Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators

Abstract
EGFR biomarkers useful in a method for identifying a mammal that will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises (a) exposing the mammal to the EGFR modulator and (b) measuring in the mammal the level of the at least one biomarker, wherein a difference in the level of the at least one biomarker measured in (b) compared to the level of the biomarker in a mammal that has not been exposed to the EGFR modulator indicates that the mammal will respond therapeutically to the method of treating cancer.
Description
FIELD OF THE INVENTION

The present invention relates generally to the field of pharmacogenomics, and more specifically to methods and procedures to determine sensitivity in patients to allow the development of individualized genetic profiles which aid in treating diseases and disorders based on patient response at a molecular level.


BACKGROUND OF THE INVENTION

Cancer is a disease with extensive histoclinical heterogeneity. Although conventional histological and clinical features have been correlated to prognosis, the same apparent prognostic type of tumors varies widely in its responsiveness to therapy and consequent survival of the patient.


New prognostic and predictive markers, which would facilitate an individualization of therapy for each patient, are needed to accurately predict patient response to treatments, such as small molecule or biological molecule drugs, in the clinic. The problem may be solved by the identification of new parameters that could better predict the patient's sensitivity to treatment. The classification of patient samples is a crucial aspect of cancer diagnosis and treatment. The association of a patient's response to a treatment with molecular and genetic markers can open up new opportunities for treatment development in non-responding patients, or distinguish a treatment's indication among other treatment choices because of higher confidence in the efficacy. Further, the pre-selection of patients who are likely to respond well to a medicine, drug, or combination therapy may reduce the number of patients needed in a clinical study or accelerate the time needed to complete a clinical development program (M. Cockett et al., 2000, Current Opinion in Biotechnology, 11:602-609).


The ability to predict drug sensitivity in patients is particularly challenging because drug responses reflect not only properties intrinsic to the target cells, but also a host's metabolic properties. Efforts to use genetic information to predict drug sensitivity have primarily focused on individual genes that have broad effects, such as the multidrug resistance genes, mdr1 and mrp1 (P. Sonneveld, 2000, J. Intern. Med., 247:521-534).


The development of microarray technologies for large scale characterization of gene mRNA expression pattern has made it possible to systematically search for molecular markers and to categorize cancers into distinct subgroups not evident by traditional histopathological methods (J. Khan et al., 1998, Cancer Res., 58:5009-5013; A. A. Alizadeh et al., 2000, Nature, 403:503-511; M. Bittner et al., 2000, Nature, 406:536-540; J. Khan et al., 2001, Nature Medicine, 7(6):673-679; and T. R. Golub et al., 1999, Science, 286:531-537; U. Mon et al., 1999, Proc. Natl. Acad. Sci. USA, 96:6745-6750). Such technologies and molecular tools have made it possible to monitor the expression level of a large number of transcripts within a cell population at any given time (see, e.g., Schena et al., 1995, Science, 270:467-470; Lockhart et al., 1996, Nature Biotechnology, 14:1675-1680; Blanchard et al., 1996, Nature Biotechnology, 14:1649; U.S. Pat. No. 5,569,588 to Ashby et al.).


Recent studies demonstrate that gene expression information generated by microarray analysis of human tumors can predict clinical outcome (L. J. van't Veer et al., 2002, Nature, 415:530-536; M. West et al., 2001, Proc. Natl. Acad. Sci. USA, 98:11462-11467; T. Sorlie et al., 2001, Proc. Natl. Acad. Sci. USA, 98:10869-10874; M. Shipp et al., 2002, Nature Medicine, 8(1):68-74). These findings bring hope that cancer treatment will be vastly improved by better predicting the response of individual tumors to therapy.


Needed are new and alternative methods and procedures to determine drug sensitivity in patients to allow the development of individualized genetic profiles which are necessary to treat diseases and disorders based on patient response at a molecular level.


SUMMARY OF THE INVENTION

The invention provides methods and procedures for determining patient sensitivity to one or more Epidermal Growth Factor Receptor (EGFR) modulators. The invention also provides methods of determining or predicting whether an individual requiring therapy for a disease state such as cancer will or will not respond to treatment, prior to administration of the treatment, wherein the treatment comprises one or more EGFR modulators. The one or more EGFR modulators are compounds that can be selected from, for example, one or more EGFR specific ligands, one or more small molecule EGFR inhibitors, or one or more EGFR binding monoclonal antibodies.


In one aspect, the invention provides a method for identifying a mammal that will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises: (a) measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1; (b) exposing the mammal to the EGFR modulator; (c) following the exposing of step (b), measuring in the mammal the level of the at least one biomarker, wherein a difference in the level of the at least one biomarker measured in step (c) compared to the level of the at least one biomarker measured in step (a) indicates that the mammal will respond therapeutically to said method of treating cancer.


As used herein, respond therapeutically refers to the alleviation or abrogation of the cancer. This means that the life expectancy of an individual affected with the cancer will be increased or that one or more of the symptoms of the cancer will be reduced or ameliorated. The term encompasses a reduction in cancerous cell growth or tumor volume. Whether a mammal responds therapeutically can be measured by many methods well known in the art, such as PET imaging.


The mammal can be, for example, a human, rat, mouse, dog rabbit, pig sheep, cow, horse, cat, primate, or monkey.


The method of the invention can be, for example, an in vitro method and wherein the at least one biomarker is measured in at least one mammalian biological sample from the mammal. The biological sample can comprise, for example, at least one of whole fresh blood, peripheral blood mononuclear cells, frozen whole blood, fresh plasma, frozen plasma, urine, saliva, skin, hair follicle, or tumor tissue.


In another aspect, the invention provides a method for identifying a mammal that will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises: (a) exposing the mammal to the EGFR modulator; (b) following the exposing of step (a), measuring in the mammal the level of the at least one biomarker selected from the biomarkers of Table 1, wherein a difference in the level of the at least one biomarker measured in step (b), compared to the level of the biomarker in a mammal that has not been exposed to said EGFR modulator, indicates that the mammal will respond therapeutically to said method of treating cancer.


In yet another aspect, the invention provides a method for testing or predicting whether a mammal will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises: (a) measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1; (b) exposing the mammal to the EGFR modulator; (c) following the exposing of step (b), measuring in the mammal the level of the at least one biomarker, wherein a difference in the level of the at least one biomarker measured in step (c) compared to the level of the at least one biomarker measured in step (a) indicates that the mammal will respond therapeutically to said method of treating cancer.


In another aspect, the invention provides a method for determining whether a compound inhibits EGFR activity in a mammal, comprising: (a) exposing the mammal to the compound; and (b) following the exposing of step (a), measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1, wherein a difference in the level of said biomarker measured in step (b), compared to the level of the biomarker in a mammal that has not been exposed to said compound, indicates that the compound inhibits EGFR activity in the mammal.


In yet another aspect, the invention provides a method for determining whether a mammal has been exposed to a compound that inhibits EGFR activity, comprising (a) exposing the mammal to the compound; and (b) following the exposing of step (a), measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1, wherein a difference in the level of said biomarker measured in step (b), compared to the level of the biomarker in a mammal that has not been exposed to said compound, indicates that the mammal has been exposed to a compound that inhibits EGFR activity.


In another aspect, the invention provides a method for determining whether a mammal is responding to a compound that inhibits EGFR activity, comprising (a) exposing the mammal to the compound; and (b) following the exposing of step (a), measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1, wherein a difference in the level of said biomarker measured in step (b), compared to the level of the biomarker in a mammal that has not been exposed to said compound, indicates that the mammal is responding to the compound that inhibits EGFR activity.


As used herein, “responding” encompasses responding by way of a biological and cellular response, as well as a clinical response (such as improved symptoms, a therapeutic effect, or an adverse event), in a mammal


The invention also provides an isolated biomarker selected from the biomarkers of Table 1. The biomarkers of the invention comprise sequences selected from the nucleotide and amino acid sequences provided in Table 1 and the Sequence Listing, as well as fragments and variants thereof.


The invention also provides a biomarker set comprising two or more biomarkers selected from the biomarkers of Table 1.


The invention also provides kits for determining or predicting whether a patient would be susceptible or resistant to a treatment that comprises one or more EGFR modulators. The patient may have a cancer or tumor such as, for example, a colon cancer or tumor.


In one aspect, the kit comprises a suitable container that comprises one or more specialized microarrays of the invention, one or more EGFR modulators for use in testing cells from patient tissue specimens or patient samples, and instructions for use. The kit may further comprise reagents or materials for monitoring the expression of a biomarker set at the level of mRNA or protein.


In another aspect, the invention provides a kit comprising two or more biomarkers selected from the biomarkers of Table 1.


In yet another aspect, the invention provides a kit comprising at least one of an antibody and a nucleic acid for detecting the presence of at least one of the biomarkers selected from the biomarkers of Table 1. In one aspect, the kit further comprises instructions for determining whether or not a mammal will respond therapeutically to a method of treating cancer comprising administering a compound that inhibits EGFR activity. In another aspect, the instructions comprise the steps of (a) measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1, (b) exposing the mammal to the compound, (c) following the exposing of step (b), measuring in the mammal the level of the at least one biomarker, wherein a difference in the level of the at least one biomarker measured in step (c) compared to the level of the at least one biomarker measured in step (a) indicates that the mammal will respond therapeutically to said method of treating cancer.


The invention also provides screening assays for determining if a patient will be susceptible or resistant to treatment with one or more EGFR modulators.


The invention also provides a method of monitoring the treatment of a patient having a disease treatable by one or more EGFR modulators.


The invention also provides individualized genetic profiles which are necessary to treat diseases and disorders based on patient response at a molecular level.


The invention also provides specialized microarrays, e.g., oligonucleotide microarrays or cDNA microarrays, comprising one or more biomarkers having expression profiles that correlate with either sensitivity or resistance to one or more EGFR modulators.


The invention also provides antibodies, including polyclonal or monoclonal, directed against one or more biomarkers of the invention.


The invention will be better understood upon a reading of the detailed description of the invention when considered in connection with the accompanying figures.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 illustrates the gene filtering process.



FIG. 2 illustrates the cell line filtering process.



FIG. 3 illustrates the cell line IC50 data.



FIG. 4 illustrates the T-test Results I.



FIG. 5 illustrates the T-test Results II.



FIG. 6 illustrates the T-test Results III.





DETAILED DESCRIPTION OF THE INVENTION

The invention provides biomarkers that respond to the modulation of a specific signal transduction pathway and also correlate with EGFR modulator sensitivity or resistance. These biomarkers can be employed for predicting response to one or more EGFR modulators. In one aspect, the biomarkers of the invention are those provided in Table 1 and the Sequence Listing, including both polynucleotide and polypeptide sequences.









TABLE 1







BIOMARKERS









Unigene title and SEQ

Affymetrix


ID NOS:
Affymetrix Description
Probe Set





Cadherin 17, LI
gb: U07969.1 /DEF = Human intestinal
209847_at


cadherin (liver-intestine)
peptide-associated transporter HPT-1


SEQ ID NOS: 1
mRNA, complete cds. /FEA = mRNA


(nucleotide) and 67
/PROD = intestinal peptide-associated


(amino acid)
transporter HPT-1 /DB_XREF = gi: 483391



/UG = Hs.89436 cadherin 17, LI cadherin



(liver-intestine) /FL = gb: NM_004063.1



gb: U07969.1


Carcinoembryonic
gb: BC005008.1 /DEF = Homo sapiens,
203757_s_at


antigen-related cell
carcinoembryonic antigen-related cell


adhesion molecule 6
adhesion molecule 6 (non-specific cross


(non-specific cross
reacting antigen), clone MGC: 10467,


reacting antigen)
mRNA, complete cds. /FEA = mRNA


SEQ ID NOS: 2
/PROD = carcinoembryonic antigen-related


(nucleotide) and 68
cell adhesionmolecule 6 (non-specific


(amino acid)
cross reacting antigen)



/DB_XREF = gi: 13477106 /UG = Hs.73848



carcinoembryonic antigen-related cell



adhesion molecule 6 (non-specific cross



reacting antigen) /FL = gb: BC005008.1



gb: M18216.1 gb: M29541.1



gb: NM_002483.1


Carcinoembryonic
gb: M18728.1 /DEF = Human nonspecific
211657_at


antigen-related cell
crossreacting antigen mRNA, complete


adhesion molecule 6
cds. /FEA = mRNA /GEN = NCA; NCA;


(non-specific cross
NCA /PROD = non-specific cross reacting


reacting antigen)
antigen /DB_XREF = gi: 189084


SEQ ID NOS: 3
/FL = gb: M18728.1


(nucleotide) and 69


(amino acid)


Lectin, galactoside-
gb: NM_002305.2 /DEF = Homo sapiens
201105_at


binding, soluble, 1
lectin, galactoside-binding, soluble, 1


(galectin 1)
(galectin 1) (LGALS1), mRNA.


SEQ ID NOS: 4
/FEA = mRNA /GEN = LGALS1


(nucleotide) and 70
/PROD = beta-galactosidase binding lectin


(amino acid)
precursor /DB_XREF = gi: 6006015



/UG = Hs.227751 lectin, galactoside-



binding, soluble, 1 (galectin 1)



/FL = gb: BC001693.1 gb: J04456.1



gb: NM_002305.2


Transmembrane
gb: AF270487.1 /DEF = Homo sapiens
211689_s_at


protease, serine 2
androgen-regulated serine protease


SEQ ID NOS: 5
TMPRSS2 precursor (TMPRSS2) mRNA,


(nucleotide) and 71
complete cds. /FEA = mRNA


(amino acid)
/GEN = TMPRSS2 /PROD = androgen-



regulated serine protease



TMPRSS2precursor



/DB_XREF = gi: 13540003



/FL = gb: AF270487.1


Mucin 5, subtypes A and
Consensus includes gb: AW192795
214303_x_at


C,
/FEA = EST /DB_XREF = gi: 6471494


tracheobronchial/gastric
/DB_XREF = est: x151d08.x1


SEQ ID NOS: 6
/CLONE = IMAGE: 2678223


(nucleotide), 7
/UG = Hs.103707 apomucin


(nucleotide) and 72


(amino acid)


3-hydroxy-3-
gb: NM_005518.1 /DEF = Homo sapiens 3-
204607_at


methylglutaryl-
hydroxy-3-methylglutaryl-Coenzyme A


Coenzyme A synthase 2
synthase 2 (mitochondrial) (HMGCS2),


(mitochondrial)
mRNA. /FEA = mRNA /GEN = HMGCS2


SEQ ID NOS: 8
/PROD = 3-hydroxy-3-methylglutaryl-


(nucleotide) and 73
Coenzyme A synthase 2(mitochondrial)


(amino acid)
/DB_XREF = gi: 5031750 /UG = Hs.59889 3-



hydroxy-3-methylglutaryl-Coenzyme A



synthase 2 (mitochondrial)



/FL = gb: NM_005518.1


Interferon-stimulated
gb: NM_005101.1 /DEF = Homo sapiens
205483_s_at


protein, 15 kDa
interferon-stimulated protein, 15 kDa


SEQ ID NOS: 9
(ISG15), mRNA. /FEA = mRNA


(nucleotide) and 74
/GEN = ISG15 /PROD = interferon-


(amino acid)
stimulated protein, 15 kDa



/DB_XREF = gi: 4826773 /UG = Hs.833



interferon-stimulated protein, 15 kDa



/FL = gb: M13755.1 gb: NM_005101.1


Dopa decarboxylase
gb: NM_000790.1 /DEF = Homo sapiens
205311_at


(aromatic L-amino acid
dopa decarboxylase (aromatic L-amino


decarboxylase)
acid decarboxylase) (DDC), mRNA.


SEQ ID NOS: 10
/FEA = mRNA /GEN = DDC /PROD = dopa


(nucleotide) and 75
decarboxylase (aromatic L-amino


(amino acid)
aciddecarboxylase)



/DB_XREF = gi: 4503280 /UG = Hs.150403



dopa decarboxylase (aromatic L-amino



acid decarboxylase) /FL = gb: BC000485.1



gb: M76180.1 gb: M88700.1



gb: NM_000790.1


Serine (or cysteine)
gb: NM_000602.1 /DEF = Homo sapiens
202628_s_at


proteinase inhibitor,
serine (or cysteine) proteinase inhibitor,


clade E (nexin,
clade E (nexin, plasminogen activator


plasminogen activator
inhibitor type 1), member 1 (SERPINE1),


inhibitor type 1),
mRNA. /FEA = mRNA /GEN = SERPINE1


member 1
/PROD = serine (or cysteine) proteinase


SEQ ID NOS: 11
inhibitor, cladeE (nexin, plasminogen


(nucleotide) and 76
activator inhibitor type 1), member1


(amino acid)
/DB_XREF = gi: 10835158 /UG = Hs.82085



serine (or cysteine) proteinase inhibitor,



clade E (nexin, plasminogen activator



inhibitor type 1), member 1



/FL = gb: NM_000602.1 gb: M16006.1


FXYD domain-
gb: BC005238.1 /DEF = Homo sapiens,
202489_s_at


containing ion transport
FXYD domain-containing ion transport


regulator 3
regulator 3, clone MGC: 12265, mRNA,


SEQ ID NOS: 12
complete cds. /FEA = mRNA


(nucleotide) and 77
/PROD = FXYD domain-containing ion


(amino acid)
transport regulator3



/DB_XREF = gi: 13528881 /UG = Hs.301350



FXYD domain-containing ion transport



regulator 3 /FL = gb: NM_005971.2



gb: BC005238.1


Putative integral
gb: NM_018407.1 /DEF = Homo sapiens
208029_s_at


membrane transporter
putative integral membrane transporter


SEQ ID NOS: 13
(LC27), mRNA. /FEA = mRNA


(nucleotide) and 78
/GEN = LC27 /PROD = putative integral


(amino acid)
membrane transporter



/DB_XREF = gi: 8923827



/FL = gb: NM_018407.1


Protease inhibitor 3,
gb: NM_002638.1 /DEF = Homo sapiens
203691_at


skin-derived (SKALP)
protease inhibitor 3, skin-derived (SKALP)


SEQ ID NOS: 14
(PI3), mRNA. /FEA = mRNA /GEN = PI3


(nucleotide) and 79
/PROD = protease inhibitor 3, skin-derived


(amino acid)
(SKALP) /DB_XREF = gi: 4505786



/UG = Hs.112341 protease inhibitor 3, skin-



derived (SKALP) /FL = gb: NM_002638.1


Caudal type homeo box
gb: U51096.1 /DEF = Human homeobox
206387_at


transcription factor 2
protein Cdx2 mRNA, complete cds.


SEQ ID NOS: 15
/FEA = mRNA /PROD = homeobox protein


(nucleotide) and 80
Cdx2 /DB_XREF = gi: 1777773


(amino acid)
/UG = Hs.77399 caudal type homeo box



transcription factor 2 /FL = gb: U51096.1



gb: NM_001265.1


Fibroblast growth factor
gb: NM_000142.2 /DEF = Homo sapiens
204379_s_at


receptor 3
fibroblast growth factor receptor 3


(achondroplasia,
(achondroplasia, thanatophoric dwarfism)


thanatophoric dwarfism)
(FGFR3), transcript variant 1, mRNA.


SEQ ID NOS: 16
/FEA = mRNA /GEN = FGFR3


(nucleotide) and 81
/PROD = fibroblast growth factor receptor


(amino acid)
3, isoform 1precursor



/DB_XREF = gi: 13112046 /UG = Hs.1420



fibroblast growth factor receptor 3



(achondroplasia, thanatophoric dwarfism)



/FL = gb: NM_000142.2 gb: M58051.1


Hypothetical protein
Consensus includes gb: AL041124
213343_s_at


PP1665
/FEA = EST /DB_XREF = gi: 5410060


SEQ ID NOS: 17
/DB_XREF = est: DKFZp434D0316_s1


(nucleotide), 18
/CLONE = DKFZp434D0316 /UG = Hs.6748


(nucleotide) and 82
hypothetical protein PP1665


(amino acid)


Protease inhibitor 3,
Cluster Incl. L10343: Huma elafin gene,
41469_at


skin-derived (SKALP)
complete cds /cds = (516,869) /gb = L10343


SEQ ID NOS:19
/gi = 190337 /ug = Hs.112341 /len = 871


(nucleotide) and 83


(amino acid)


A kinase (PRKA)
gb: AB003476.1 /DEF = Homo sapiens
210517_s_at


anchor protein (gravin)
mRNA for gravin, complete cds.


12
/FEA = mRNA /PROD = gravin


SEQ ID NOS: 20
/DB_XREF = gi: 2081606 /UG = Hs.788 A


(nucleotide) and 84
kinase (PRKA) anchor protein (gravin) 12


(amino acid)
/FL = gb: AB003476.1


Lymphocyte antigen 75
gb: NM_002349.1 /DEF = Homo sapiens
205668_at


SEQ ID NOS: 21
lymphocyte antigen 75 (LY75), mRNA.


(nucleotide) and 85
/FEA = mRNA /GEN = LY75


(amino acid)
/PROD = lymphocyte antigen 75



/DB_XREF = gi: 4505052 /UG = Hs.153563



lymphocyte antigen 75



/FL = gb: AF011333.1 gb: AF064827.1



gb: NM_002349.1


Mucin 5, subtypes A and
Consensus includes gb: AI521646
214385_s_at


C,
/FEA = EST /DB_XREF = gi: 4435781


tracheobronchial/gastric
/DB_XREF = est: to66a06.x1


SEQ ID NOS: 22
/CLONE = IMAGE: 2183218


(nucleotide)
/UG = Hs.102482 mucin 5, subtype B,



tracheobronchial


Metallothionein 1G
gb: NM_005950.1 /DEF = Homo sapiens
204745_x_at


SEQ ID NOS: 23
metallothionein 1G (MT1G), mRNA.


(nucleotide) and 86
/FEA = mRNA /GEN = MT1G


(amino acid)
/PROD = metallothionein 1G



/DB_XREF = gi: 10835229 /UG = Hs.173451



metallothionein 1G /FL = gb: NM_005950.1


Tumor necrosis factor
gb: NM_003823.1 /DEF = Homo sapiens
206467_x_at


receptor superfamily,
tumor necrosis factor receptor superfamily,


member 6b, decoy
member 6b, decoy (TNFRSF6B), mRNA.


SEQ ID NOS: 24
/FEA = mRNA /GEN = TNFRSF6B


(nucleotide) and 87
/PROD = decoy receptor 3


(amino acid)
/DB_XREF = gi: 4507584 /UG = Hs.278556



tumor necrosis factor receptor superfamily,



member 6b, decoy /FL = gb: AF104419.1



gb: NM_003823.1 gb: AF134240.1



gb: AF217794.1


Mucin 3B
Consensus includes gb: AB038783.1
214898_x_at


SEQ ID NOS: 25
/DEF = Homo sapiens MUC3B mRNA for


(nucleotide) and 88
intestinal mucin, partial cds. /FEA = mRNA


(amino acid)
/GEN = MUC3B /PROD = intestinal mucin



/DB_XREF = gi: 9929917 /UG = Hs.129782



mucin 3A, intestinal


Metallothionein 1x
gb: NM_005952.1 /DEF = Homo sapiens
208581_x_at


SEQ ID NOS: 26
metallothionein 1X (MT1X), mRNA.


(nucleotide) and 89
/FEA = CDS /GEN = MT1X


(amino acid)
/PROD = metallothionein 1X



/DB_XREF = gi: 10835231 /UG = Hs.278462



metallothionein 1X /FL = gb: NM_005952.1


GRO3 oncogene
gb: NM_002090.1 /DEF = Homo sapiens
207850_at


SEQ ID NOS: 27
GRO3 oncogene (GRO3), mRNA.


(nucleotide) and 90
/FEA = mRNA /GEN = GRO3


(amino acid)
/PROD = GRO3 oncogene



/DB_XREF = gi: 4504156 /UG = Hs.89690



GRO3 oncogene /FL = gb: M36821.1



gb: NM_002090.1


Transforming growth
gb: NM_000358.1 /DEF = Homo sapiens
201506_at


factor, beta-induced,
transforming growth factor, beta-induced,


68 kD
68 kD (TGFBI), mRNA. /FEA = mRNA


SEQ ID NOS: 28
/GEN = TGFBI /PROD = transforming


(nucleotide) and 91
growth factor, beta-induced, 68 kD


(amino acid)
/DB_XREF = gi: 4507466 /UG = Hs.118787



transforming growth factor, beta-induced,



68 kD /FL = gb: BC000097.1 gb: BC004972.1



gb: M77349.1 gb: NM_000358.1


Bone morphogenetic
gb: M60316.1 /DEF = Human transforming
209591_s_at


protein 7 (osteogenic
growth factor-beta (tgf-beta) mRNA,


protein 1)
complete cds. /FEA = mRNA /GEN = tgf-


SEQ ID NOS: 29
beta /PROD = transforming growth factor-


(nucleotide) and 92
beta /DB_XREF = gi: 339563


(amino acid)
/UG = Hs.170195 bone morphogenetic



protein 7 (osteogenic protein 1)



/FL = gb: M60316.1 gb: NM_001719.1


Annexin A10
gb: AF196478.1 /DEF = Homo sapiens
210143_at


SEQ ID NOS: 30
annexin 14 (ANX14) mRNA, complete


(nucleotide) and 93
cds. /FEA = mRNA /GEN = ANX14


(amino acid)
/PROD = annexin 14



/DB_XREF = gi: 6274496 /UG = Hs.188401



annexin A10 /FL = gb: AF196478.1



gb: NM_007193.2


Metallothionein 1F
Consensus includes gb: M10943
217165_x_at


(functional)
/DEF = Human metallothionein-If gene


SEQ ID NOS: 31
(hMT-If) /FEA = CDS


(nucleotide) and 94
/DB_XREF = gi: 187540 /UG = Hs.203936


(amino acid)
metallothionein 1F (functional)


Annexin A1
gb: NM_000700.1 /DEF = Homo sapiens
201012_at


SEQ ID NOS: 32
annexin A1 (ANXA1), mRNA.


(nucleotide) and 95
/FEA = mRNA /GEN = ANXA1


(amino acid)
/PROD = annexin I /DB_XREF = gi: 4502100



/UG = Hs.78225 annexin A1



/FL = gb: BC001275.1 gb: NM_000700.1


Secretory leukocyte
gb: NM_003064.1 /DEF = Homo sapiens
203021_at


protease inhibitor
secretory leukocyte protease inhibitor


(antileukoproteinase)
(antileukoproteinase) (SLPI), mRNA.


SEQ ID NOS: 33
/FEA = mRNA /GEN = SLPI


(nucleotide) and 96
/PROD = secretory leukocyte protease


(amino acid)
inhibitor(antileukoproteinase)



/DB_XREF = gi: 4507064 /UG = Hs.251754



secretory leukocyte protease inhibitor



(antileukoproteinase)



/FL = gb: NM_003066.1 gb: AF114471.1



gb: NM_003064.1


Polymeric
gb: NM_002644.1 /DEF = Homo sapiens
204213_at


immunoglobulin
polymeric immunoglobulin receptor


receptor
(PIGR), mRNA. /FEA = mRNA


SEQ ID NOS: 34
/GEN = PIGR /PROD = polymeric


(nucleotide) and 97
immunoglobulin receptor


(amino acid)
/DB_XREF = gi: 11342673 /UG = Hs.288579



polymeric immunoglobulin receptor



/FL = gb: NM_002644.1


Carcinoembryonic
gb: NM_004363.1 /DEF = Homo sapiens
201884_at


antigen-related cell
carcinoembryonic antigen-related cell


adhesion molecule 5
adhesion molecule 5 (CEACAM5),


SEQ ID NOS: 35
mRNA. /FEA = mRNA /GEN = CEACAM5


(nucleotide) and 98
/PROD = carcinoembryonic antigen-related


(amino acid)
cell adhesionmolecule 5



/DB_XREF = gi: 11386170 /UG = Hs.220529



carcinoembryonic antigen-related cell



adhesion molecule 5



/FL = gb: NM_004363.1 gb: M29540.1


Protein tyrosine
gb: NM_002847.1 /DEF = Homo sapiens
203029_s_at


phosphatase, receptor
protein tyrosine phosphatase, receptor type,


type, N polypeptide 2
N polypeptide 2 (PTPRN2), mRNA.


SEQ ID NOS: 36
/FEA = mRNA /GEN = PTPRN2


(nucleotide) and 99
/PROD = protein tyrosine phosphatase,


(amino acid)
receptor type, Npolypeptide 2



/DB_XREF = gi: 11386148 /UG = Hs.74624



protein tyrosine phosphatase, receptor type,



N polypeptide 2 /FL = gb: NM_002847.1



gb: U66702.1 gb: AF007555.1


Cystic fibrosis
gb: NM_000492.2 /DEF = Homo sapiens
205043_at


transmembrane
cystic fibrosis transmembrane conductance


conductance regulator,
regulator, ATP-binding cassette (sub-


ATP-binding cassette
family C, member 7) (CFTR), mRNA.


(sub-family C, member
/FEA = mRNA /GEN = CFTR /PROD = cystic


7)
fibrosis transmembrane


SEQ ID NOS: 37
conductanceregulator, ATP-binding


(nucleotide) and 100
cassette (sub-family C, member 7)


(amino acid)
/DB_XREF = gi: 6995995 /UG = Hs.663



cystic fibrosis transmembrane conductance



regulator, ATP-binding cassette (sub-



family C, member 7)



/FL = gb: NM_000492.2


DVS27-related protein
gb: AB024518.1 /DEF = Homo sapiens
209821_at


SEQ ID NOS: 38
mRNA for DVS27-related protein,


(nucleotide) and 101
complete cds. /FEA = mRNA


(amino acid)
/GEN = DVS27 /PROD = DVS27-related



protein /DB_XREF = gi: 4520327



/UG = Hs.58589 glycogenin 2



/FL = gb: AB024518.1


Insulin-like growth
gb: NM_000597.1 /DEF = Homo sapiens
202718_at


factor binding protein 2
insulin-like growth factor binding protein 2


(36 kD)
(36 kD) (IGFBP2), mRNA. /FEA = mRNA


SEQ ID NOS: 39
/GEN = IGFBP2 /PROD = insulin-like


(nucleotide) and 102
growth factor binding protein 2(36 kD)


(amino acid)
/DB_XREF = gi: 10835156 /UG = Hs.162



insulin-like growth factor binding protein 2



(36 kD) /FL = gb: NM_000597.1



gb: BC004312.1 gb: M35410.1


Inhibitor of DNA
gb: NM_002167.1 /DEF = Homo sapiens
207826_s_at


binding 3, dominant
inhibitor of DNA binding 3, dominant


negative helix-loop-
negative helix-loop-helix protein (ID3),


helix protein
mRNA. /FEA = mRNA /GEN = ID3


SEQ ID NOS: 40
/PROD = inhibitor of DNA binding 3,


(nucleotide) and 103
dominant negativehelix-loop-helix protein


(amino acid)
/DB_XREF = gi: 10835060 /UG = Hs.76884



inhibitor of DNA binding 3, dominant



negative helix-loop-helix protein



/FL = gb: NM_002167.1


Phospholipase A2,
Consensus includes gb: X00452.1
203649_s_at


group IIA (platelets,
/DEF = Human mRNA for DC classII


synovial fluid)
histocompatibility antigen alpha-chain.


SEQ ID NOS: 41
/FEA = mRNA /PROD = DC classII


(nucleotide) and 104
histocompatibility antigenalpha-chain


(amino acid)
/DB_XREF = gi: 32265 /UG = Hs.198253



major histocompatibility complex, class II,



DQ alpha 1


Purkinje cell protein 4
gb: NM_006198.1 /DEF = Homo sapiens
205549_at


SEQ ID NOS: 42
Purkinje cell protein 4 (PCP4), mRNA.


(nucleotide) and 105
/FEA = mRNA /GEN = PCP4


(amino acid)
/PROD = Purkinje cell protein 4



/DB_XREF = gi: 5453857 /UG = Hs.80296



Purkinje cell protein 4 /FL = gb: U52969.1



gb: NM_006198.1


G protein-coupled
Consensus includes gb: AL524520
213880_at


receptor 49
/FEA = EST /DB_XREF = gi: 12788013


SEQ ID NOS: 43
/DB_XREF = est: AL524520


(nucleotide), 44
/CLONE = CS0DC007YG21 (3 prime)


(nucleotide) and 106
/UG = Hs.285529 G protein-coupled


(amino acid)
receptor 49


Fucosyltransferase 3
Consensus includes gb: AW080549
214088_s_at


(galactoside 3(4)-L-
/FEA = EST /DB_XREF = gi: 6035701


fucosyltransferase,
/DB_XREF = est: xc33a08.x1


Lewis blood group
/CLONE = IMAGE: 2586038


included)
/UG = Hs.169238 fucosyltransferase 3


SEQ ID NOS: 45
(galactoside 3(4)-L-fucosyltransferase,


(nucleotide), 46
Lewis blood group included)


(nucleotide) and 107


(amino acid)


Interferon, alpha-
gb: NM_005532.1 /DEF = Homo sapiens
202411_at


inducible protein 27
interferon, alpha-inducible protein 27


SEQ ID NOS: 47
(IFI27), mRNA. /FEA = mRNA


(nucleotide) and 108
/GEN = IFI27 /PROD = interferon, alpha-


(amino acid)
inducible protein 27



/DB_XREF = gi: 5031780 /UG = Hs.278613



interferon, alpha-inducible protein 27



/FL = gb: NM_005532.1


Serine (or cysteine)
gb: NM_002639.1 /DEF = Homo sapiens
204855_at


proteinase inhibitor,
serine (or cysteine) proteinase inhibitor,


clade B (ovalbumin),
clade B (ovalbumin), member 5


member 5
(SERPINB5), mRNA. /FEA = mRNA


SEQ ID NOS: 48
/GEN = SERPINB5 /PROD = serine (or


(nucleotide) and 109
cysteine) proteinase inhibitor, cladeB


(amino acid)
(ovalbumin), member 5



/DB_XREF = gi: 4505788 /UG = Hs.55279



serine (or cysteine) proteinase inhibitor,



clade B (ovalbumin), member 5



/FL = gb: NM_002639.1 gb: U04313.1



Homo sapiens CD44

gb: AF098641.1 /DEF = Homo sapiens
210916_s_at


isoform RC (CD44)
CD44 isoform RC (CD44) mRNA,


mRNA, complete cds
complete cds. /FEA = mRNA /GEN = CD44


SEQ ID NOS: 49
/PROD = CD44 isoform RC


(nucleotide) and 110
/DB_XREF = gi: 3832517 /UG = Hs.306278


(amino acid)

Homo sapiens CD44 isoform RC (CD44)




mRNA, complete cds /FL = gb: AF098641.1


Solute carrier family 7
gb: NM_012244.1 /DEF = Homo sapiens
202752_x_at


(cationic amino acid
solute carrier family 7 (cationic amino acid


transporter, y+ system),
transporter, y+ system), member 8


member 8
(SLC7A8), mRNA. /FEA = mRNA


SEQ ID NOS: 50
/GEN = SLC7A8 /PROD = solute carrier


(nucleotide) and 111
family 7 (cationic amino acidtransporter,


(amino acid)
y+ system), member 8



/DB_XREF = gi: 6912669 /UG = Hs.22891



solute carrier family 7 (cationic amino acid



transporter, y+ system), member 8



/FL = gb: AB037669.1 gb: AF171669.1



gb: NM_012244.1


Membrane protein,
gb: NM_002436.2 /DEF = Homo sapiens
202974_at


palmitoylated 1 (55 kD)
membrane protein, palmitoylated 1 (55 kD)


SEQ ID NOS: 51
(MPP1), mRNA. /FEA = mRNA


(nucleotide) and 112
/GEN = MPP1 /PROD = palmitoylated


(amino acid)
membrane protein 1



/DB_XREF = gi: 6006024 /UG = Hs.1861



membrane protein, palmitoylated 1 (55 kD)



/FL = gb: BC002392.1 gb: M64925.1



gb: NM_002436.2


Tumor protein p53 (Li-
gb: K03199.1 /DEF = Human p53 cellular
211300_s_at


Fraumeni syndrome)
tumor antigen mRNA, complete cds.


SEQ ID NOS: 52
/FEA = mRNA /GEN = TP53


(nucleotide) and 113
/DB_XREF = gi: 189478 /UG = Hs.1846


(amino acid)
tumor protein p53 (Li-Fraumeni syndrome)



/FL = gb: K03199.1


S100 calcium-binding
gb: NM_005980.1 /DEF = Homo sapiens
204351_at


protein P
S100 calcium-binding protein P (S100P),


SEQ ID NOS: 53
mRNA. /FEA = mRNA /GEN = S100P


(nucleotide) and 114
/PROD = S100 calcium-binding protein P


(amino acid)
/DB_XREF = gi: 5174662 /UG = Hs.2962



S100 calcium-binding protein P



/FL = gb: NM_005980.1


Serine (or cysteine)
gb: AF119873.1 /DEF = Homo sapiens
211429_s_at


proteinase inhibitor,
PRO2275 mRNA, complete cds.


clade A (alpha-1
/FEA = mRNA /PROD = PRO2275


antiproteinase,
/DB_XREF = gi: 7770182 /UG = Hs.297681


antitrypsin), member 1
serine (or cysteine) proteinase inhibitor,


SEQ ID NOS: 54
clade A (alpha-1 antiproteinase,


(nucleotide) and 115
antitrypsin), member 1


(amino acid)
/FL = gb: AF119873.1


Eukaryotic translation
gb: NM_001970.1 /DEF = Homo sapiens
201123_s_at


initiation factor 5A
eukaryotic translation initiation factor 5A


SEQ ID NOS: 55
(EIF5A), mRNA. /FEA = mRNA


(nucleotide) and 116
/GEN = EIF5A /PROD = eukaryotic


(amino acid)
translation initiation factor 5A



/DB_XREF = gi: 4503544 /UG = Hs.119140



eukaryotic translation initiation factor 5A



/FL = gb: BC000751.1 gb: BC001832.1



gb: M23419.1 gb: NM_001970.1


Old astrocyte
Consensus includes gb: AF055009.1
213059_at


specifically induced
/DEF = Homo sapiens clone 24747 mRNA


substance
sequence. /FEA = mRNA


SEQ ID NOS: 56
/DB_XREF = gi: 3005731 /UG = Hs.13456


(nucleotide), 57

Homo sapiens clone 24747 mRNA



(nucleotide) and 117
sequence


(amino acid)


UDP glycosyltransferase
gb: NM_019093.1 /DEF = Homo sapiens
208596_s_at


1 family, polypeptide
UDP glycosyltransferase 1 family,


A3
polypeptide A3 (UGT1A3), mRNA.


SEQ ID NOS: 58
/FEA = CDS /GEN = UGT1A3 /PROD = UDP


(nucleotide) and 118
glycosyltransferase 1 family,


(amino acid)
polypeptideA3 /DB_XREF = gi: 13487899



/UG = Hs.326543 UDP glycosyltransferase



1 family, polypeptide A3



/FL = gb: NM_019093.1


Alpha-2-HS-
gb: AF130057.1 /DEF = Homo sapiens clone
210929_s_at


glycoprotein
FLB5539 PRO1454 mRNA, complete cds.


SEQ ID NOS: 59
/FEA = mRNA /PROD = PRO1454


(nucleotide) and 119
/DB_XREF = gi: 11493420 /UG = Hs.323288


(amino acid)

Homo sapiens clone FLB5539 PRO1454




mRNA, complete cds /FL = gb: AF130057.1


ESTs, Highly similar to
Consensus includes gb: AV691323
215125_s_at


A39092
/FEA = EST /DB_XREF = gi: 10293186


glucuronosyltransferase
/DB_XREF = est: AV691323


[H. sapiens]
/CLONE = GKCEWFL11 /UG = Hs.2056


SEQ ID NOS: 60
UDP glycosyltransferase 1 family,


(nucleotide), 61
polypeptide A9


(nucleotide) and 120


(amino acid)


UDP glycosyltransferase
gb: NM_000463.1 /DEF = Homo sapiens
207126_x_at


1 family, polypeptide
UDP glycosyltransferase 1 family,


A1
polypeptide A1 (UGT1A1), mRNA.


SEQ ID NOS: 62
/FEA = mRNA /GEN = UGT1A1


(nucleotide) and 121
/PROD = UDP glycosyltransferase 1 family,


(amino acid)
polypeptideA1 /DB_XREF = gi: 8850235



/UG = Hs.278896 UDP glycosyltransferase



1 family, polypeptide A1



/FL = gb: M57899.1 gb: NM_000463.1


Serine (or cysteine)
gb: NM_000295.1 /DEF = Homo sapiens
202833_s_at


proteinase inhibitor,
serine (or cysteine) proteinase inhibitor,


clade A (alpha-1
clade A (alpha-1 antiproteinase,


antiproteinase,
antitrypsin), member 1 (SERPINA1),


antitrypsin), member 1
mRNA. /FEA = mRNA /GEN = SERPINA1


SEQ ID NOS: 63
/PROD = serine (or cysteine) proteinase


(nucleotide) and 122
inhibitor, cladeA (alpha-1 antiproteinase,


(amino acid)
antitrypsin), member 1



/DB_XREF = gi: 4505792 /UG = Hs.297681



serine (or cysteine) proteinase inhibitor,



clade A (alpha-1 antiproteinase,



antitrypsin), member 1



/FL = gb: AF130068.1 gb: M11465.1



gb: K01396.1 gb: NM_000295.1


Nerve growth factor
gb: NM_014380.1 /DEF = Homo sapiens
217963_s_at


receptor (TNFRSF16)
p75NTR-associated cell death executor;


associated protein 1
ovarian granulosa cell protein (13 kD)


SEQ ID NOS: 64
(DXS6984E), mRNA. /FEA = mRNA


(nucleotide) and 123
/GEN = DXS6984E /PROD = p75NTR-


(amino acid)
associated cell death executor;



ovariangranulosa cell protein (13 kD)



/DB_XREF = gi: 7657043 /UG = Hs.17775



p75NTR-associated cell death executor;



ovarian granulosa cell protein (13 kD)



/FL = gb: NM_014380.1 gb: AF187064.1


Collagen, type XVIII,
Consensus includes gb: NM_030582.1
209081_s_at


alpha 1
/DEF = Homo sapiens collagen, type XVIII,


SEQ ID NOS: 65
alpha 1 (COL18A1), mRNA. /FEA = CDS


(nucleotide) and 124
/GEN = COL18A1 /PROD = collagen, type


(amino acid)
XVIII, alpha 1 /DB_XREF = gi: 13385619



/UG = Hs.78409 collagen, type XVIII, alpha



1 /FL = gb: NM_030582.1 gb: AF018081.1



gb: AF184060.1 gb: NM_016214.1


Collagen, type IX, alpha 3
gb: NM_001853.1 /DEF = Homo sapiens
204724_s_at


SEQ ID NOS: 66
collagen, type IX, alpha 3 (COL9A3),


(nucleotide) and 125
mRNA. /FEA = mRNA /GEN = COL9A3


(amino acid)
/PROD = collagen, type IX, alpha 3



/DB_XREF = gi: 4502966 /UG = Hs.53563



collagen, type IX, alpha 3



/FL = gb: L41162.1 gb: NM_001853.1









The biomarkers have expression levels in the cells that are dependent on the activity of the EGFR signal transduction pathway and that are also highly correlated with EGFR modulator sensitivity exhibited by the cells. Biomarkers serve as useful molecular tools for predicting a response to EGFR modulators, preferably biological molecules, small molecules, and the like that affect EGFR kinase activity via direct or indirect inhibition or antagonism of EGFR kinase function or activity.


EGFR Modulators

As used herein, the term “EGFR modulator” is intended to mean a compound or drug that is a biological molecule or a small molecule that directly or indirectly modulates EGFR activity or the EGFR signal transduction pathway. Thus, compounds or drugs as used herein is intended to include both small molecules and biological molecules. Direct or indirect modulation includes activation or inhibition of EGFR activity or the EGFR signal transduction pathway. In one aspect, inhibition refers to inhibition of the binding of EGFR to an EGFR ligand such as, for example, EGF. In another aspect, inhibition refers to inhibition of the kinase activity of EGFR.


EGFR modulators include, for example, EGFR specific ligands, small molecule EGFR inhibitors, and EGFR monoclonal antibodies. In one aspect, the EGFR modulator inhibits EGFR activity and/or inhibits the EGFR signal transduction pathway. In another aspect, the EGFR modulator is an EGFR monoclonal antibody that inhibits EGFR activity and/or inhibits the EGFR signal transduction pathway.


EGFR modulators include biological molecules or small molecules. Biological molecules include all lipids and polymers of monosaccharides, amino acids, and nucleotides having a molecular weight greater than 450. Thus, biological molecules include, for example, oligosaccharides and polysaccharides; oligopeptides, polypeptides, peptides, and proteins; and oligonucleotides and polynucleotides. Oligonucleotides and polynucleotides include, for example, DNA and RNA.


Biological molecules further include derivatives of any of the molecules described above. For example, derivatives of biological molecules include lipid and glycosylation derivatives of oligopeptides, polypeptides, peptides, and proteins.


Derivatives of biological molecules further include lipid derivatives of oligosaccharides and polysaccharides, e.g., lipopolysaccharides. Most typically, biological molecules are antibodies, or functional equivalents of antibodies. Functional equivalents of antibodies have binding characteristics comparable to those of antibodies, and inhibit the growth of cells that express EGFR. Such functional equivalents include, for example, chimerized, humanized, and single chain antibodies as well as fragments thereof.


Functional equivalents of antibodies also include polypeptides with amino acid sequences substantially the same as the amino acid sequence of the variable or hypervariable regions of the antibodies. An amino acid sequence that is substantially the same as another sequence, but that differs from the other sequence by means of one or more substitutions, additions, and/or deletions, is considered to be an equivalent sequence. Preferably, less than 50%, more preferably less than 25%, and still more preferably less than 10%, of the number of amino acid residues in a sequence are substituted for, added to, or deleted from the protein.


The functional equivalent of an antibody is preferably a chimerized or humanized antibody. A chimerized antibody comprises the variable region of a non-human antibody and the constant region of a human antibody. A humanized antibody comprises the hypervariable region (CDRs) of a non-human antibody. The variable region other than the hypervariable region, e.g., the framework variable region, and the constant region of a humanized antibody are those of a human antibody.


Suitable variable and hypervariable regions of non-human antibodies may be derived from antibodies produced by any non-human mammal in which monoclonal antibodies are made. Suitable examples of mammals other than humans include, for example, rabbits, rats, mice, horses, goats, or primates.


Functional equivalents further include fragments of antibodies that have binding characteristics that are the same as, or are comparable to, those of the whole antibody. Suitable fragments of the antibody include any fragment that comprises a sufficient portion of the hypervariable (i.e., complementarity determining) region to bind specifically, and with sufficient affinity, to EGFR tyrosine kinase to inhibit growth of cells that express such receptors.


Such fragments may, for example, contain one or both Fab fragments or the F(ab′)2 fragment. Preferably, the antibody fragments contain all six complementarity determining regions of the whole antibody, although functional fragments containing fewer than all of such regions, such as three, four, or five CDRs, are also included.


In one aspect, the fragments are single chain antibodies, or Fv fragments. Single chain antibodies are polypeptides that comprise at least the variable region of the heavy chain of the antibody linked to the variable region of the light chain, with or without an interconnecting linker. Thus, Fv fragment comprises the entire antibody combining site. These chains may be produced in bacteria or in eukaryotic cells.


The antibodies and functional equivalents may be members of any class of immunoglobulins, such as IgG, IgM, IgA, IgD, or IgE, and the subclasses thereof. In one aspect, the antibodies are members of the IgG1 subclass. The functional equivalents may also be equivalents of combinations of any of the above classes and subclasses.


In one aspect, EGFR antibodies can be selected from chimerized, humanized, fully human, and single chain antibodies derived from the murine antibody 225 described in U.S. Pat. No. 4,943,533 to Mendelsohn et al., including, for example, cetuximab.


In another aspect, the EGFR antibody can be selected from the antibodies described in U.S. Pat. No. 6,235,883 to Jakobovits et al., U.S. Pat. No. 5,558,864 to Bendi et al., and U.S. Pat. No. 5,891,996 to Mateo de Acosta del R10 et al.


In addition to the biological molecules discussed above, the EGFR modulators useful in the invention may also be small molecules. Any molecule that is not a biological molecule is considered herein to be a small molecule. Some examples of small molecules include organic compounds, organometallic compounds, salts of organic and organometallic compounds, saccharides, amino acids, and nucleotides. Small molecules further include molecules that would otherwise be considered biological molecules, except their molecular weight is not greater than 450. Thus, small molecules may be lipids, oligosaccharides, oligopeptides, and oligonucleotides and their derivatives, having a molecular weight of 450 or less.


It is emphasized that small molecules can have any molecular weight. They are merely called small molecules because they typically have molecular weights less than 450. Small molecules include compounds that are found in nature as well as synthetic compounds. In one embodiment, the EGFR modulator is a small molecule that inhibits the growth of tumor cells that express EGFR. In another embodiment, the EGFR modulator is a small molecule that inhibits the growth of refractory tumor cells that express EGFR. In yet another embodiment, the EGFR modulator is erlotinib HCl or gefitinib.


Numerous small molecules have been described as being useful to inhibit EGFR. For example, U.S. Pat. No. 5,656,655 to Spada et al. discloses styryl substituted heteroaryl compounds that inhibit EGFR. The heteroaryl group is a monocyclic ring with one or two heteroatoms, or a bicyclic ring with 1 to about 4 heteroatoms, the compound being optionally substituted or polysubstituted.


U.S. Pat. No. 5,646,153 to Spada et al. discloses bis mono and/or bicyclic aryl heteroaryl, carbocyclic, and heterocarbocyclic compounds that inhibit EGFR.


U.S. Pat. No. 5,679,683 to Bridges et al. discloses tricyclic pyrimidine compounds that inhibit the EGFR. The compounds are fused heterocyclic pyrimidine derivatives described at column 3, line 35 to column 5, line 6.


U.S. Pat. No. 5,616,582 to Barker discloses quinazoline derivatives that have receptor tyrosine kinase inhibitory activity.


Fry et al., Science 265, 1093-1095 (1994) in FIG. 1 discloses a compound having a structure that inhibits EGFR.


Osherov et al. disclose tyrphostins that inhibit EGFR/HER1 and HER 2, particularly those in Tables I, II, III, and IV.


U.S. Pat. No. 5,196,446 to Levitzki et al. discloses heteroarylethenediyl or heteroarylethendeiylaryl compounds that inhibit EGFR, particularly from column 2, line 42 to column 3, line 40.


Panek et al., Journal of Pharmacology and Experimental Therapeutics 283, 1433-1444 (1997) discloses a compound identified as PD166285 that inhibits the EGFR, PDGFR, and FGFR families of receptors. PD166285 is identified as 6-(2,6-dichlorophenyl)-2-(4-(2-diethylaminoethyoxy)phenylamino)-8-methyl-8H-pyrido(2,3-d)pyrimidin-7-one having the structure shown in FIG. 1 on page 1436.


Biomarkers and Biomarker Sets

The invention includes individual biomarkers and biomarker sets having both diagnostic and prognostic value in disease areas in which signaling through EGFR or the EGFR pathway is of importance, e.g., in cancers or tumors, in immunological disorders, conditions or dysfunction, or in disease states in which cell signaling and/or cellular proliferation controls are abnormal or aberrant. The biomarker sets comprise a plurality of biomarkers such as, for example, a plurality of the biomarkers provided in Table 1, that highly correlate with resistance or sensitivity to one or more EGFR modulators.


The biomarker sets of the invention enable one to predict or reasonably foretell the likely effect of one or more EGFR modulators in different biological systems or for cellular responses. The biomarker sets can be used in in vitro assays of EGFR modulator response by test cells to predict in vivo outcome. In accordance with the invention, the various biomarker sets described herein, or the combination of these biomarker sets with other biomarkers or markers, can be used, for example, to predict how patients with cancer might respond to therapeutic intervention with one or more EGFR modulators.


A biomarker set of cellular gene expression patterns correlating with sensitivity or resistance of cells following exposure of the cells to one or more EGFR modulators provides a useful tool for screening one or tumor samples before treatment with the EGFR modulator. The screening allows a prediction of cells of a tumor sample exposed to one or more EGFR modulators, based on the expression results of the biomarker set, as to whether or not the tumor, and hence a patient harboring the tumor, will or will not respond to treatment with the EGFR modulator.


The biomarker or biomarker set can also be used as described herein for monitoring the progress of disease treatment or therapy in those patients undergoing treatment for a disease involving an EGFR modulator.


The biomarkers also serve as targets for the development of therapies for disease treatment. Such targets may be particularly applicable to treatment of colon disease, such as colon cancers or tumors. Indeed, because these biomarkers are differentially expressed in sensitive and resistant cells, their expression patterns are correlated with relative intrinsic sensitivity of cells to treatment with EGFR modulators. Accordingly, the biomarkers highly expressed in resistant cells may serve as targets for the development of new therapies for the tumors which are resistant to EGFR modulators, particularly EGFR inhibitors.


Microarrays

The invention also includes specialized microarrays, e.g., oligonucleotide microarrays or cDNA microarrays, comprising one or more biomarkers, showing expression profiles that correlate with either sensitivity or resistance to one or more EGFR modulators. Such microarrays can be employed in in vitro assays for assessing the expression level of the biomarkers in the test cells from tumor biopsies, and determining whether these test cells are likely to be resistant or sensitive to EGFR modulators. For example, a specialized microarray can be prepared using all the biomarkers, or subsets thereof, as described herein and shown in Table 1. Cells from a tissue or organ biopsy can be isolated and exposed to one or more of the EGFR modulators. Following application of nucleic acids isolated from both untreated and treated cells to one or more of the specialized microarrays, the pattern of gene expression of the tested cells can be determined and compared with that of the biomarker pattern from the control panel of cells used to create the biomarker set on the microarray. Based upon the gene expression pattern results from the cells that underwent testing, it can be determined if the cells show a resistant or a sensitive profile of gene expression. Whether or not the tested cells from a tissue or organ biopsy will respond to one or more of the EGFR modulators and the course of treatment or therapy can then be determined or evaluated based on the information gleaned from the results of the specialized microarray analysis.


Antibodies

The invention also includes antibodies, including polyclonal or monoclonal, directed against one or more of the polypeptide biomarkers. Such antibodies can be used in a variety of ways, for example, to purify, detect, and target the biomarkers of the invention, including both in vitro and in vivo diagnostic, detection, screening, and/or therapeutic methods.


Kits

The invention also includes kits for determining or predicting whether a patient would be susceptible or resistant to a treatment that comprises one or more EGFR modulators. The patient may have a cancer or tumor such as, for example, a colon cancer or tumor. Such kits would be useful in a clinical setting for use in testing a patient's biopsied tumor or cancer samples, for example, to determine or predict if the patient's tumor or cancer will be resistant or sensitive to a given treatment or therapy with an EGFR modulator. The kit comprises a suitable container that comprises: one or more microarrays, e.g., oligonucleotide microarrays or cDNA microarrays, that comprise those biomarkers that correlate with resistance and sensitivity to EGFR modulators, particularly EGFR inhibitors; one or more EGFR modulators for use in testing cells from patient tissue specimens or patient samples; and instructions for use. In addition, kits contemplated by the invention can further include, for example, reagents or materials for monitoring the expression of biomarkers of the invention at the level of mRNA or protein, using other techniques and systems practiced in the art such as, for example, RT-PCR assays, which employ primers designed on the basis of one or more of the biomarkers described herein, immunoassays, such as enzyme linked immunosorbent assays (ELISAs), immunoblotting, e.g., Western blots, or in situ hybridization, and the like, as further described herein.


Application of Biomarkers and Biomarker Sets

The biomarkers and biomarker sets may be used in different applications. Biomarker sets can be built from any combination of biomarkers listed in Table 1 to make predictions about the likely effect of any EGFR modulator in different biological systems. The various biomarkers and biomarkers sets described herein can be used, for example, as diagnostic or prognostic indicators in disease management, to predict how patients with cancer might respond to therapeutic intervention with compounds that modulate the EGFR, and to predict how patients might respond to therapeutic intervention that modulates signaling through the entire EGFR regulatory pathway.


While the data described herein were generated in cell lines that are routinely used to screen and identify compounds that have potential utility for cancer therapy, the biomarkers have both diagnostic and prognostic value in other diseases areas in which signaling through EGFR or the EGFR pathway is of importance, e.g., in immunology, or in cancers or tumors in which cell signaling and/or proliferation controls have gone awry.


In accordance with the invention, cells from a patient tissue sample, e.g., a tumor or cancer biopsy, can be assayed to determine the expression pattern of one or more biomarkers prior to treatment with one or more EGFR modulators. Success or failure of a treatment can be determined based on the biomarker expression pattern of the cells from the test tissue (test cells), e.g., tumor or cancer biopsy, as being relatively similar or different from the expression pattern of a control set of the one or more biomarkers. Thus, if the test cells show a biomarker expression profile which corresponds to that of the biomarkers in the control panel of cells which are sensitive to the EGFR modulator, it is highly likely or predicted that the individual's cancer or tumor will respond favorably to treatment with the EGFR modulator. By contrast, if the test cells show a biomarker expression pattern corresponding to that of the biomarkers of the control panel of cells which are resistant to the EGFR modulator, it is highly likely or predicted that the individual's cancer or tumor will not respond to treatment with the EGFR modulator.


The invention also provides a method of monitoring the treatment of a patient having a disease treatable by one or more EGFR modulators. The isolated test cells from the patient's tissue sample, e.g., a tumor biopsy or tumor sample, can be assayed to determine the expression pattern of one or more biomarkers before and after exposure to an EGFR modulator wherein, preferably, the EGFR modulator is an EGFR inhibitor. The resulting biomarker expression profile of the test cells before and after treatment is compared with that of one or more biomarkers as described and shown herein to be highly expressed in the control panel of cells that are either resistant or sensitive to an EGFR modulator. Thus, if a patient's response is sensitive to treatment by an EGFR modulator, based on correlation of the expression profile of the one or biomarkers, the patient's treatment prognosis can be qualified as favorable and treatment can continue. Also, if, after treatment with an EGFR modulator, the test cells don't show a change in the biomarker expression profile corresponding to the control panel of cells that are sensitive to the EGFR modulator, it can serve as an indicator that the current treatment should be modified, changed, or even discontinued. This monitoring process can indicate success or failure of a patient's treatment with an EGFR modulator and such monitoring processes can be repeated as necessary or desired.


The biomarkers of the invention can be used to predict an outcome prior to having any knowledge about a biological system. Essentially, a biomarker can be considered to be a statistical tool. Biomarkers are useful primarily in predicting the phenotype that is used to classify the biological system. In an embodiment of the invention, the goal of the prediction is to classify cancer cells as having an active or inactive EGFR pathway. Cancer cells with an inactive EGFR pathway can be considered resistant to treatment with an EGFR modulator. An inactive EGFR pathway is defined herein as a non-significant expression of the EGFR or by a classification as “resistant” or “sensitive” based on the IC50 value of each colon cell line to EGFR inhibitor compound as exemplified herein.


However, although the complete function of all of the biomarkers are not currently known, some of the biomarkers are likely to be directly or indirectly involved in the EGFR signaling pathway. In addition, some of the biomarkers may function in the metabolic or other resistance pathways specific to the EGFR modulators tested. Notwithstanding, knowledge about the function of the biomarkers is not a requisite for determining the accuracy of a biomarker according to the practice of the invention.


EXAMPLES
Example 1
Identification of Biomarkers

The biomarkers of Table 1 were identified as follows.


Colon Tumors and Patients:

Forty colon tumors collected from the University of London between 1998 and 2002. The median age of the patients was 70 years (range: 26-91 years). The patients were diagnosed as follows: 6 patients were designated as Duke's A, 14 as Duke's B, and 20 as Duke's C. None of the patients were treated pre-operatively, and 13 were treated post-operatively.


Determination of Relative Drug Sensitivity in Colon Cancer Cell Lines:

The cell line filtering process used is illustrated in FIG. 2.


The colon cancer cell lines were grown using standard cell culture conditions: RPMI 1640 supplemented to contain 10% fetal bovine serum, 100 IU/ml penicillin, 100 mg/ml streptomycin, 2 mM L-glutamine and 10 mM Hepes (all from GibcoBRL, Rockville, Md.). Twenty-one colon cancer cell lines were examined for their relative sensitivity to a pair of small molecule EGFR inhibitors, erlotinib HCl and gefitinib. Cytotoxicity was assessed in cells by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphenyl)-2H-tetrazolium, inner salt)assay (T. L. Riss et al., 1992, Mol. Biol. Cell, 3 (Suppl.):184a). To carry out the assays, the colon cancer cells were plated at 4,000 cells/well in 96 well microtiter plates and 24 hours later serial diluted drugs were added. The concentration range for the EGFR inhibitor compounds used in the cytotoxicity assays was 50 ug/ml to 0.0016 ug/ml (roughly 100 uM to 0.0032 uM). The cells were incubated at 37° C. for 72 hours at which time the tetrazolium dye MTS (333 ug/ml final concentration in combination with the electron coupling agent phenazine methosulfate) was added. A dehydrogenase enzyme in live cells reduces the MTS to a form that absorbs light at 492 nm that can be quantified spectrophotometrically. The greater the absorbency, the greater the number of live cells. The results, provided below in Table 2 and FIG. 3, are expressed as an IC50, which is the drug concentration required to inhibit cell proliferation to 50% of that of untreated cells.









TABLE 2







Colon Cell Lines











Cell Line
ATCC No.
Avg. IC50







CaCo2
HTB-37
 5.4



Colo 201
CCL-224
10+



Colo 205
CCL-222
10+



CS-1

10+



Difi

 1



DLD-1

20



Geo

 3.6



HCT116
CCL-247
67+



HCT116S542

53



HCT-8
CCL-244
10+



HT-29
HTB-38
10+



Lovo
CCL-229LS174T
 3



LS1034

68+



RKORM13

29



SW1116

20



SW403

 6.2



SW480
CCL-228
10+



SW837
CCL-235
 7



SW948

73+



T84
CCL-248
10+



WiDr

67+










Resistance/Sensitivity Classification:

Two separate analyses were performed using different cut-offs to define EGFR-inhibitor resistance. For the first (designated “6-15”), the 6 cell lines with an IC50 at or below 7 uM were defined as sensitive and the remaining 15 cell lines were defined as resistant. For the second (designated “3-18”), the 3 cell lines with an IC50 below 4 uM were defined as sensitive and the remaining 18 cell lines were defined as resistant.


Gene Expression Profiling:

RNA was isolated from 50-70% confluent cell lines or colon cancer tumor tissue using the Rneasy kits from Qiagen (Valencia, Calif.). The quality of RNA was checked by measuring the 28S:18: ribosomal RNA ratio using and Agilent 2100 bioanalyzer (Agilent Technologies, Rockville, Md.). Concentration of total RNA was determined spectrophotemetrically. 10 ug of total RNA was used to prepare biotyinylated probes according to the Affymetrix Genechip Expression Analysis Technical Manual. Targets were hybridized to human HG-U133A gene chips according to the manufacturers instructions. Data were preprocessed using the MAS 5.0 software (Affymetrix, Santa Clara, Calif.). The trimmed mean intensity for each chip was scaled to 1,500 to account for minor differences in global chip intensity so that the overall expression level for each sample is comparable.


Data Analysis


All 22,215 probes (gene sequences) present on the U133A chip were considered as potential predictive biomarkers. To restrict the analysis to gene sequences expressed at a moderate level in colon tumor(s), gene sequences without at least one expression value of 2× the mean value for the array (3000 expression units) were removed leaving 6988 gene sequences. Next, to identify genes with variable expression in colon tumors (and therefore more likely to be able to correlate with variability in response to treatment), gene sequences with a VARP value (using log 10-transformed data)<0.1 were removed leaving 745 gene sequences. Next, the same expression and variance filters were applied to the remaining 745 gene sequences using the colon cell line data, reducing to 332 gene sequences for analysis (FIG. 1).


The 332 gene sequences were then subjected to a two-sided T-test using the Resistance/sensitivity classifications of the cell lines described above (FIG. 3). A total of 12 gene sequences had a p-value of <0.05 for both analyses (T-test Results I, FIG. 4). For the “6-15” analysis, 19 gene sequences were found to have a p-value <0.05 (T-Test Results II, FIG. 5). For the “3-18” analysis, 29 gene sequences were found to have a p-value <0.05 (T-test Results III, FIG. 6). Table 1 provides the biomarkers identified using the two-sided T-test.


Example 2
Untreated Xenograph Profiles

In Example 1, biomarkers were identified using sensitivity resistance profiles of cell lines to gefitinib and erlotinib HCl. The present example provided efficacy data for cetuximab (C225) in the colon cancer xenograft models Geo (sensitive to C225) and HT29 (resistant to C225).


In Vivo Antitumor Testing

Tumors were propagated in nude mice as subcutaneous (sc) transplants using tumor fragments obtained from donor mice. Tumor passage occurred approximately every two to four weeks. Tumors were then allowed to grow to the pre-determined size window (usually between 100-200 mg, tumors outside the range were excluded) and animals were evenly distributed to various treatment and control groups. Animals were treated with C225 (1 mg/mouse q3d X 10, 14, ip). Treated animals were checked daily for treatment related toxicity/mortality. Each group of animals was weighed before the initiation of treatment (Wt1) and then again following the last treatment dose (Wt2). The difference in body weight (Wt2-Wt1) provided a measure of treatment-related toxicity. Tumor response was determined by measurement of tumors with a caliper twice a week, until the tumors reached a predetermined target size of 1 gm or became necrotic. Tumor weights (mg) were estimated from the formula:





Tumor weight=(length×width2)/2


Antitumor activity was determined in terms of primary tumor growth inhibition. This was determined in two ways: (i) calculating the relative median tumor weight (MTW) of treated (T) and control (C) mice at various time points (effects were expressed as % T/C); and (ii) calculating the tumor growth delay (T-C value), defined as the difference in time (days) required for the treated tumors (T) to reach a predetermined target size compared to those of the control group (C). Statistical evaluations of data were performed using Gehan's generalized Wilcoxon test for comparisons of time to reach tumor target size (Gehan 1965). Statistical significance was declared at p<0.05. Antitumor activity was defined as a continuous MTW % T/C≦50% for at least 1 tumor volume doubling time (TVDT) any time after the start of treatment, where TVDT (tumor volume doubling time)=median time (days) for control tumors to reach target size−median time (days) for control tumors to reach half the target size. In addition, treatment groups had to be accompanied by a statistically significant tumor growth delay (T-C value) (p<0.05) to be termed active.


Treated animals were checked daily for treatment related toxicity/mortality. When death occurred, the day of death was recorded. Treated mice dying prior to having their tumors reach target size were considered to have died from drug toxicity. No control mice died bearing tumors less than target size. Treatment groups with more than one death caused by drug toxicity were considered to have had excessively toxic treatments and their data were not included in the evaluation of the compound's antitumor efficacy.


Table 3 provides the resulting untreated xenograph profiles.









TABLE 3







Untreated Xenograph Profiles












Differential expression in





Geo (sensitive) and HT-29
Absence and




(resistant) untreated
Presence of


Biomarker
Probe
xenografts
HT-29 and Geo





transforming growth
201506_at
Higher 373X in Geo than
HT-29 Absent


factor, beta-induced,

HT-29 (Absent)
Geo Present


68 kD


carcinoembryonic
201884_at
Higher 85X in Geo than HT-
HT-29 Absent


antigen-related cell

29 (Absent)
Geo Present


adhesion molecule 5


nerve growth factor
217963_s_at
Higher 50X in Geo than HT-
HT-29 Absent


receptor (TNFRSF16)

29 (Absent)
Geo Present


associated protein 1


carcinoembryonic
211657_at
Higher 23X in Geo than HT-
HT-29 Absent


antigen-related cell

29(Absent)
Geo Present


adhesion molecule 6


(non-specific cross


reacting antigen)


annexin A1
201012_at
Higher 16X in Geo than HT-
HT-29 Absent




29 (Absent)
Geo Present


tumor protein p53 (Li-
211300_s_at
Higher 11X in Geo than HT-
HT-29 Absent


Fraumeni syndrome)

29 (Absent)
Geo Present


DVS27-related protein
209821_at
Higher 9X in Geo than HT-
HT-29 Absent




29 (Absent)
Geo Present


cystic fibrosis
205043_at
Higher 7X in Geo than HT-
HT-29 Absent


transmembrane

29 (Absent)
Geo Present


conductance regulator,


ATP-binding cassette


(sub-family C,


member 7)


serine (or cysteine)
211429_s_at
Higher 7X in Geo than HT-
HT-29 Absent


proteinase inhibitor,

29 (Absent)
Geo Present


clade A (alpha-1


antiproteinase,


antitrypsin), member 1


bone morphogenetic
209591_s_at
Higher 4X in Geo than HT-
HT-29 Absent


protein 7 (osteogenic

29 (Absent)
Geo Present


protein 1)


interferon-stimulated
205483_s_at
Higher 3X in Geo than HT-
HT-29 Absent


protein, 15 kDa

29(Absent)
Geo Present


S100 calcium-binding
204351_at
Higher 11X in Geo than HT-
HT-29 Present


protein P

29
Geo Present


carcinoembryonic
203757_s_at
Higher 8X in Geo than HT-
HT-29 Present


antigen-related cell

29
Geo Present


adhesion molecule 6


(non-specific cross


reacting antigen)


putative integral
208029_s_at
Higher 7X in Geo than HT-
HT-29 Present


membrane transporter

29
Geo Present


cadherin 17, LI
209847_at
Higher 4X in Geo than HT-
HT-29 Present


cadherin (liver-

29
Geo Present


intestine)


FXYD domain-
202489_s_at
Higher 3X in Geo than HT-
HT-29 Present


containing ion

29
Geo Present


transport regulator 3


insulin-like growth
202718_at
Higher 3X in Geo than HT-
HT-29 Present


factor binding protein

29
Geo Present


2 (36 kD)


eukaryotic translation
201123_s_at
Higher 3X in Geo than HT-
HT-29 Present


initiation factor 5A

29
Geo Present


3-hydroxy-3-
204607_at
Higher 2X in Geo than HT-
HT-29 Present


methylglutaryl-

29
Geo Present


Coenzyme A synthase


2 (mitochondrial)


serine (or cysteine)
202833_s_at
Higher 21X in HT-29 than
HT-29 Present


proteinase inhibitor,

Geo
Geo Present


clade A (alpha-1


antiproteinase,


antitrypsin), member 1


transmembrane
211689_s_at
Higher 7X in HT-29 than
HT-29 Present


protease, serine 2

Geo
Geo Present


protease inhibitor 3,
41469_at
Higher 6X in HT-29 than
HT-29 Present


skin-derived (SKALP)

Geo
Geo Present


serine (or cysteine)
204855_at
Higher 4X in HT-29 than
HT-29 Present


proteinase inhibitor,

Geo
Geo Present


clade B (ovalbumin),


member 5


fibroblast growth
204379_s_at
Higher 3X in HT-29 than
HT-29 Present


factor receptor 3

Geo
Geo Present


(achondroplasia,


thanatophoric


dwarfism)


mucin 3B
214898_x_at
Higher 3X in HT-29 than
HT-29 Present




Geo
Geo Present


fucosyltransferase 3
214088_s_at
Higher 3X in HT-29 than
HT-29 Present


(galactoside 3(4)-L-

Geo
Geo Present


fucosyltransferase,


Lewis blood group


included)


phospholipase A2,
203649_s_at
Higher 2X in HT-29 than
HT-29 Present


group IIA (platelets,

Geo
Geo Present


synovial fluid)


A kinase (PRKA)
210517_s_at
Higher 339X in HT-29 than
HT-29 Present


anchor protein

Geo (Absent)
Geo Absent


(gravin) 12


serine (or cysteine)
202628_s_at
Higher 280X in HT-29 than
HT-29 Present


proteinase inhibitor,

Geo (Absent)
Geo Absent


clade E (nexin,


plasminogen activator


inhibitor type 1),


member 1


ESTs, Highly similar
215125_s_at
Higher 75X in HT-29 than
HT-29 Present


to A39092

Geo (Absent)
Geo Absent


glucuronosyltransferase


[H. sapiens]


Purkinje cell protein 4
205549_at
Higher 38X in HT-29 than
HT-29 Present




Geo (Absent)
Geo Absent


lectin, galactoside-
201105_at
Higher 33X in HT-29 than
HT-29 Present


binding, soluble, 1

Geo (Absent)
Geo Absent


(galectin 1)


old astrocyte
213059_at
Higher 29X in HT-29 than
HT-29 Present


specifically induced

Geo (Absent)
Geo Absent


substance


UDP
208596_s_at
Higher 23X in HT-29 than
HT-29 Present


glycosyltransferase 1

Geo (Absent)
Geo Absent


family, polypeptide


A3


hypothetical protein
213343_s_at
Higher 21X in HT-29 than
HT-29 Present


PP1665

Geo (Absent)
Geo Absent


membrane protein,
202974_at
Higher 9X in HT-29 than
HT-29 Present


palmitoylated 1

Geo (Absent)
Geo Absent


(55 kD)


caudal type homeo
206387_at
Higher 8X in HT-29 than
HT-29 Present


box transcription

Geo (Absent)
Geo Absent


factor 2


polymeric
204213_at
Higher 7X in HT-29 than
HT-29 Present


immunoglobulin

Geo (Absent)
Geo Absent


receptor


mucin 5, subtypes A
214385_s_at
Higher 6X in HT-29 than
HT-29 Present


and C,

Geo (Absent)
Geo Absent


tracheobronchial/gastric


metallothionein 1G
204745_x_at
Higher 2X in HT-29 than
HT-29 Present




Geo (Absent)
Geo Absent


inhibitor of DNA
207826_s_at
Higher 2X in HT-29 than
HT-29 Present


binding 3, dominant

Geo (Absent)
Geo Absent


negative helix-loop-


helix protein


lymphocyte antigen 75
205668_at
not differentially expressed
HT-29 Present





Geo Absent


secretory leukocyte
203021_at
not differentially expressed
HT-29 Present


protease inhibitor


Geo Absent


(antileukoproteinase)


dopa decarboxylase
205311_at
not differentially expressed
HT-29 Present


(aromatic L-amino


Geo Absent


acid decarboxylase)


G protein-coupled
213880_at
not differentially expressed
HT-29 Present


receptor 49


Geo Absent


interferon, alpha-
202411_at
not differentially expressed
HT-29 Present


inducible protein 27


Geo Absent



Homo sapiens CD44

210916_s_at
not differentially expressed
HT-29 Present


isoform RC (CD44)


Geo Absent


mRNA, complete cds


mucin 5, subtypes A
214303_x_at
absent in HT-29 and Geo
HT-29 Absent


and C,


Geo Absent


tracheobronchial/gastric


UDP
207126_x_at
absent in HT-29 and Geo
HT-29 Absent


glycosyltransferase 1


Geo Absent


family, polypeptide


A1


metallothionein 1F
217165_x_at
absent in HT-29 and Geo
HT-29 Absent


(functional)


Geo Absent


GRO3 oncogene
207850_at
absent in HT-29 and Geo
HT-29 Absent





Geo Absent


protease inhibitor 3,
203691_at
absent in HT-29 and Geo
HT-29 Absent


skin-derived (SKALP)


Geo Absent


annexin A10
210143_at
absent in HT-29 and Geo
HT-29 Absent





Geo Absent


protein tyrosine
203029_s_at
absent in HT-29 and Geo
HT-29 Absent


phosphatase, receptor


Geo Absent


type, N polypeptide 2


solute carrier family 7
202752_x_at
absent in HT-29 and Geo
HT-29 Absent


(cationic amino acid


Geo Absent


transporter, y+


system), member 8


collagen, type XVIII,
209081_s_at
absent in HT-29 and Geo
HT-29 Absent


alpha 1


Geo Absent


collagen, type IX,
204724_s_at
absent in HT-29 and Geo
HT-29 Absent


alpha 3


Geo Absent


alpha-2-HS-
210929_s_at
?
HT-29 Absent


glycoprotein


Geo Absent


metallothionein 1X
208581_x_at
?
HT-29 Absent





Geo Absent


tumor necrosis factor
206467_x_at
?
HT-29 Absent


receptor superfamily,


Geo Absent


member 6b, decoy









Example 3
Production of Antibodies Against the Biomarkers

Antibodies against the biomarkers can be prepared by a variety of methods. For example, cells expressing an biomarker polypeptide can be administered to an animal to induce the production of sera containing polyclonal antibodies directed to the expressed polypeptides. In one aspect, the biomarker protein is prepared and isolated or otherwise purified to render it substantially free of natural contaminants, using techniques commonly practiced in the art. Such a preparation is then introduced into an animal in order to produce polyclonal antisera of greater specific activity for the expressed and isolated polypeptide.


In one aspect, the antibodies of the invention are monoclonal antibodies (or protein binding fragments thereof). Cells expressing the biomarker polypeptide can be cultured in any suitable tissue culture medium, however, it is preferable to culture cells in Earle's modified Eagle's medium supplemented to contain 10% fetal bovine serum (inactivated at about 56° C.), and supplemented to contain about 10 g/l nonessential amino acids, about 1,00 U/ml penicillin, and about 100 μg/ml streptomycin.


The splenocytes of immunized (and boosted) mice can be extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line can be employed in accordance with the invention, however, it is preferable to employ the parent myeloma cell line (SP2/0), available from the ATCC. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al. (1981, Gastroenterology, 80:225-232). The hybridoma cells obtained through such a selection are then assayed to identify those cell clones that secrete antibodies capable of binding to the polypeptide immunogen, or a portion thereof.


Alternatively, additional antibodies capable of binding to the biomarker polypeptide can be produced in a two-step procedure using anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens and, therefore, it is possible to obtain an antibody that binds to a second antibody. In accordance with this method, protein specific antibodies can be used to immunize an animal, preferably a mouse. The splenocytes of such an immunized animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones that produce an antibody whose ability to bind to the protein-specific antibody can be blocked by the polypeptide. Such antibodies comprise anti-idiotypic antibodies to the protein-specific antibody and can be used to immunize an animal to induce the formation of further protein-specific antibodies.


Example 4
Immunofluorescence Assays

The following immunofluorescence protocol may be used, for example, to verify EGFR biomarker protein expression on cells or, for example, to check for the presence of one or more antibodies that bind EGFR biomarkers expressed on the surface of cells. Briefly, Lab-Tek II chamber slides are coated overnight at 4° C. with 10 micrograms/milliliter (μg/ml) of bovine collagen Type II in DPBS containing calcium and magnesium (DPBS++). The slides are then washed twice with cold DPBS++ and seeded with 8000 CHO—CCR5 or CHO pC4 transfected cells in a total volume of 125 μl and incubated at 37° C. in the presence of 95% oxygen/5% carbon dioxide.


The culture medium is gently removed by aspiration and the adherent cells are washed twice with DPBS++ at ambient temperature. The slides are blocked with DPBS++ containing 0.2% BSA (blocker) at 0-4° C. for one hour. The blocking solution is gently removed by aspiration, and 125 μl of antibody containing solution (an antibody containing solution may be, for example, a hybridoma culture supernatant which is usually used undiluted, or serum/plasma which is usually diluted, e.g., a dilution of about 1/100 dilution). The slides are incubated for 1 hour at 0-4° C. Antibody solutions are then gently removed by aspiration and the cells are washed five times with 400 μl of ice cold blocking solution. Next, 125 μl of 1 μg/ml rhodamine labeled secondary antibody (e.g., anti-human IgG) in blocker solution is added to the cells. Again, cells are incubated for 1 hour at 0-4° C.


The secondary antibody solution is then gently removed by aspiration and the cells are washed three times with 400 μl of ice cold blocking solution, and five times with cold DPBS++. The cells are then fixed with 125 μl of 3.7% formaldehyde in DPBS++ for 15 minutes at ambient temperature. Thereafter, the cells are washed five times with 400 μl of DPBS++ at ambient temperature. Finally, the cells are mounted in 50% aqueous glycerol and viewed in a fluorescence microscope using rhodamine filters.

Claims
  • 1. A method for identifying a mammal that will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises: (a) measuring in the mammal the level of at least one biomarker selected from the biomarkers of Table 1;(b) exposing the mammal to the EGFR modulator;(c) following the exposing of step (b), measuring in the mammal the level of the at least one biomarker,wherein a difference in the level of the at least one biomarker measured in step (c) compared to the level of the at least one biomarker measured in step (a) indicates that the mammal will respond therapeutically to said method of treating cancer.
  • 2. The method of claim 1 wherein the method is an in vitro method, and wherein the at least one biomarker is measured in at least one mammalian biological sample from the mammal.
  • 3. A method for identifying a mammal that will respond therapeutically to a method of treating cancer comprising administering an EGFR modulator, wherein the method comprises: (a) exposing the mammal to the EGFR modulator;(b) following the exposing of step (a), measuring in the mammal the level of the at least one biomarker selected from the biomarkers of Table 1,wherein a difference in the level of the at least one biomarker measured in step (b), compared to the level of the biomarker in a mammal that has not been exposed to said EGFR modulator, indicates that the mammal will respond therapeutically to said method of treating cancer.
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2005/000638 1/7/2005 WO 00 6/18/2009
Provisional Applications (1)
Number Date Country
60535151 Jan 2004 US