected intradermally (ID) on the back with antigen in complete Freunds adjuvant (500 μg antigen per rabbit). The rabbits were boosted with the same antigen in incomplete Freund adjuvant (250 μg antigen per rabbit) every three weeks. After the fifth boost, the bleeds were collected. The sera were purified by Protein A-affinity chromatography as previously described (see ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor, supra.). The eluted immunoglobulins are then loaded onto a resin-LGRGRLGs*TGAKMQG Knotes column. After washing the column extensively, the phospho-SATB1 (Ser 47) antibodies were eluted and kept in antibody storage buffer.


The antibody (commercially available from Cell Signaling Technology, Inc. in Beverly, Mass.) was further tested for phospho-specificity by Western blot analysis as show in FIG. 3 of the Drawings (for further details see product #4028 in the 2009-2010 Cell Signaling Technology, Inc. catalogue or on the Cell Signaling Technology, Inc. website).

Claims
  • 1. An isolated phosphorylation site-specific antibody or antigen-binding fragment thereof that specifically binds a human signaling protein selected from Column A of Table 1, Rows 4, 15, 58, 61, 63, 72, 75, 102, 138, 145, 161, 168, 172, 177, 190, 203, 228, 253, 282, 283, and 354 only when phosphorylated at the serine or threonine listed in corresponding Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 3, 14, 57, 60, 62, 71, 74, 101, 137, 144, 160, 167, 171, 176, 189, 202, 227, 252, 281, 282, and 353), wherein said antibody does not bind said signaling protein when not phosphorylated at said serine or threonine.
  • 2. An isolated phosphorylation site-specific antibody or antigen-binding fragment thereof that specifically binds a human signaling protein selected from Column A of Table 1, Rows 4, 15, 58, 61, 63, 72, 75, 102, 138, 145, 161, 168, 172, 177, 190, 203, 228, 253, 282, 283, and 354 only when not phosphorylated at the serine or threonine listed in corresponding Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 3, 14, 57, 60, 62, 71, 74, 101, 137, 144, 160, 167, 171, 176, 189, 202, 227, 252, 281, 282, and 353), wherein said antibody does not bind said signaling protein when phosphorylated at said serine or threonine.
  • 3. A method selected from the group consisting of: (a) a method for detecting a human signaling protein selected from Column A of Table 1, Rows 4, 15, 58, 61, 63, 72, 75, 102, 138, 145, 161, 168, 172, 177, 190, 203, 228, 253, 282, 283, and 354 wherein said human signaling protein is phosphorylated at the serine or threonine listed in corresponding Column D of Table 1, comprised within the corresponding phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 3, 14, 57, 60, 62, 71, 74, 101, 137, 144, 160, 167, 171, 176, 189, 202, 227, 252, 281, 282, and 353), comprising the step of adding an isolated phosphorylation-specific antibody or antigen-binding fragment thereof according to claim 1, to a sample comprising said human signaling protein under conditions that permit the binding of said antibody or antigen binding fragment thereof to said human signaling protein, and detecting bound antibody or antigen binding fragment thereof;(b) a method for quantifying the amount of a human signaling protein listed in Column A of Table 1, Rows 4, 15, 58, 61, 63, 72, 75, 102, 138, 145, 161, 168, 172, 177, 190, 203, 228, 253, 282, 283, and 354 that is phosphorylated at the corresponding serine or thereonine listed in the corresponding Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 (SEQ ID NOs: 3, 14, 57, 60, 62, 71, 74, 101, 137, 144, 160, 167, 171, 176, 189, 202, 227, 252, 281, 282, and 353.), in a sample using a heavy-isotope labeled peptide (AQUA™ peptide), said labeled peptide comprising the phosphorylated serine or threonine listed in corresponding Column D of Table 1, comprised within the phosphorylatable peptide sequence listed in corresponding Column E of Table 1 as an internal standard; and(c) a method comprising step (a) followed by step (b)
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. 61/204,617, filed Jan. 7, 2009, the contents of which is incorporated herein, in its entirety, by reference.

Provisional Applications (1)
Number Date Country
61204617 Jan 2009 US