Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
A METHOD OF DIAGNOSING NEOPLASMS
Document Type and Number:
WIPO Patent Application WO/2009/052573
Kind Code:
A1
Abstract:
The present invention relates generally to nucleic acid molecules, the RNA and protein expression profiles of which are indicative of the onset, predisposition to the onset and/or progression of a neoplasm. More particularly, the present invention is directed to nucleic acid molecules, the expression profiles of which are indicative of the onset and/or progression of a large intestine neoplasm, such as an adenoma or an adenocarcinoma. The expression profiles of the present invention are useful in a range of applications including, but not limited to, those relating to the diagnosis and/or monitoring of colorectal neoplasms, such as colorectal adenocarcinomas. Accordingly, in a related aspect the present invention is directed to a method of screening a subject for the onset, predisposition to the onset and/or progression of a neoplasm by screening for modulation in the expression profile of one or more nucleic acid molecule markers.

Inventors:
LAPOINTE LAWRENCE CLARENCE (AU)
DUNNE ROBERT (AU)
YOUNG GRAEME P (AU)
LOCKETT TREVOR JOHN (AU)
WILSON WILLIAM J (AU)
Application Number:
PCT/AU2008/001571
Publication Date:
April 30, 2009
Filing Date:
October 23, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CLINICAL GENOMICS PTY LTD (AU)
COMMW SCIENT IND RES ORG (AU)
LAPOINTE LAWRENCE CLARENCE (AU)
DUNNE ROBERT (AU)
YOUNG GRAEME P (AU)
LOCKETT TREVOR JOHN (AU)
WILSON WILLIAM J (AU)
International Classes:
C12Q1/68
Domestic Patent References:
WO2001073027A22001-10-04
WO1995025116A11995-09-21
WO1995035505A11995-12-28
Foreign References:
US20060188889A12006-08-24
US20060035237A12006-02-16
US20070037204A12007-02-15
US5837832A1998-11-17
US6410229B12002-06-25
US5470967A1995-11-28
US5714331A1998-02-03
EP0225807A21987-06-16
US5419966A1995-05-30
US5445934A1995-08-29
US5384261A1995-01-24
US5700637A1997-12-23
Other References:
See also references of EP 2215258A4
MOORE ET AL., BBA, vol. 1402, 1988, pages 239 - 249
WEISSLEDER ET AL., NATURE, vol. 6, 2000, pages 351 - 355
WEDEMEYER ET AL., CLINICAL CHEMISTRY, vol. 48, no. 9, 2002, pages 1398 - 1405
ALON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 96, June 1999 (1999-06-01), pages 6745 - 6750
SCHENA ET AL., SCIENCE, vol. 270, 1995, pages 467 - 470
DERISI ET AL., NATURE GENETICS, vol. 14, 1996, pages 457 - 460
NIELSEN ET AL., SCIENCE, vol. 254, 1991, pages 1497 - 1500
NIELSEN, CURR. OPIN. BIOTECHNOL., vol. 10, 1999, pages 71 - 75
URDEA ET AL., NUCLEIC ACIDS SYMP. SER., vol. 24, 1991, pages 197 - 200
PEVZNER ET AL., J. BIOMOL. STRUC. & DYN., vol. 9, 1991, pages 399 - 410
MASKOS; SOUTHERN, NUC. ACIDS RES., vol. 20, 1992, pages 1679 - 84
T. SANO; C. R. CANTOR, BIO/TECHNOLOGY, vol. 9, 1991, pages 1378 - 81
SMITH ET AL., SCIENCE, vol. 258, 1992, pages 1122 - 1126
PEASE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, no. 11, 1994, pages 5022 - 5026
GUO ET AL., NUCLEIC ACIDS RES., vol. 22, 1994, pages 5456 - 5465
GERMER ET AL., GENOME RES., vol. 10, 2000, pages 258 - 266
HEID ET AL., GENOME RES., vol. 6, 1996, pages 986 - 994
KRAUS, M.; AARONSON, S., METHODS ENZYMOL., vol. 200, 1991, pages 546 - 556
BONNER ET AL., J. MOL. BIOL., vol. 81, 1973, pages 123
"Affymetrix. GeneChip expression data analysis fundamentals", AFFYMETRIX, SANTA CLARA, CA USA, 2001
HUBBELL ET AL., BIOINFORMATICS, vol. 18, 2002, pages 1585 - 1592
IRIZARRY ET AL., NUCLEIC ACID RESEARCH, vol. 31, 2003
G. K. SMYTH., STATISTICAL APPLICATIONS IN GENETICS AND MOLECULAR BIOLOGY, vol. 3, no. 1, 2004
HUBBELL E.W.; W. M. LIU; R. MEI.: "Robust estimators for expression analysis", BIOINFORMATICS, vol. 18, 2002, pages 1585 - 1592
IRIZARRY R.W; B. M. BOLSTAD; F. COLLIN; L. M. COPE; B. HOBBS; T. P. SPEED: "Summaries ofaffymetrix genechip probe level data", NUCLEIC ACID RESEARCH, vol. 31, 2003
SMYTH G.K.: "Linear models and empirical bayes methods for assessing differential expression in microarray experiments", STATISTICAL APPLICATIONS IN GENETICS AND MOLECULAR BIOLOGY, vol. 3, no. 1, 2004, pages 3
T. SANO; C. R. CANTOR, BIOLTECHNOLOGY, vol. 9, 1991, pages 1378 - 81
WEISSLEDER ET AL., NATURE MEDICINE, vol. 6, 2000, pages 351 - 355
Attorney, Agent or Firm:
OBRANOVICH, Tania, D. et al. (1 Nicholson StreetMelbourne, Victoria 3000, AU)
Download PDF:
Claims:

CLAIMS:

1. A method of screening for the onset or predisposition to the onset of a large intestine neoplasm or monitoring the progress of a neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 201328_at 221577_x_at 205828_at 201341_at 221922_at 205886_at 201416_at 60474_at 205890_s_at 201417_at 222696_at 205910_s_at 201468_s_at 223447_at 20594 l_s_at 201506_at 223970_at 206224_at 201563_at 22554 l_at 206976_s_at 201656_at 225835_at 207173_x_at 201925_s_at 226360_at 207457_s_at 201926_s_at 227174_at 208079_s_at 202286_s_at 227475_at 208712_at 202718_at 228303_at 209218_at 20283 l_at 228653_at 209309_at 202833_s_at 228754_at 209752_at 202935_s_at 228915_at 209773_s_at 202936_s_at 229215_at 209774_x_at 203124_s_at 231832_at 209792_s_at 203256_at 231941_s_at 209875_s_at 203313_s_at 232176_at 209955_s_at 203510_at 232252_at 210052_s_at 203860_at 23248 l_s_at 210511_s_at 203895_at 23433 l_s_at 210559_s_at 203896_s_at 235210_s_at 210766_s_at 20396 l_at 235976_at 211506_s_at 203962_s_at 236894_at 212281_s_at 204259_at 238017_at 212344_at 20435 l_at 212353_at 20440 l_at 238984_at 212354_at 204404 at 241031 at 213905 x at

204855_at 200660_at 214022_s_at

204885_s_at 200832_s_at 214974_x_at

205174_s_at 200903_s_at 215091_s_at

205366_s_at 201014_s_at 217430_x_at

205470_s_at 201112_s_at 217996_at

205513_at 201195_s_at 218507_at

205765_at 201261_x_at 218963_s_at

205825_at 201292_at 218984_at

205927_s_at 201338_x_at 219787_s_at

205983_at 201479_at 21991 l_s_at

206239_s_at 201577_at 221729_at

206286_s_at 201601_x_at 221730_at

207158_at 201666_at 221731_x_at

207850_at 202310_s_at 221923_s_at

209369_at 20231 1_s_at 37892_at

210445_at 202404_s_at 222449_at

210519_s_at 20243 l_s_at 222450_at

211429_s_at 202504_at 222549_at

212063_at 202779_s_at 222608_s_at

212070_at 202859_x_at 223062_s_at

212190_at 202954_at 224428_s_at

21253 l_at 202998_s_at 224646_x_at

212942_s_at 203083_at 224915_x_at

213880_at 203213_at 225295_at

213975_s_at 203878_s_at 225520_at

214235_at 20405 l_s_at 225664_at

214651_s_at 204127_at 22568 l_at

217523_at 204170_s_at 225767_at

217867_x_at 204320_at 225799_at

218086_at 204470_at 225806_at

21821 l_s_at 204475_at 226227_x_at

218704_at 204580_at 226237_at

218796_at 204620_s_at 226311_at

218872_at 204702_s_at 226777_at

219630_at 20536 l_s_at 226835_s_at

219682_s_at 205476_at 227140_at

219727_at 205479_s_at 229802_at

219955 at 205713 s at 231766 s at

219956_at 205815_at 232151 _at; and/or

200665_s_at

(ϋ) KIAAl 199 CDCA7 L0XL2

FOXQl RNF43 TDGFl AZGPl

MMPl CCL20 MTHFDlL MYC

TCNl CTSE ANLN COMP

MMP7 MSLN H19 AURKA

WDR72 TIMPl FAP PAICS

INHBA PCSKl DACHl PUS7

COLI lAl CSTl VCAN ZNRF3

GDF 15 BGN SQLE CCNDl

CTHRCl AXIN2 REG3A CSElL

COLlAl MET TESC PFDN4

LGR5 SOX9 UBE2C C20orf42

DUSP27 TMEPAI TMEM97 SLCl 1A2

SERPINB5 CDHI l TRIM29 NFE2L3

ASCL2 MMPI l KLKI l CEL

SULFl CXCLl LY6G6D NLFl

SLC6A6 QPCT SLC7A5 NPDCl

TACSTD2 PDZKlIPl FABP6 ENCl

NEBL CD55 SLITRK6 SERPINE2

PCCA ECT2 MLPH UBE2S

FLJ37644 COL12A1 HOXA9 T0P2A

KRT23 LlTDl TBX3 CDC2

CXCL3 WDR51B BACE2 RFC3

MMP3 HOXB6 GPX2 LILRBl

SFRP4 FAM84A TPX2 NPMl

UBD COL5A2 KCNN4 RDHE2

SCD MMP 12 LOC541471 PLAU

DPEPl SORD CYP3A5 HSPHl

LCN2 PSATl ANXA3 GTF3A

PLCB4 CXCL5 CYP3A5P2 KLKlO

SPINKl IGFBP2 C14orf94 GPSM2

DUOX2 TGIFl RP5-875H10.1 NMEl

SOX4 CXCL2 RPL22L1 MUC20

THBS2 SLCO4A1 APOBECl RPESP

CLDNl CKS2 HIG2 RRM2

REGlB PHLDAl COLlOAl DKCl

CDH3 SERPINAl AHCY CD44

SPARC COL 1A2 IL8

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

2. The method according to claim 1 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 22568 l_at; 227140_at; and/or

(ii) CTHRCl.

3. The method according to claim 1 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 227475_at, 204475_at, 202859_x_at, 202404_s_at; and/or

(ii) FOXQl, MMPl, IL8, COL1A2.

4. The method according to claim 1 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 205513_at, 204259_at, 227174_at, 21051 l_s_at, 37892_at; and/or

(ii) TCNl, MMP7, WDR72, INHBA, COLl IAl.

5. The method according to claim 1, wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

221577_x_at 212942_s_at 202310_s_at

213880_at 232252_at 204855_at

229215_at 212354_at 228754_at

202286_s_at 203962_s_at 203860_at

202935_s_at 218963_s_at 207850_at

205828_at 20405 l_s_at 205890_s_at

200832_s_at 205983_at 21253 l_at; and/or

(ϋ) KIAAl 199 GDF 15 COLlAl

LGR5 DUSP27 SERPINB5

ASCL2 SULFl SLC6A6

TACSTD2 NEBL PCCA

FL J37644 KRT23 CXCL3

MMP3 SFRP4 UBD

SCD DPEPl LCN2.

6. The method according to any one of claims 1-5 wherein said control level is a nonneoplastic level.

7. The method according to any one of claims 1-5 wherein said neoplastic cell is an adenoma or an adenocarcinoma.

8. The method according to any one of claims 1 to 3 wherein said neoplasm is an adenoma and said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

201328_at 205825_at 219956_at

201468_s_at 205927_s_at 221922_at

201656_at 206239_s_at 223447_at

201925_s_at 207158_at 223970_at

201926_s_at 210445_at 225835_at

202718_at 210519_s_at 226360_at

20283 l_at 211429_s_at 228303_at

202833_s_at 212063_at 228653_at

203124_s_at 213975_s_at 228915_at

203313_s_at 214235_at 231832_at

203860 at 214651 s at 231941 s at

203895_at 217523_at 232176_at

203896_s_at 217867_x_at 23248 l_s_at

20440 l_at 218086_at 23433 l_s_at

204885_s_at 21821 l_s_at 235210_s_at

205174_s_at 218796_at 235976_at

205366_s_at 219630_at 236894_at

205470_s_at 219682_s_at 238017_at

205513_at 219727_at 238984_at

205765_at 219955_at; 24103 l_at; and/or

(«) APOBECl HOXB6 QPCT

BACE2 IGFBP2 RDHE2

C20orf42 ITGA6 REG4

CD44 KCNN4 RETNLB

CD55 KLKI l RP5-875H10.1

CTSE LlTDl RPESP

CYP3A5 LILRBl SERPINAl

CYP3A5P2 MLPH SLCl 1A2

DACHl MSLN SLCl 2 A2

DUOX2 MUC20 SLITRK6

ETS2 NLFl SPINKl

FABP6 NPDCl TBX3

FAM84A NQOl TCNl

GALNT6 PCCA TGIFl

GPSM2 PCSKl WDR51B

GPX2 PDZKlIPl ZNRF3

HOXA9 PLCB4.

9. The method according to claim 1 wherein said neoplasm is a cancer and said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200660_at 205476_at 217430_x_at

200665_s_at 205479_s_at 217996_at

200832_s_at 205713_s_at 218507_at

200903_s_at 205815_at 218963_s_at

201014_s_at 205828_at 218984_at

201112 s at 205886 at 219787 s at

201195 s_at 205890_ s at 219911_ s_at

201261_ x_at 205910_ s at 221729_ at

201292 at 205941_ s_at 221730_ at

201338_ x_at 206224_ at 221731_ x at

201479_ at 206976_ _s_at 221923_ s_at

201577 . at 207173_ x_at 37892_at

201601 . _x_at 207457_ _s_at 222449_ at

201666 . at 208079_ s at 222450_ at

202310_ _s_at 208712_ at 222549_ at

202311 , s at 209218_ at 222608_ s at

202403_ s at 209309 . at 223062_ s at

202404_ s at 209752_ at 224428_ s at

202431 , s at 209773_ s_at 224646_ x at

202504_ _at 209774_ x at 224915_ x at

202779 , s at 209792 s_at 225295_ at

202859 , x at 209875_ s_at 225520_ at

202954 . at 209955_ _s_at 225664_ at

202998_ _s_at 210052_ s_at 225681_ at

203083_ at 210511_ s_at 225767_ at

203213_ _at 210559_ s_at 225799_ at

203878 . _s_at 210766 . s_at 225806_ at

204051_ _s_at 211506 . s_at 226227_ x_at

204127_ at 212281 . s_at 226237_ at

204170 . _s_at 212344 . at 226311_ at

204320_ at 212353_ at 226777_ at

204470 at 212354 . at 226835_ s_at

204475 . at 213905 x_at 227140_ at

204580 at 214022 . s at 229802_ at

204620 . s at 214974 . x_at 231766_ s at

204702 . _s_at 215091 . s_at; 232151_ at; and/or

205361 . _s_at

(») AHCY DKCl PSATl

ANLN ECT2 PUS7

AURKA FAP REGlA

AZGPl GTF3A REGlB

BGN H19 REG3A

C14orf94 HIG2 RFC3

C20orω99 HSPHl RRM2

CCL20 IFITMl SlOOAI l

CCNDl IL8 SCD

CDC2 INHBA SFRP4

CDCA7 KLKlO SLC39A10

CDHI l KRT23 SLC7A5

CEL LOC541471 SLCO4A1

CKS2 LOXL2 SPARC

CLDNl LY6G6D SPPl

COLlOAl MMPl SQLE

COLI lAl MMPI l SULFl

COL12A1 MMP 12 THBS2

COLlAl MMP3 TIMPl

COL1A2 MTHFDlL TMEM97

COL5A2 MYC TMEPAI

COL8A1 NFE2L3 TOP2A

COMP NMEl TPX2

CSElL NPMl TRIM29

CSTl PAICS UBD

CTHRCl PFDN4 UBE2C

CXCLl PHLDAl UBE2S

CXCL2 PLAU VCAN

CXCL5.

10. A method of screening for the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

202286_s_at 235976_at 209309_at

204259_at 236894_at 211506_s_at

204885_s_at 214974_x_at

205174_s_at 238984_at 219787_s_at

205825_at 24103 l_at 37892_at

207850_at 202311_s_at 222608_s_at

213880_at 204320_at 223062_s_at

217523_at 204475_at 225806_at

227174 at 204702 s at 226237 at

228915_at 205910_s_at 227140_at

232252_at 206224_at 229802_at; and/or

(M) MMPl PCSKl ANLN

MMP7 CSTl DACHl

LGR5 QPCT COLI lAl

WDR72 ECT2 C14orf94

COLI lAl SLITRK6 AZGPl

COLlAl LlTDl REG4

DUSP27 KIAAl 199 NFE2L3

NLFl PSATl CEL

IL8 CXCL5 CD44

TACSTD2 CXCL3 COL8A1

MSLN in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of a neoplastic cell or a cell predisposed to the onset of a neoplastic state.

11. The method according to claim 10 wherein said neoplastic cell is an adenoma or adenocarcinoma.

12. The method according to any one of claims 10 to 11 wherein said neoplasm is an adenoma and said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

204885_s_at, 217523_at, 236894_at, 205174_s_at, 228915_at, 238984_at, 205825_at, 235976_at, 24103 l_at; and/or

(ii) CD44, MSLN, QPCT, DACHl, NLFl, REG4, LlTDl, PCSKl, SLITRK6.

13. The method according to any one of claims 13 to 15 wherein said neoplasm is a cancer and said one or more genes or transcripts are selected from:

(i) the gene or genes detected by Affymetrix probeset IDs:

202311_s_at 209309_at 223062_s_at

204320_at 211506_s_at 225806_at

204475_at 214974_x_at 226237_at

204702 s at 219787 s at 227140 at

205910_s_at 37892_at 229802_at; and/or

206224_at 222608_s_at

(ϋ) ANLN COLlAl IL8

AZGPl COL8A1 MMPl

C14orf94 CSTl NFE2L3

CEL CXCL5 PSATl

COLI lAl ECT2.

14. The method according to any one of claims 10-13 wherein said background level is the background of the testing methodology.

15. A method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(Q the gene, genes or 1 transcripts detected by Affymetrix probeset IDs

200884_ at 214234_ _s_at 226248_ s at

203240_ at 214235_ at 226302 . _at

203963_ at 214433_ _s_at 227676 . at

204508_ s_at 215125_ _s_at 227719 . .at

204607_ at 215867_ x at 227725 . at

204811_ s_at 217109_ at 228232 . s at

204895_ x at 217110_ s_at 229070 . at

204897_ at 218211_ s_at 231832 . _at

205259_ at 219543 . at 232176 . .at

205765_ at 219955_ at 232481 . _s_at

205927_ s_at 221841_ _s_at 235976 . at

208063_ s_at 221874 , .at 236894 . at

208937_ s_at 223969 s_at 237521 . _x_at

210107_ at 223970_ at 242601 . .at

213106_ at; and/oi

(ϋ) CLCAl CTSE ATP8B1

FCGBP C6orfl05 CACN A2D2

HMGCS2 CKB KLF4

RETNLB ATP8A1 CYP3A5P2

LlTDl MUC4 CAPN9

SLITRK6 UGTlAl NR3C2

VSIG2 SELENBPl PBLD

LOC253012 PTGER4 CA12

ST6GALNAC1 MLPH WDR51B

IDl KIAA 1324 FAM3D

CYP3A5 in said cell or cellular population wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer cell level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

16. A method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200600_at 204006_s_at 213428_s_at

200665_s_at 20405 l_s_at 213524_s_at

200832_s_at 204122_at 213869_x_at

200974_at 204320_at 213905_x_at

200986_at 204475_at 214247_s_at

201058_s_at 204620_s_at 215049_x_at

201069_at 205479_s_at 215076_s_at

201105_at 205547_s_at 215646_s_at

201141_at 205828_at 216442_x_at

201147_s_at 207173_x_at 217430_x_at

201150_s_at 20719 l_s_at 217762_s_at

201162_at 208747_s_at 217763_s_at

201163_s_at 208782_at 217764_s_at

201185_at 208788_at 218468_s_at

201261_x_at 208850_s_at 218469_at

201289_at 20885 l_s_at 218559_s_at

201426_s_at 209101_at 218638_s_at

201438_at 209156_s_at 219087_at

201616 s at 209218 at 221011 s at

201645_at 209395_at 221729_at

201667_at 209396_s_at 221730_at

201744_s_at 209596_at 221731_x_at

201792_at 209875_s_at 37892_at

201842_s_at 209955_s_at 223122_s_at

201852_x_at 210095_s_at 223235_s_at

201859_at 210495_x_at 224560_at

201893_x_at 210511_s_at 224694_at

202237_at 210764_s_at 224724_at

202238_s_at 210809_s_at 225664_at

202283_at 211161_s_at 22568 l_at

20229 l_s_at 211571_s_at 225710_at

202310_s_at 211719_x_at 225799_at

20231 1_s_at 211813_x_at 226237_at

202403_s_at 211896_s_at 226311_at

202404_s_at 211959_at 226694_at

202450_s_at 211964_at 226777_at

202620_s_at 211966_at 226930_at

202766_s_at 211980_at 227099_s_at

202859_x_at 211981_at 227140_at

202878_s_at 212077_at 227566_at

202917_s_at 212344_at 229218_at

202998_s_at 212353_at 229802_at

203083_at 212354_at 231579_s_at

203325_s_at 212464_s_at 231766_s_at

203382_s_at 212488_at 231879_at

203477_at 212489_at 232458_at

203570_at 212667_at 233555_s_at

203645_s_at 213125_at 234994_at

203878_s_at; and/or

(ϋ) COL1A2 LGALSl SRGN

CTHRCl ELO VL5 LBH

FNl MGP CTGF

POSTN MMP2 TNC

SPPl LOXL2 G0S2

MMPl MYL9 SQLE

SPARC DCN EFEMPl

LUM CALDl APOE

GREMl FBNl MSN

IL8 MMP3 IGFBP3

IGFBP5 IGFBP7 SERPINFl

SFRP2 FSTLl ISLR

SULFl COL4A2 HNT

ASPN VCAN COL5A1

COL6A3 SMOC2 OLFML2B

COL8A1 HTRAl KIAA1913

COL 12Al CYR61 PALM2-AKAP2

COL5A2 FAP SERPINGl

CDHI l VIM TYROBP

THBS2 TIMP2 ACTA2

COL15A1 SCD COL3A1

COLI lAl TIMP3 PLOD2

S100A8 AEBPl MMPI l

FNDCl GJAl CD 163

SFRP4 NNMT FCGR3B

INHBA COLlAl PLAU

COL6A2 SULF2 MAFB

ANTXRl COL6A1 LOC541471

GPNMB SPON2 LOC387763

BGN CTSK CHI3L1

TAGLN MXRA5 THYl

COL4A1 CIS LOXLl

RAB31 DKK3 CD93

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal adenoma cell level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

17. The method according to claim 15 wherein said one or more genes or transcripts are selected from:

(i) the gene or genes detected by Affymetrix probeset IDs: 210107_at; and/or

(H) CLCAl.

18. The method according to claim 15 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

203240_at, 219955_at, 24260 l_at, 204607_at, 23248 l_s_at, 227725_at, 223969_s_at, 228232_s_at; and/or

(ii) FCGBP, LlTDl, LOC253012, HMGCS2, SLITRK6, ST6GALNAC1, RETNLB, VSIG2.

19. The method according to claim 16 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

202404_s_at, 210809_s_at, 227140_at, 22568 l_at, 209875_s_at, 204475_at, 212464_s_at; and/or

(ii) COL 1A2, FNl, SPPl, CTHRCl, POSTN, MMPl.

20. The method according to claim 16 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200665_s_at, 218468_s_at, 211959_at, 201744_s_at, 202859_x_at; and/or

(ii) SPARC, GREMl, IGFBP5, LUM, IL8.

21. The method according to claim 16 wherein said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

223122_s_at 207173_x_at 21051 l_s_at

212353_at 203083_at 209156_s_at

219087_at 203477_at 224694_at

201438_at 37892_at 201141_at

226237_at 202917_s_at 213905_x_at

225664 at 226930 at 205547 s at

221730_at 20405 l_s_at; and/or

(ϋ) SFRP2 CDHI l INHBA

SULFl THBS2 COL6A2

ASPN COL15A1 ANTXRl

COL6A3 COLI lAl GPNMB

COL8A1 S100A8 BGN

COL 12Al FNDCl TAGLN

COL5A2 SFRP4.

22. The method according to claim 15 wherein said control level is the background level of neoplastic tissue and said one or more genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 235976_at 236894_at; and/or

(ii) SLITRK6 LlTDl.

23. The method according to claim 16 wherein said control level is the background level of neoplastic tissue and said one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

20231 l_s_at, 209396_s_at, 226237_at, 204320_at, 215646_s_at, 227140_at, 204475_at, 37892_at, 229802_at, 209395_at; and/or,

(ii) COLlAl, VCAN, CHI3L1, MMPl, COL8A1, COLI lAl.

24. The method according to any one of claims 1-23 wherein said cell is a colorectal cell.

25. The method according to any one of claims 1 to 24 wherein said biological sample is a faecal sample, enema wash, surgical resection, tissue biopsy or blood sample.

26. The method according to any one of claims 1 to 25 wherein said level of expression is mRNA expression or protein expression.

27. The method according to any one of claims 1-26 wherein said individual is a human.

28. A molecular array, which array comprises a plurality of:

(i) nucleic acid molecules comprising a nucleotide sequence corresponding to any one or more of the neoplastic marker genes listed in any one of claims 1-23 or a sequence exhibiting at least 80% identity thereto or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(ii) nucleic acid molecules comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iii) nucleic acid probes or oligonucleotides comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iv) probes capable of binding to any one or more of the proteins encoded by the nucleic acid molecules of (i) or a derivative, fragment or, homologue thereof

wherein the level of expression of said marker genes of (i) or proteins of (iv) is indicative of the neoplastic state of a cell or cellular subpopulation derived from the large intestine.

29. A diagnostic kit for assaying biological samples said kit comprising an agent for detecting one or more of the neoplastic markers listed in claims 1-23 and reagents useful for facilitating the detecting by said agent.

30. The method according to any one of claims 1-9 wherein said control level is a nonneoplastic level.

Description:

A METHOD OF DIAGNOSING NEOPLASMS

FIELD OF THE INVENTION The present invention relates generally to nucleic acid molecules, the RNA and protein expression profiles of which are indicative of the onset, predisposition to the onset and/or progression of a neoplasm. More particularly, the present invention is directed to nucleic acid molecules, the expression profiles of which are indicative of the onset and/or progression of a large intestine neoplasm, such as an adenoma or an adenocarcinoma. The expression profiles of the present invention are useful in a range of applications including, but not limited to, those relating to the diagnosis and/or monitoring of colorectal neoplasms, such as colorectal adenocarcinomas. Accordingly, in a related aspect the present invention is directed to a method of screening a subject for the onset, predisposition to the onset and/or progression of a neoplasm by screening for modulation in the expression profile of one or more nucleic acid molecule markers.

BACKGROUND OF THE INVENTION

Bibliographic details of the publications referred to by author in this specification are collected alphabetically at the end of the description.

The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Adenomas are benign tumours, or neoplasms, of epithelial origin which are derived from glandular tissue or exhibit clearly defined glandular structures. Some adenomas show recognisable tissue elements, such as fibrous tissue (fibroadenomas) and epithelial structure, while others, such as bronchial adenomas, produce active compounds that might give rise to clinical syndromes.

Adenomas may progress to become an invasive neoplasm and are then termed adenocarcinomas. Accordingly, adenocarcinomas are defined as malignant epithelial tumours arising from glandular structures, which are constituent parts of many organs of the body. The term adenocarcinoma is also applied to tumours showing a glandular growth pattern. These tumours may be sub-classified according to the substances that they produce, for example mucus secreting and serous adenocarcinomas, or to the microscopic arrangement of

their cells into patterns, for example papillary and follicular adenocarcinomas. These carcinomas may be solid or cystic (cystadenocarcinomas). Each organ may produce tumours showing a variety of histological types, for example the ovary may produce both mucinous and cystadenocarcinoma.

Adenomas in different organs behave differently. In general, the overall chance of carcinoma being present within an adenoma (i.e. a focus of cancer having developed within a benign lesion) is approximately 5%. However, this is related to size of an adenoma. For instance, in the large bowel (colon and rectum specifically) occurrence of a cancer within an adenoma is rare in adenomas of less than 1 centimetre. Such a development is estimated at 40 to 50% in adenomas which are greater than 4 centimetres and show certain histopathological change such as villous change, or high grade dysplasia. Adenomas with higher degrees of dysplasia have a higher incidence of carcinoma. In any given colorectal adenoma, the predictors of the presence of cancer now or the future occurrence of cancer in the organ include size (especially greater than 9mm) degree of change from tubular to villous morphology, presence of high grade dysplasia and the morphological change described as "serrated adenoma". In any given individual, the additional features of increasing age, familial occurrence of colorectal adenoma or cancer, male gender or multiplicity of adenomas, predict a future increased risk for cancer in the organ - so-called risk factors for cancer. Except for the presence of adenomas and its size, none of these is objectively defined and all those other than number and size are subject to observer error and to confusion as to precise definition of the feature in question. Because such factors can be difficult to assess and define, their value as predictors of current or future risk for cancer is imprecise.

Once a sporadic adenoma has developed, the chance of a new adenoma occurring is approximately 30% within 26 months.

Colorectal adenomas represent a class of adenomas which are exhibiting an increasing incidence, particularly in more affluent countries. The causes of adenoma, and of progression to adenocarcinoma, are still the subject of intensive research. To date it has been speculated that in addition to genetic predisposition, environmental factors (such as diet) play a role in the development of this condition. Most studies indicate that the relevant environmental factors relate to high dietary fat, low fibre, low vegetable intake, smoking, obesity, physical inactivity and high refined carbohydrates.

Colonic adenomas are localised areas of dysplastic epithelium which initially involve just one or several crypts and may not protrude from the surface, but with increased growth in size, usually resulting from an imbalance in proliferation and/or apoptosis, they may protrude. Adenomas can be classified in several ways. One is by their gross appearance and the major

descriptors include degrees of protrusion: flat sessile (i.e. protruding but without a distinct stalk) or pedunculated (i.e. having a stalk). Other gross descriptors include actual size in the largest dimension and actual number in the colon/rectum. While small adenomas (less than say 5 or 10 millimetres) exhibit a smooth tan surface, pedunculated and especially larger adenomas tend to have a cobblestone or lobulated red-brown surface. Larger sessile adenomas may exhibit a more delicate villous surface. Another set of descriptors include the histopathological classification; the prime descriptors of clinical value include degree of dysplasia (low or high), whether or not a focus of invasive cancer is present, degree of change from tubular gland formation to villous gland formation (hence classification is tubular, villous or tubulo villous), presence of admixed hyperplastic change and of so-called "serrated" adenomas and its subgroups. Adenomas can be situated at any site in the colon and/or rectum although they tend to be more common in the rectum and distal colon. All of these descriptors, with the exception of number and size, are relatively subjective and subject to interobserver disagreement.

The various descriptive features of adenomas are of value not just to ascertain the neoplastic status of any given adenomas when detected, but also to predict a person's future risk of developing colorectal adenomas or cancer. Those features of an adenoma or number of adenomas in an individual that point to an increased future risk for cancer or recurrence of new adenomas include: size of the largest adenoma (especially 10mm or larger), degree of villous change (especially at least 25% such change and particularly 100% such change), high grade dysplasia, number (3 or more of any size or histological status) or presence of serrated adenoma features. None except size or number is objective and all are relatively subjective and subject to interobserver disagreement. These predictors of risk for future neoplasia (hence "risk") are vital in practice because they are used to determine the rate and need for and frequency of future colonoscopic surveillance. More accurate risk classification might thus reduce workload of colonoscopy, make it more cost-effective and reduce the risk of complications from unnecessary procedures.

Adenomas are generally asymptomatic, therefore rendering difficult their diagnosis and treatment at a stage prior to when they might develop invasive characteristics and so became cancer. It is technically impossible to predict the presence or absence of carcinoma based on the gross appearance of adenomas, although larger adenomas are more likely to show a region of malignant change than are smaller adenomas. Sessile adenomas exhibit a higher incidence of malignancy than pedunculated adenomas of the same size. Some adenomas result in blood loss which might be observed or detectable in the stools; while sometimes visible by eye, it is often, when it occurs, microscopic or "occult". Larger adenomas tend to bleed more than smaller adenomas. However, since blood in the stool, whether overt or occult, can also be

indicative of non-adenomatous conditions, the accurate diagnosis of adenoma is rendered difficult without the application of highly invasive procedures such as colonoscopy combined with tissue acquisition by either removal (i.e. polypectomy) or biopsy and subsequent histopathological analysis.

Accordingly, there is an on-going need to elucidate the causes of adenoma and to develop more informative diagnostic protocols or aids to diagnosis that enable one to direct colonoscopy at people more likely to have adenomas. These adenomas may be high risk, advanced or neither of these. Furthermore, it can be difficult after colonoscopy to be certain that all adenomas have been removed, especially in a person who has had multiple adenomas. An accurate screening test may minimise the need to undertake an early second colonoscopy to ensure that the colon has been cleared of neoplasms. Accordingly, the identification of molecular markers for adenomas would provide means for understanding the cause of adenomas and cancer, improving diagnosis of adenomas including development of useful screening tests, elucidating the histological stage of an adenoma, characterising a patient's future risk for colorectal neoplasia on the basis of the molecular state of an adenoma and facilitating treatment of adenomas.

To date, research has focused on the identification of gene mutations which lead to the development of colorectal neoplasms. In work leading up to the present invention, however, it has been determined that changes in expression profiles of genes which are also expressed in healthy individuals are indicative of the development of neoplasms of the large intestine, such as adenomas and adenocarcinomas. It has been further determined that in relation to neoplasms of the large intestine, diagnosis can be made based on screening for one or more of a panel of these differentially expressed genes. In a related aspect, it has still further been determined that to the extent that neoplastic tissue has been identified either by the method of the invention or by some other method, the present invention provides still further means of characterising that tissue as an adenoma or a cancer. In yet another aspect, it has been determined that a proportion of these genes are characterised by gene expression which occurs in the context of a neoplastic state but not in the context of a non-neoplastic state, thereby facilitating the development of qualitative analyses which do not require a relative analysis to be performed against a non-neoplastic or normal control reference level. Accordingly, the inventors have identified a panel of genes which facilitate the diagnosis of adenocarcinoma and adenoma development and/or the monitoring of conditions characterised by the development of these types of neoplasms.

SUMMARY OF THE INVENTION

Throughout this specification and the claims which follow, unless the context requires

otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

As used herein, the term "derived from" shall be taken to indicate that a particular integer or group of integers has originated from the species specified, but has not necessarily been obtained directly from the specified source. Further, as used herein the singular forms of "a", "and" and "the" include plural referents unless the context clearly dictates otherwise.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

The subject specification contains amino acid and nucleotide sequence information prepared using the programme Patentln Version 3.4, presented herein after the bibliography. Each amino acid and nucleotide sequence is identified in the sequence listing by the numeric indicator <210> followed by the sequence identifier (eg. <210>l, <210>2, etc). The length, type of sequence (amino acid, DNA, etc.) and source organism for each sequence is indicated by information provided in the numeric indicator fields <211>m <212> and <213>, respectively. Amino acid and nucleotide sequences referred to in the specification are identified by the indicator SEQ ID NO: followed by the sequence identifier (eg. SEQ ID

NO:1, SEQ ID NO: 2, etc). The sequence identifier referred to in the specification correlates to the information provided in numeric indicator field <400> in the sequence listing, which is followed by the sequence identifier (eg. <400>l, <400>2, etc). That is SEQ ID NO: 1 as detailed in the specification correlates to the sequence indicated as <400>l in the sequence listing.

One aspect of the present invention is directed to a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

201328_at 221577_x_at 205828_at

201341_at 221922_at 205886_at

201416_at 60474_at 205890_s_at

201417_at 222696_at 205910_s_at

201468_s_at 223447_at 205941_s_at

201506 at 223970 at 206224 at

201563_at 225541_at 206976_s_at

201656_at 225835_at 207173_x_at

201925_s_at 226360_at 207457_s_at

201926_s_at 227174_at 208079_s_at

202286_s_at 227475_at 208712_at

202718_at 228303_at 209218_at

20283 l_at 228653_at 209309_at

202833_s_at 228754_at 209752_at

202935_s_at 228915_at 209773_s_at

202936_s_at 229215_at 209774_x_at

203124_s_at 231832_at 209792_s_at

203256_at 231941_s_at 209875_s_at

203313_s_at 232176_at 209955_s_at

203510_at 232252_at 210052_s_at

203860_at 23248 l_s_at 21051 l_s_at

203895_at 23433 l_s_at 210559_s_at

203896_s_at 235210_s_at 210766_s_at

20396 l_at 235976_at 211506_s_at

203962_s_at 236894_at 212281_s_at

204259_at 238017_at 212344_at

20435 l_at 23802 l_s_at 212353_at

20440 l_at 238984_at 212354_at

204404_at 24103 l_at 213905_x_at

204855_at 200660_at 214022_s_at

204885_s_at 200832_s_at 214974_x_at

205174_s_at 200903_s_at 215091_s_at

205366_s_at 201014_s_at 217430_x_at

205470_s_at 201112_s_at 217996_at

205513_at 201195_s_at 218507_at

205765_at 201261_x_at 218963_s_at

205825_at 201292_at 218984_at

205927_s_at 201338_x_at 219787_s_at

205983_at 201479_at 21991 l_s_at

206239_s_at 201577_at 221729_at

206286_s_at 201601_x_at 221730_at

207158_at 201666_at 221731_x_at

207850_at 202310_s_at 221923_s_at

209369 at 202311 s at 37892 at

210445_at 202404_ _s_at 222449 . at

210519_s_at 202431 . _s_at 222450 . at

211429_s_at 202504_ at 222549 . at

212063_at 202779_ s at 222608 . _s_at

212070_at 202859 , x at 223062 . _s_at

212190_at 202954_ .at 224428 . _s_at

212531_at 202998_ _s_at 224646 . _x_at

212942_s_at 203083_ at 224915 . x_at

213880_at 203213_ at 225295 . at

213975_s_at 203878_ _s_at 225520 . at

214235_at 204051 . _s_at 225664 . at

21465 l_s_at 204127_ at 225681 . _at

217523_at 204170 . _s_at 225767 . at

217867_x_at 204320_ at 225799 . _at

218086_at 204470 . _at 225806 . at

21821 l_s_at 204475 , .at 226227 . x at

218704_at 204580_ at 226237 . .at

218796_at 204620 . _s_at 226311 . at

218872_at 204702 _s_at 226777 . at

219630_at 205361 . _s_at 226835 . _s_at

219682_s_at 205476 . at 227140 . .at

219727_at 205479 . _s_at 229802 . at

219955_at 205713 . _s_at 231766 . _s_at

219956_at 205815 . at 232151 . at; and/or

200665_s_at

(ϋ) FOXQl RNF43 CDCA7 LOXL2

MMPl CCL20 TDGFl AZGPl

TCNl CTSE MTHFDlL MYC

MMP7 MSLN ANLN COMP

WDR72 TIMPl H19 AURKA

INHBA PCSKl FAP PAICS

COLI lAl CSTl DACHl PUS7

GDF 15 BGN VCAN ZNRF3

CTHRCl AXIN2 SQLE CCNDl

COLlAl MET REG3A CSElL

LGR5 SOX9 TESC PFDN4

DUSP27 TMEPAI UBE2C C20orf42

SERPINB5 CDHI l TMEM97 SLCl 1A2

ASCL2 MMPI l TRIM29 NFE2L3

SULFl CXCLl KLKI l CEL

SLC6A6 QPCT LY6G6D NLFl

TACSTD2 PDZKlIPl SLC7A5 NPDCl

NEBL CD55 FABP6 ENCl

PCCA ECT2 SLITRK6 SERPINE2

FL J37644 COLl 2Al MLPH UBE2S

KRT23 LlTDl HOXA9 TOP2A

CXCL3 WDR51B TBX3 CDC2

MMP3 HOXB6 BACE2 RFC3

SFRP4 FAM84A GPX2 LILRBl

UBD COL5A2 TPX2 NPMl

SCD MMP 12 KCNN4 RDHE2

DPEPl SORD LOC541471 PLAU

LCN2 PSATl CYP3A5 HSPHl

PLCB4 CXCL5 ANXA3 GTF3A

SPINKl IGFBP2 CYP3A5P2 KLKlO

DUOX2 TGIFl C14orf94 GPSM2

SOX4 CXCL2 RP5-875H10.1 NMEl

THBS2 SLCO4A1 RPL22L1 MUC20

CLDNl CKS2 APOBECl RPESP

REGlB PHLDAl HIG2 RRM2

CDH3 SERPINAl COLlOAl DKCl

SPARC COL 1A2 AHCY CD44

IL8

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

In another aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 225681_at; 227140_at; and/or

(ii) CTHRCl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

In yet another aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

227475_at

204475_at

202859_x_at

202404_s_at; and/or

(ii) FOXQl, MMPl, IL8, COL1A2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

In still another aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 205513_at, 204259_at, 227174_at, 21051 l_s_at, 37892_at; and/or

(ii) TCN 1 , MMP7, WDR72, INHBA, COLI lAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Preferably, said control level is a non-neoplastic level.

In still yet another aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

221577_x_at 212942_s_at 202310_s_at

213880_at 232252_at 204855_at

229215_at 212354_at 228754_at

202286_s_at 203962_s_at 203860_at

202935_s_at 218963_s_at 207850_at

205828_at 20405 l_s_at 205890_s_at

200832_s_at 205983_at 21253 l_at; and/or

(ϋ) LGR5 GDF 15 COLlAl

ASCL2 DUSP27 SERPINB5

TACSTD2 SULFl SLC6A6

FLJ37644 NEBL PCCA

MMP3 KRT23 CXCL3

SCD SFRP4 UBD

DPEPl LCN2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

In a further aspect the present invention is directed to a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

201328_at 205825_at 219956_at

201468_s_at 205927_s_at 221922_at

201656_at 206239_s_at 223447_at

201925_s_at 207158_at 223970_at

201926_s_at 210445_at 225835_at

202718_at 210519_s_at 226360_at

20283 l_at 211429_s_at 228303_at

202833_s_at 212063_at 228653_at

203124_s_at 213975_s_at 228915_at

203313_s_at 214235_at 231832_at

203860 at 214651 s at 231941 s at

203895_at 217523_ at 232176_at

203896_s_at 217867_ x_at 23248 l_s_at

20440 l_at 218086_ at 23433 l_s_at

204885_s_at 218211_ s_at 235210_s_at

205174_s_at 218796_ at 235976_at

205366_s_at 219630_ at 236894_at

205470_s_at 219682_ s_at 238017_at

205513_at 219727_ at 238984_at

205765_at 219955_ at; 24103 l_at; and/or

(ϋ) APOBECl HOXB6 QPCT

BACE2 IGFBP2 RDHE2

C20orf42 ITGA6 REG4

CD44 KCNN4 RETNLB

CD55 KLKI l RP5-875H10.1

CTSE LlTDl RPESP

CYP3A5 LILRBl SERPINAl

CYP3A5P2 MLPH SLCl 1A2

DACHl MSLN SLCl 2 A2

DUOX2 MUC20 SLITRK6

ETS2 NLFl SPINKl

FABP6 NPDCl TBX3

FAM84A NQOl TCNl

GALNT6 PCCA TGIFl

GPSM2 PCSKl WDR51B

GPX2 PDZKlIPl ZNRF3

HOXA9 PLCB4

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another further aspect of the present invention there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200660_at 205476_ at 217430_x_at

200665_s_at 205479_ s_at 217996_at

200832_s_at 205713_ s_at 218507_at

200903_s_at 205815_ at 218963_s_at

201014_s_at 205828_ at 218984_at

201112_s_at 205886_ .at 219787_s_at

201195_s_at 205890 , s at 21991 l_s_at

201261_x_at 205910_ s at 221729_at

201292_at 205941_ s at 221730_at

201338_x_at 206224_ at 221731_x_at

201479_at 206976 . _s_at 221923_s_at

201577_at 207173 . x_at 37892_at

201601_x_at 207457 . _s_at 222449_at

201666_at 208079 . _s_at 222450_at

202310_s_at 208712_ at 222549_at

202311_s_at 209218 . .at 222608_s_at

202403_s_at 209309 . at 223062_s_at

202404_s_at 209752 . at 224428_s_at

20243 l_s_at 209773. _s_at 224646_x_at

202504_at 209774 . x at 224915_x_at

202779_s_at 209792 . s at 225295_at

202859_x_at 209875 . _s_at 225520_at

202954_at 209955 . _s_at 225664_at

202998_s_at 210052 . _s_at 22568 l_at

203083_at 210511 . _s_at 225767_at

203213_at 210559 . _s_at 225799_at

203878_s_at 210766 . _s_at 225806_at

20405 l_s_at 211506 . _s_at 226227_x_at

204127_at 212281 . s at 226237_at

204170_s_at 212344 . at 226311_at

204320_at 212353 . _at 226777_at

204470_at 212354 . _at 226835_s_at

204475_at 213905 . x at 227140_at

204580_at 214022 . _s_at 229802_at

204620_s_at 214974 x at 231766_s_at

204702_s_at 215091 s at; 23215 l_at; and/or

205361 s at

(») AHCY DKCl PSATl

ANLN ECT2 PUS7

AURKA FAP REGlA

AZGPl GTF3A REGlB

BGN H19 REG3A

C14orf94 HIG2 RFC3

C20orfl99 HSPHl RRM2

CCL20 IFITMl SlOOAI l

CCNDl IL8 SCD

CDC2 INHBA SFRP4

CDCA7 KLKlO SLC39A10

CDHI l KRT23 SLC7A5

CEL LOC541471 SLCO4A1

CKS2 LOXL2 SPARC

CLDNl LY6G6D SPPl

COLlOAl MMPl SQLE

COLI lAl MMPI l SULFl

COLl 2Al MMP 12 THBS2

COLlAl MMP3 TIMPl

COL1A2 MTHFDlL TMEM97

COL5A2 MYC TMEPAI

COL8A1 NFE2L3 TOP2A

COMP NMEl TPX2

CSElL NPMl TRIM29

CSTl PAICS UBD

CTHRCl PFDN4 UBE2C

CXCLl PHLDAl UBE2S

CXCL2 PLAU VCAN

CXCL5

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a cancer cell or a cell predisposed to the onset of a cancerous state.

In yet another further aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

202286_s_at 235976_at 209309_at

204259_at 236894_at 211506_s_at

204885_s_at 214974_x_at

205174_s_at 238984_at 219787_s_at

205825_at 24103 l_at 37892_at

207850_at 202311_s_at 222608_s_at

213880_at 204320_at 223062_s_at

217523_at 204475_at 225806_at

227174_at 204702_s_at 226237_at

228915_at 205910_s_at 227140_at

232252_at 206224_at 229802_at; and/or

(») MMPl PCSKl ANLN

MMP7 CSTl DACHl

LGR5 QPCT COLI lAl

WDR72 ECT2 C14orf94

COLI lAl SLITRK6 AZGPl

COLlAl LlTDl REG4

DUSP27 KIAAl 199 NFE2L3

NLFl PSATl CEL

IL8 CXCL5 CD44

TACSTD2 CXCL3 COL8A1

MSLN

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of a neoplastic cell or a cell predisposed to the onset of a neoplastic state.

Yet another aspect of the present invention provides a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 204885_s_at 217523_at 236894_at

205174_s_at ,228915_at 238984_at

205825 at 235976 at 24103 l_at; and/or

(ϋ) CD44 MSLN QPCT

DACHl NLFl REG4

LlTDl PCSKl SLITRK6

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In yet still another aspect there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene or genes detected by Affymetrix probeset IDs:

202311_s_at 209309_at 223062_ s_at

204320_at 211506_s_at 225806_ at

204475_at 214974_x_at 226237_ at

204702_s_at 219787_s_at 227140_ at

205910_s_at 37892_at 229802_ at; and/or

206224_at 222608_s_at

(ϋ) ANLN COLlAl IL8

AZGPl COL8A1 MMPl

C14orf94 CSTl NFE2L3

CEL CXCL5 PSATl

COLI lAl ECT2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of a cancer cell or a cell predisposed to the onset of a cancerous state.

In still yet another aspect of the present invention, there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 200884 at 214234 s at 226248 s at

203240_at 214235_at 226302_at

203963_at 214433_s_at 227676_at

204508_s_at 215125_s_at 227719_at

204607_at 215867_x_at 227725_at

204811_s_at 217109_at 228232_s_at

204895_x_at 217110_s_at 229070_at

204897_at 21821 l_s_at 231832_at

205259_at 219543_at 232176_at

205765_at 219955_at 23248 l_s_at

205927_s_at 221841_s_at 235976_at

208063_s_at 221874_at 236894_at

208937_s_at 223969_s_at 23752 l_x_at

210107_at 223970_at 242601 at

213106_at; and/or

(ϋ) CLCAl CTSE ATP8B1

FCGBP C6orfl05 CACNA2D2

HMGCS2 CKB KLF4

RETNLB ATP8A1 CYP3A5P2

LlTDl MUC4 CAPN9

SLITRK6 UGTlAl NR3C2

VSIG2 SELENBPl PBLD

LOC253012 PTGER4 CA12

ST6GALNAC1 MLPH WDR51B

IDl KIAA 1324 FAM3D

CYP3A5

in said cell or cellular population wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer cell level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another aspect there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200600 at 204006 s at 213428 s at

200665_s_at 20405 l_s_at 213524_s_at

200832_s_at 204122_at 213869_x_at

200974_at 204320_at 213905_x_at

200986_at 204475_at 214247_s_at

201058_s_at 204620_s_at 215049_x_at

201069_at 205479_s_at 215076_s_at

201105_at 205547_s_at 215646_s_at

201141_at 205828_at 216442_x_at

201147_s_at 207173_x_at 217430_x_at

201150_s_at 207191_s_at 217762_s_at

201162_at 208747_s_at 217763_s_at

201163_s_at 208782_at 217764_s_at

201185_at 208788_at 218468_s_at

201261_x_at 208850_s_at 218469_at

201289_at 20885 l_s_at 218559_s_at

201426_s_at 209101_at 218638_s_at

201438_at 209156_s_at 219087_at

201616_s_at 209218_at 22101 l_s_at

201645_at 209395_at 221729_at

201667_at 209396_s_at 221730_at

201744_s_at 209596_at 221731_x_at

201792_at 209875_s_at 37892_at

201842_s_at 209955_s_at 223122_s_at

201852_x_at 210095_s_at 223235_s_at

201859_at 210495_x_at 224560_at

201893_x_at 21051 l_s_at 224694_at

202237_at 210764_s_at 224724_at

202238_s_at 210809_s_at 225664_at

202283_at 211161_s_at 22568 l_at

20229 l_s_at 211571_s_at 225710_at

202310_s_at 211719_x_at 225799_at

202311_s_at 211813_x_at 226237_at

202403_s_at 211896_s_at 22631 1_at

202404_s_at 211959_at 226694_at

202450_s_at 211964_at 226777_at

202620_s_at 211966_at 226930_at

202766_s_at 211980_at 227099_s_at

202859 x at 211981 at 227140 at

202878_s_at 212077_at 227566_at

202917_s_at 212344_at 229218_at

202998_s_at 212353_at 229802_at

203083_at 212354_at 231579_s_at

203325_s_at 212464_s_at 231766_s_at

203382_s_at 212488_at 231879_at

203477_at 212489_at 232458_at

203570_at 212667_at 233555_s_at

203645_s_at 213125_at 234994_at

203878_s_at; and/or

(ϋ) COL 1A2 LGALSl SRGN

CTHRCl ELO VL5 LBH

FNl MGP CTGF

POSTN MMP2 TNC

SPPl L0XL2 G0S2

MMPl MYL9 SQLE

SPARC DCN EFEMPl

LUM CALDl APOE

GREMl FBNl MSN

IL8 MMP3 IGFBP3

IGFBP5 IGFBP7 SERPINFl

SFRP2 FSTLl ISLR

SULFl COL4A2 HNT

ASPN VCAN COL5A1

COL6A3 SMOC2 OLFML2B

COL8A1 HTRAl KIAA1913

COL 12Al CYR61 PALM2-AKAP2

COL5A2 FAP SERPINGl

CDHI l VIM TYROBP

THBS2 TIMP2 ACTA2

COL15A1 SCD COL3A1

COLI lAl TIMP3 PLOD2

S100A8 AEBPl MMPI l

FNDCl GJAl CD163

SFRP4 NNMT FCGR3B

INHBA COLlAl PLAU

COL6A2 SULF2 MAFB

ANTXRl COL6A1 LOC541471

GPNMB SPON2 LOC387763

BGN CTSK CHI3L1

TAGLN MXRA5 THYl

COL4A1 CIS LOXLl

RAB31 DKK3 CD93

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal adenoma cell level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

In a further aspect there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene or genes detected by Affymetrix probeset IDs: 210107_at; and/or

(ii) CLCAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer control level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another aspect there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 203240_at 219955_at 24260 l at

204607_at 23248 l_s_at 227725_at

223969_s_at 228232_s_at; and/or

(») FCGBP LlTDl LOC253012

HMGCS2 SLITRK6 ST6GALNAC1

RETNLB VSIG2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer control level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

A further aspect of the present invention is directed to a method of characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

202404_s_at, 210809_s_at, 227140_at, 22568 l_at, 209875_s_at, 204475_at, 212464_s_at; and/or,

(ii) COL1A2, FNl, SPPl, CTHRCl, POSTN, MMPl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

In yet another further aspect the present invention is directed to a method of characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200665_s_at, 218468_s_at, 211959_at, 201744_s_at, 202859_x_at; and/or,

(ii) SPARC, GREMl, IGFBP5, LUM, IL8,

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

In still yet another further aspect the present invention is directed to a method of characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

223122_s_at 207173_x_at 210511_s_at

212353_at 203083_at 209156_s_at

219087_at 203477_at 224694_at

201438_at 37892_at 201141_at

226237_at 202917_s_at 213905_x_at

225664_at 226930_at 205547_s_at

221730_at 20405 l_s_at; and/or

(«) SFRP2 CDHI l INHBA

SULFl THBS2 COL6A2

ASPN COLl 5Al ANTXRl

COL6A3 COLI lAl GPNMB

COL8A1 S100A8 BGN

COL 12Al FNDCl TAGLN

COL5A2 SFRP4

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

Yet another aspect of the present invention provides a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 235976_at 236894_at; and/or

(ii) SLITRK6 LlTDl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to neoplastic tissue background levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In still another aspect the present invention provides a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

20231 l_s_at, 209396_s_at, 226237_at, 204320_at, 215646_s_at, 227140_at, 204475_at, 37892_at, 229802_at, 209395_at; and/or

(H) COLlAl, VCAN, CHI3L1, MMPl, COL8A1, COLI lAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to neoplastic tissue background neoplastic cell levels is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

A related aspect of the present invention provides a molecular array, which array comprises a plurality of:

(i) nucleic acid molecules comprising a nucleotide sequence corresponding to any one or more of the neoplastic marker genes hereinbefore described or a sequence exhibiting at least 80% identity thereto or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(ii) nucleic acid molecules comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iii) nucleic acid probes or oligonucleotides comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iv) probes capable of binding to any one or more of the proteins encoded by the nucleic acid molecules of (i) or a derivative, fragment or, homologue thereof

wherein the level of expression of said marker genes of (i) or proteins of (iv) is indicative of the neoplastic state of a cell or cellular subpopulation derived from the large intestine.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 is a graphical representation of nuclear factor (erythroid-derived-2)-like 2. Blk=normal, grn=inflamed, red=adenoma,blue=cancer

Figure 2 is a graphical representation of SLIT and NTRK-like family, member 6 (left) and LINEl type transposase domain containing 1 (right).

Figure 3 is a graphical representation of collagen type XI, alpha 1.

Figure 4 is an image of the immunohistochemical staining for MSLN in a normal tissue shows mild staining in the cytoplasm of the colonocytes.

Figure 5 is an image of the immunohistochemical staining for MSLN in a representative cancer tissue shows moderate staining in the cytoplasm of the colonic epithelia.

Figure 6 is an image of the immunohistochemical staining of MSLN in a moderately differentiated adenoma shows strong staining in the multilayered colonic epithelium.

Figure 7 is a graphical representation depicting Probeset 205828_at Expression profile across 68 clinical specimens comprising 30 non-disease controls (black), 19 adenomas (red) and 19 adenocarcinoma (green).

Figure 8 is a graphical representation of the measurement of MMP3 expression in protein extract from 27 clinical stool specimens. Protein extraction was performed on 27 clinical stool specimens comprising 6 non-disease controls (circle), 10 adenoma (triangle) and 11 adenocarcinoma (cross) emulsified using a PBS pH 7.4 solution containing 0.05% Tween-20 and Ix Protease Inhibitor Cocktail (Roche). Proteins were further solubilised by 30 minutes incubation in an ultrasonic water bath. Solubilised proteins were isolated by centrifugation and endogenous levels of MMP3 in the resulting protein extracts were using measured using a commercially available Luminex bead-based suspension immunoassay as recommended by manufacturer (R&D Systems).

Figure 9 is a graphical representation of Affymetrix probeset ID 205828 at which is known to hybrising to transcripts of the MMP3 gene depicting a gene expression profile in 68 clinical specimens comprising 30 non-disease controls (black), 19 adenomas (red) and 19 cancers (green).

Figure 10 is a schematic representation of predicted RNA variants derived from hCG_1815491. cDNA clones derived from map region 8579310 to 8562303 on human chromosome 16 were used to locate exon sequences. Arrows: Oligo nucleotide primer sets were designed to allow measurement of individual RNA variants by PCR. Primers covering splice junctions are shown as spanning intron sequences which is not included in

the actual oligonucleotide primer sequence.

DETAILED DESCRIPTION OF THE INVENTION

The present invention is predicated, in part, on the elucidation of gene expression profiles which characterise large intestine cellular populations in terms of their neoplastic state and, more particularly, whether they are malignant or pre-malignant. This finding has now facilitated the development of routine means of screening for the onset or predisposition to the onset of a large intestine neoplasm or characterising cellular populations derived from the large intestine based on screening for upregulation of the expression of these molecules, relative to control expression patterns and levels. To this end, in addition to assessing expression levels of the subject genes relative to normal or non-neoplastic levels, it has been determined that a proportion of these genes are expressed only in the diseased state, thereby facilitating the development of a simple qualitative test based on requiring assessment only relative to test background levels.

In accordance with the present invention, it has been determined that the genes detailed above are modulated, in terms of differential changes to their levels of expression, depending on whether the cell expressing that gene is neoplastic or not. It should be understood that reference to a gene "expression product" or "expression of a gene" is a reference to either a transcription product (such as primary RNA or mRNA) or a translation product such as protein. These genes and their expression products, whether they be RNA transcripts or encoded proteins, are collectively referred to as "neoplastic markers".

Accordingly, one aspect of the present invention is directed to a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 201328_at 221577_x_at 205828_at

201341_at 221922_at 205886_at

201416_at 60474_at 205890_s_at

201417_at 222696_at 205910_s_at

201468 s at 223447 at 205941 s at

201506_at 223970_at 206224_at

201563_at 225541_at 206976_s_at

201656_at 225835_at 207173_x_at

201925_s_at 226360_at 207457_s_at

201926_s_at 227174_at 208079_s_at

202286_s_at 227475_at 208712_at

202718_at 228303_at 209218_at

20283 l_at 228653_at 209309_at

202833_s_at 228754_at 209752_at

202935_s_at 228915_at 209773_s_at

202936_s_at 229215_at 209774_x_at

203124_s_at 231832_at 209792_s_at

203256_at 231941_s_at 209875_s_at

203313_s_at 232176_at . 209955_s_at

2035 lO at 232252_at 210052_s_at

203860_at 23248 l_s_at 21051 l_s_at

203895_at 23433 l_s_at 210559_s_at

203896_s_at 235210_s_at 210766_s_at

203961 at 235976_at 211506_s_at

203962_s_at 236894_at 212281_s_at

204259_at 238017_at 212344_at

20435 l_at 238021_s_at 212353_at

20440 l_at 238984_at 212354_at

204404_at 24103 l_at 213905_x_at

204855_at 200660_at 214022_s_at

204885_s_at 200832_s_at 214974_x_at

205174_s_at 200903_s_at 215091_s_at

205366_s_at 201014_s_at 217430_x_at

205470_s_at 201112_s_at 217996_at

205513_at 201195_s_at 218507_at

205765_at 201261_x_at 218963_s_at

205825_at 201292_at 218984_at

205927_s_at 201338_x_at 219787_s_at

205983_at 201479_at 21991 l_s_at

206239_s_at 201577_at 221729_at

206286_s_at 201601_x_at 221730_at

207158_at 201666_at 221731_x_at

207850 at 202310 s at 221923 s at

209369_at 202311_s_at 37892_at

210445_at 202404_s_at 222449_at

210519_s_at 20243 l_s_at 222450_at

211429_s_at 202504_at 222549_at

212063_at 202779_s_at 222608_s_at

212070_at 202859_x_at 223062_s_at

212190_at 202954_at 224428_s_at

21253 l_at 202998_s_at 224646_x_at

212942_s_at 203083_at 224915_x_at

213880_at 203213_at 225295_at

213975_s_at 203878_s_at 225520_at

214235_at 20405 l_s_at 225664_at

21465 l_s_at 204127_at 22568 l_at

217523_at 204170_s_at 225767_at

217867_x_at 204320_at 225799_at

218086_at 204470_at 225806_at

218211_s_at 204475_at 226227_x_at

218704_at 204580_at 226237_at

218796_at 204620_s_at 22631 l_at

218872_at 204702_s_at 226777_at

219630_at 20536 l_s_at 226835_s_at

219682_s_at 205476_at 227140_at

219727_at 205479_s_at 229802_at

219955_at 205713_s_at 231766_s_at

219956_at 205815_at 23215 l_at; and/or

200665_s_at

(H) CTHRCl CDCA7 LOXL2

FOXQl RNF43 TDGFl AZGPl

MMPl CCL20 MTHFDlL MYC

TCNl CTSE ANLN COMP

MMP7 MSLN H19 AURKA

WDR72 TIMPl FAP PAICS

INHBA PCSKl DACHl PUS7

COLI lAl CSTl VCAN ZNRF3

GDF 15 BGN SQLE CCNDl

COLlAl AXIN2 REG3A CSElL

LGR5 MET TESC PFDN4

DUSP27 SOX9 UBE2C C20orf42

SERPINB5 TMEPAI TMEM97 SLCl 1A2

ASCL2 CDHI l TRIM29 NFE2L3

SULFl MMPI l KLKI l CEL

SLC6A6 CXCLl LY6G6D NLFl

TACSTD2 QPCT SLC7A5 NPDCl

NEBL PDZKlIPl FABP6 ENCl

PCCA CD55 SLITRK6 SERPINE2

FLJ37644 ECT2 MLPH UBE2S

KRT23 COL 12Al HOXA9 TOP2A

CXCL3 LlTDl TBX3 CDC2

MMP3 WDR51B BACE2 RFC3

SFRP4 HOXB6 GPX2 LILRBl

UBD FAM84A TPX2 NPMl

SCD COL5A2 KCNN4 RDHE2

DPEPl MMP 12 LOC541471 PLAU

LCN2 SORD CYP3A5 HSPHl

PLCB4 PSATl ANXA3 GTF3A

SPINKl CXCL5 CYP3A5P2 KLKlO

DUOX2 IGFBP2 C14orf94 GPSM2

SOX4 TGIFl RP5-875H10.1 NMEl

THBS2 CXCL2 RPL22L1 MUC20

CLDNl SLCO4A1 APOBECl RPESP

REGlB CKS2 HIG2 RRM2

CDH3 PHLDAl COLlOAl DKCl

SPARC SERPINAl AHCY CD44

COL 1A2 IL8

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Reference to "large intestine" should be understood as a reference to a cell derived from one of the six anatomical regions of the large intestine, which regions commence after the terminal region of the ileum, these being:

(i) the cecum;

(ii) the ascending colon;

(iii) the transverse colon;

(iv) the descending colon; (v) the sigmoid colon; and (vi) the rectum.

Reference to "neoplasm" should be understood as a reference to a lesion, tumour or other encapsulated or unencapsulated mass or other form of growth which comprises neoplastic cells. A "neoplastic cell" should be understood as a reference to a cell exhibiting abnormal growth. The term "growth" should be understood in its broadest sense and includes reference to proliferation. In this regard, an example of abnormal cell growth is the uncontrolled proliferation of a cell. Another example is failed apoptosis in a cell, thus prolonging its usual life span. The neoplastic cell may be a benign cell or a malignant cell. In a preferred embodiment, the subject neoplasm is an adenoma or an adenocarcinoma. Without limiting the present invention to any one theory or mode of action, an adenoma is generally a benign tumour of epithelial origin which is either derived from epithelial tissue or exhibits clearly defined epithelial structures. These structures may take on a glandular appearance. It can comprise a malignant cell population within the adenoma, such as occurs with the progression of a benign adenoma to a malignant adenocarcinoma.

Preferably, said neoplastic cell is an adenoma or adenocarcinoma and even more preferably a colorectal adenoma or adenocarcinoma.

Each of the genes and transcripts detailed in sub-paragraphs (i) and (ii), above, would be well known to the person of skill in the art, as would their encoded proteins. The identification of the expression products of these genes and transcripts as markers of neoplasia occurred by virtue of differential expression analysis using Affymetrix HGU133Aor HGUl 33B gene chips. To this end, each gene chip is characterised by approximately 45,000 probe sets which detect the RNA transcribed from the genome. On average, approximately 11 probe pairs detect overlapping or consecutive regions of the RNA transcript. In general, the genes from which the RNA transcripts described herein are identified by the Affymetrix probesets are well known and characterised genes. However, to the extent that some of the probesets detect RNA transcripts which are not yet defined, these transcripts are indicated as "the gene, genes or transcripts detected by Affymetrix probe x". In some cases a number of genes and/or transcripts may be detectable by a single probeset. It should be understood, however, that this is not intended as a limitation as to how the expression level of the subject gene or transcript can be detected. In the first instance, it would be understood that the subject gene transcript is also detectable by other probesets which would be present on the Affymetrix gene chip. The reference to a single probesets is merely included as an identifier of the gene transcript of interest. In terms of actually screening for the transcript, however, one may utilise a probe or probeset directed to any region of the transcript and not just to the 3' terminal 600bp

transcript region to which the Affymetrix probesets are often directed.

Reference to each of the genes and transcripts detailed above and their transcribed and translated expression products should therefore be understood as a reference to all forms of these molecules and to fragments or variants thereof. As would be appreciated by the person of skill in the art, some genes are known to exhibit allelic variation between individuals. Accordingly, the present invention should be understood to extend to such variants which, in terms of the present diagnostic applications, achieve the same outcome despite the fact that minor genetic variants between the actual nucleic acid sequences may exist between individuals or that within one individual there may exist two or more splice variants of one subject gene. The present invention should therefore be understood to extend to all forms of RNA (eg mRNA, primary RNA transcript, miRNA, etc), cDNA and peptide isoforms which arise from alternative splicing or any other mutation, polymorphic or allelic variation. It should also be understood to include reference to any subunit polypeptides such as precursor forms which may be generated, whether existing as a monomer, multimer, fusion protein or other complex.

For example, in one embodiment of the invention, the subject gene is CDH3. Analysis of the AceView Database reveals that there exist 12 CDH3 alternative mRNA transcripts. Nine are generated by alternative splicing while three are unspliced forms. In terms of the genes encompassed by the present invention, means for determining the existence of such variants and characterising same, are described in Example 6. To the extent that the genes of the present invention are described by reference to an Affymetrix probeset, Table 9 provides details of the nucleic acid sequence to which each probeset is directed. Based on this information, the skilled person could, as a matter of routine procedure, identify the gene in respect of which that sequence forms part. A typical protocol for doing this is also outlined in Example 6.

It should be understood that the "individual" who is the subject of testing may be any human or non-human mammal. Examples of non-human mammals includes primates, livestock animals (e.g. horses, cattle, sheep, pigs, donkeys), laboratory test animals (e.g. mice, rats, rabbits, guinea pigs), companion animals (e.g. dogs, cats) and captive wild animals (e.g. deer, foxes). Preferably the mammal is a human.

The method of the present invention is predicated on the comparison of the level of the neoplastic markers of a biological sample with the control levels of these markers. The "control level" may be either a "normal level", which is the level of marker expressed by a corresponding large intestine cell or cellular population which is not neoplastic, or the background level which is detectable in a negative control sample.

The normal (or "non-neoplastic") level may be determined using tissues derived from the same individual who is the subject of testing. However, it would be appreciated that this may be quite invasive for the individual concerned and it is therefore likely to be more convenient to analyse the test results relative to a standard result which reflects individual or collective results obtained from individuals other than the patient in issue. This latter form of analysis is in fact the preferred method of analysis since it enables the design of kits which require the collection and analysis of a single biological sample, being a test sample of interest. The standard results which provide the normal level may be calculated by any suitable means which would be well known to the person of skill in the art. For example, a population of normal tissues can be assessed in terms of the level of the neoplastic markers of the present invention, thereby providing a standard value or range of values against which all future test samples are analysed. It should also be understood that the normal level may be determined from the subjects of a specific cohort and for use with respect to test samples derived from that cohort. Accordingly, there may be determined a number of standard values or ranges which correspond to cohorts which differ in respect of characteristics such as age, gender, ethnicity or health status. Said "normal level" may be a discrete level or a range of levels. An increase in the expression level of the subject genes relative to normal levels is indicative of the tissue being neoplastic.

Without limiting the present invention to any one theory or mode of action, although each of the genes hereinbefore described is differentially expressed, either singly or in combination, as between neoplastic versus non-neoplastic cells of the large intestine, and is therefore diagnostic of the existence of a large intestine neoplasm, the expression of some of these genes was found to exhibit particularly significant levels of sensitivity, specificity and positive and negative predictive value. Accordingly, in a preferred embodiment one would screen for and assess the expression level of one or more of these genes. To this end, and without limiting the present invention to any one theory or mode of action, the following markers were determined to be expressed in neoplastic tissue at a level of 3, 4, 5 or 7 fold greater than non-neoplastic tissue when assessed by virtue of the method exemplified herein.

There is therefore more particularly provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 22568 l_at; 227140_at; and/or

(ii) CTHRCl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Preferably, said control level is a non-neoplastic level.

In another embodiment, there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 227475_at, 204475_at, 202859_x_at, 202404_s_at; and/or

(ii) FOXQl, MMPl, IL8, COL 1A2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Preferably, said control level is a non-neoplastic level.

In yet another embodiment there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 205513_at, 204259_at, 227174_at, 21051 l_s_at, 37892_at; and/or

(ii) TCNl, MMP7, WDR72, INHBA, COLl IAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Preferably, said control level is a non-neoplastic level.

In still yet another preferred embodiment, there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method

comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

221577_x_at 232252_at 202310_s_at

213880_at 212354_at 204855_at

229215_at 203962_s_at 228754_at

202286_s_at 218963_s_at 203860_at

202935_s_at 20405 l_s_at 207850_at

205828_at 205983_at 205890_s_at

200832_s_at 21253 l_at; and/or

(ϋ) LGR5 GDFl 5 COLlAl

ASCL2 DUSP27 SERPINB5

TACSTD2 SULFl SLC6A6

FLJ37644 NEBL PCCA

MMP3 KRT23 CXCL3

SCD SFRP4 UBD

DPEPl LCN2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a neoplastic large intestine cell or a cell predisposed to the onset of a neoplastic state.

Preferably, said control level is a non-neoplastic level.

According to these aspects of the present invention, said large intestine tissue is preferably colorectal tissue.

The detection method of the present invention can be performed on any suitable biological sample. To this end, reference to a "biological sample" should be understood as a reference to any sample of biological material derived from an animal such as, but not limited to, cellular material, biofluids (eg. blood), faeces, tissue biopsy specimens, surgical specimens or fluid which has been introduced into the body of an animal and subsequently removed (such as, for example, the solution retrieved from an enema wash). The biological sample which is tested according to the method of the present invention may be tested directly or may require some form of treatment prior to testing. For example, a biopsy or surgical sample may require homogenisation prior to testing or it may require sectioning for in situ testing of the qualitative expression levels of individual genes. Alternatively, a cell sample may require

permeabilisation prior to testing. Further, to the extent that the biological sample is not in liquid form, (if such form is required for testing) it may require the addition of a reagent, such as a buffer, to mobilise the sample.

To the extent that the neoplastic marker gene expression product is present in a biological sample, the biological sample may be directly tested or else all or some of the nucleic acid or protein material present in the biological sample may be isolated prior to testing. In yet another example, the sample may be partially purified or otherwise enriched prior to analysis. For example, to the extent that a biological sample comprises a very diverse cell population, it may be desirable to enrich for a sub-population of particular interest. It is within the scope of the present invention for the target cell population or molecules derived therefrom to be treated prior to testing, for example, inactivation of live virus or being run on a gel. It should also be understood that the biological sample may be freshly harvested or it may have been stored (for example by freezing) prior to testing or otherwise treated prior to testing (such as by undergoing culturing).

The choice of what type of sample is most suitable for testing in accordance with the method disclosed herein will be dependent on the nature of the situation. Preferably, said sample is a faecal (stool) sample, enema wash, surgical resection, tissue biopsy or blood sample.

In a related aspect, it has been determined that certain of the markers hereinbefore defined are more indicative of adenoma development versus cancer development or vice versa. This is an extremely valuable finding since it enables one to more specifically characterise the likely nature of a neoplasm which is detected by virtue of the method of the present invention.

Accordingly, in a related aspect the present invention is directed to a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 201328_at 205825_at 219956_at

201468_s_at 205927_s_at 221922_at

201656_at 206239_s_at 223447_at

201925_s_at 207158_at 223970_at

201926_s_at 210445_at 225835_at

202718_at 210519_s_at 226360_at

202831_at 211429_s_at 228303_at

202833 s at 212063 at 228653 at

203124_s_at 213975_s_at 228915_at

203313_s_at 214235_at 231832_at

203860_at 214651_s_at 231941_s_at

203895_at 217523_at 232176_at

203896_s_at 217867_x_at 23248 l_s_at

20440 l_at 218086_at 23433 l_s_at

204885_s_at 21821 l_s_at 235210_s_at

205174_s_at 218796_at 235976_at

205366_s_at 219630_at 236894_at

205470_s_at 219682_s_at 238017_at

205513_at 219727_at 238984_at

205765_at 219955_at; 24103 l_at; and/or

(ϋ) APOBECl HOXB6 QPCT

BACE2 IGFBP2 RDHE2

C20orf42 ITGA6 REG4

CD44 KCNN4 RETNLB

CD55 KLKI l RP5-875H10.1

CTSE LlTDl RPESP

CYP3A5 LILRBl SERPINAl

CYP3A5P2 MLPH SLCl 1A2

DACHl MSLN SLC12A2

DUOX2 MUC20 SLITRK6

ETS2 NLFl SPINKl

FABP6 NPDCl TBX3

FAM84A NQOl TCNl

GALNT6 PCCA TGIFl

GPSM2 PCSKl WDR51B

GPX2 PDZKlIPl ZNRF3

HOXA9 PLCB4

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another preferred embodiment of this aspect of the present invention there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

e gene, genes or transcripts detected by Affymetπx probeset IDs

200660_ at 205476_at 217430_x_at

200665_ s_at 205479_s_at 217996_at

200832_ s_at 205713_s_at 218507_at

200903_ s_at 205815_at 218963_s_at

201014_ s_at 205828_at 218984_at

201112_ s_at 205886_at 219787_s_at

201195_ s_at 205890_s_at 21991 l_s_at

201261_ x at 205910_s_at 221729_at

201292_ at 20594 l_s_at 221730_at

201338_ x at 206224_at 221731_x_at

201479_ at 206976_s_at 221923_s_at

201577_ at 207173_x_at 37892_at

201601_ x at 207457_s_at 222449_at

201666_ at 208079_s_at 222450_at

202310_ s_at 208712_at 222549_at

202311_ s_at 209218_at 222608_s_at

202403_ s_at 209309_at 223062_s_at

202404_ s_at 209752_at 224428_s_at

202431_ s_at 209773_s_at 224646_x_at

202504_ .at 209774_x_at 224915_x_at

202779_ s_at 209792_s_at 225295_at

202859_ x_at 209875_s_at 225520_at

202954_ at 209955_s_at 225664_at

202998_ s at 210052_s_at 22568 l_at

203083_ at 21051 l_s_at 225767_at

203213_ at 210559_s_at 225799_at

203878_ s_at 210766_s_at 225806_at

204051_ s_at 211506_s_at 226227_x_at

204127_ at 21228 l_s_at 226237_at

204170_ s_at 212344_at 22631 l_at

204320_ at 212353_at 226777_at

204470_ _at 212354_at 226835_s_at

204475_ at 213905_x_at 227140_at

204580_ at 214022_s_at 229802_at

204620_ _s_at 214974_x_at 231766_s_at

204702 s at 215091_s_at; 23215 l_at; and/or

20536 l_s_at

(ϋ) AHCY DKCl PSATl

ANLN ECT2 PUS7

AURKA FAP REGlA

AZGPl GTF3A REGlB

BGN H19 REG3A

C14orf94 HIG2 RFC3

C20orfl99 HSPHl RRM2

CCL20 IFITMl SlOOAI l

CCNDl IL8 SCD

CDC2 INHBA SFRP4

CDCA7 KLKlO SLC39A10

CDHI l KRT23 SLC7A5

CEL LOC541471 SLCO4A1

CKS2 LOXL2 SPARC

CLDNl LY6G6D SPPl

COLlOAl MMPl SQLE

COLI lAl MMPI l SULFl

COLl 2Al MMP 12 THBS2

COLlAl MMP3 TIMPl

COL 1A2 MTHFDlL TMEM97

COL5A2 MYC TMEPAI

COL8A1 NFE2L3 TOP2A

COMP NMEl TPX2

CSElL NPMl TRIM29

CSTl PAICS UBD

CTHRCl PFDN4 UBE2C

CXCLl PHLDAl UBE2S

CXCL2 PLAU VCAN

CXCL5

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to control levels is indicative of a cancer cell or a cell predisposed to the onset of a cancerous state.

According to these aspects, said control levels are preferably non-neoplastic levels and said large intestine tissue is colorectal tissue. Even more preferably, said biological sample is a stool sample or blood sample.

In a related aspect, it has been determined that a subpopulation of the markers of the present invention are not only expressed at levels higher than normal levels, their expression pattern is uniquely characterised by the fact that expression levels above that of background control levels are not detectable in non-neoplastic tissue. This determination has therefore enabled the development of qualitative screening systems which are simply designed to detect marker expression relative to a control background level. In accordance with this aspect of the present invention, said "control level" is therefore the "background level". Preferably, said background level is of the chosen testing methodology.

According to this aspect, there is therefore provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 202286 s at 235976 at 209309 at

204259_at 236894_at 211506_s_at

204885_s_at 214974_x_at

205174_s_at 238984_at 219787_s_at

205825_at 24103 l_at 37892_at

207850_at 202311_s_at 222608_s_at

213880_at 204320_at 223062_s_at

217523_at 204475_at 225806_at

227174_at 204702_s_at 226237_at

228915_at 205910_s_at 227140_at

232252_at 206224_at 229802_at; and/or

(ϋ) MMPl PCSKl ANLN

MMP7 CSTl DACHl

LGR5 QPCT COLI lAl

WDR72 ECT2 C14orf94

COLI lAl SLITRK6 AZGPl

COLlAl LlTDl REG4

DUSP27 KIAAl 199 NFE2L3

NLFl PSATl CEL

IL8 CXCL5 CD44

TACSTD2 CXCL3 COL8A1

MSLN

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of a neoplastic cell or a cell predisposed to the onset of a neoplastic state.

In a most preferred embodiment, said genes or transcripts are selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

227140_at 227174_at 24103 l_at

204475_at 37892_at 211506_s_at

204259_at 202311_s_at 202286_s_at

213880__at 232252_at 226237_at; and/or

(») MMPl COLI lAl IL8

MMP7 COLlAl TACSTD2

LGR5 DUSP27 COL8A1

WDR72 NLFl

Preferably, said neoplasm is an adenoma or an adenocarcinoma and said gastrointestinal tissue is colorectal tissue.

In yet another embodiment, it has been determined that a further subpopulation of these markers are more characteristic of adenoma development, while others are more characteristic of cancer development. Accordingly, there is provided a convenient means of qualitatively obtaining indicative information in relation to the characteristics of the subject neoplasm.

According to this embodiment there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

204885_s_at 217523_at 236894_at

205174_s_at 228915_at 238984_at

205825_at 235976_at 24103 l_at; and/or

(ii) CD44 MSLN QPCT

DACHl NLFl REG4

LlTDl PCSKl SLITRK6

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In yet still another preferred embodiment there is provided a method of screening for the onset or predisposition to the onset of a large intestine neoplasm in an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene or genes detected by Affymetπx probeset IDs:

202311_s_at 209309_at 223062_s_at

204320_at 211506_s_ at 225806_at

204475_at 214974_x_ at 226237_at

204702_s_at 219787_s_ at 227140_at

205910_s_at 37892_at 229802_at; and/or

206224_at 222608_s_ at

(ϋ) ANLN COLlAl IL8

AZGPl COL8A1 MMPl

C14orf94 CSTl NFE2L3

CEL CXCL5 PSATl

COLI lAl ECT2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to background levels is indicative of a cancer cell or a cell predisposed to the onset of a cancerous state.

Preferably, said large intestine tissue is colorectal tissue.

More preferably, said biological sample is a blood sample or stool sample.

As detailed hereinbefore, the present invention is designed to screen for a neoplastic cell or cellular population, which is located in the large intestine. Accordingly, reference to "cell or cellular population" should be understood as a reference to an individual cell or a group of cells. Said group of cells may be a diffuse population of cells, a cell suspension, an encapsulated population of cells or a population of cells which take the form of tissue.

Reference to "expression" should be understood as a reference to the transcription and/or

translation of a nucleic acid molecule. In this regard, the present invention is exemplified with respect to screening for neoplastic marker expression products taking the form of RNA transcripts (eg primary RNA or mRNA). Reference to "RNA" should be understood to encompass reference to any form of RNA, such as primary RNA or mRNA. Without limiting the present invention in any way, the modulation of gene transcription leading to increased or decreased RNA synthesis will also correlate with the translation of some of these RNA transcripts (such as mRNA) to produce a protein product. Accordingly, the present invention also extends to detection methodology which is directed to screening for modulated levels or patterns of the neoplastic marker protein products as an indicator of the neoplastic state of a cell or cellular population. Although one method is to screen for mRNA transcripts and/or the corresponding protein product, it should be understood that the present invention is not limited in this regard and extends to screening for any other form of neoplastic marker expression product such as, for example, a primary RNA transcript. It is well within the skill of the person of skill in the art to determine the most appropriate screening target for any given situation. To this end, the genes which are known to encode an expression product which is either secreted by the cell or membrane bound is detailed in the table, below. It would be appreciated that screening for neoplastic markers which are secreted or membrane bound may provide particular advantages in terms of the design of a diagnostic screening product.

The gene or genes detected )y Affymetrix probe Nos:

200600. at 203961 . at 212077. at 224724 . at

200832_ s at 203962. _s_at 212281 . _s_at 224915 . x_at

200903_ s_at 204006 . s at 212344 . at 225295. at

200974_ at 204122. .at 212353 . at 225520. at

201058_ s at 204127. at 212354 . at 225541. at

201069_ at 204170 . s at 212942 . _s_at 225664 . at

201 105_ .at 204320 . at 213125 . .at 225710 at

201112 . _s_at 204351 . at 213524 . _s_at 225767 . at

201195. s at 204401 . at 213869 . x at 225799 . at

201328 . _at 204404 . at 213880 . at 225806. at

201341. at 204702 . s at 214022 . s at 225835 . at

201416. _at 204885 . s at 214235 . at 226227 . x at

201417 . .at 205361 . s at 214651. _s_at 226311. at

201426 . _s_at 205366 . _s_at 215049 . x at 226360 . .at

201468 . _s_at 205547 . _s_at 217523 . at 226694 . at

201479 . at 205765 . .at 217762 s at 226777 . .at

201563_ at 205825 . at 217763 . _s_at 226835 . _s_at

201601_ x at 205890 . s at 217764 s at 227099 . s at

201616_ _s_at 205927 . _s_at 217867 . x at 227140 . at

201656 at 205941 . _s_at 217996 . at 227174 . at

201667_ at 205983 . at 218086 . at 227475 . at

201925_ s_at 206286 . s at 218211 . s at 227566 . _at

201926_ s at 206976 . s at 218559 . _s_at 228303 . at

202237_ at 207158 . at 218704 . at 228653_ at

202238_ s at 207173 . x at 218796 . _at 228754 at

202286_ _s_at 207191 . s at 218872 . at 228915_ at

202431_ s at 208079 s_at 218963 . _s_at 229215 . . at

202450_ s_at 208712 . at 218984 at 229802 . at

202504_at 208782 . .at 219630 . at 231766 . s at

202620_ s_at 208788 . at 219682 . s at 231832 . at

202779_ s at 208850 . _s_at 219727 . _at 231879_ . at

202831_at 208851 . _s_at 219787 . _s_at 232151 . at

202878_ s at 209156 . s at 219911 . _s_at 232176 . at

202917_s_at 209218 . at 219955 . _at 232252 . . at

202935_s_at 209369 . at 219956 . at 232481_ s_at

202936_s_at 209596 at 221011 . _s_at 233555 . s_at

202954_at 209773 . s at 221922 . at 234331 . _s_at

203124_s_at 209955 . s at 221923 . _s_at 234994_ at

203213_at 210052 . s at 222449 . at 235210_ s_at

203256_at 210445_ _at 222450 . at 235976_ at

203313_s_at 210519 s_at 222549_ . at 236894_ at

203382_s_at 210559 . s at 222608 . _s_at 238017_ at

203645_s_at 210766 . _s_at 223062 . s_at

203860_at 211964 . at 223235 . s_at 241031_ at

203878_s at 211966 . at 224428 . s at 37892_at

203895_at 212063 . at 224646 . x at 60474_at

203896_s at 212070 . . at 224694 . at

ACTA2 DUOX2 LOC541471 SlOOAl 1

AHCY DUSP27 MAFB S100A8

ANLN ECT2 MLPH SlOOP

ANTXRl ELOVL5 MMPIl SCD

ANXA3 ENCl MMP2 SLCl 1A2

APOBECl ETS2 MSLN SLCl2A2

APOE FABP6 MSN SLC39A10

ASCL2 FAM84A MTHFDlL SLC6A6

AURKA FAP MXRA5 SLC7A5

BACE2 FCGR3B MYC SLCO4A1

C14orf94 FLJ37644 MYL9 SLITRK6

C20orfl99 FOXQl NEBL SM0C2

C20orf42 FSTLl NFE2L3 SORD

CALDl G0S2 NLFl S0X4

CCNDl GALNT6 NNMT S0X9

CD 163 GJAl NPDCl SQLE

CD44 GPR56 NPMl SULFl

CD55 GPSM2 NQOl SULF2

CD93 GPX2 0LFML2B TACSTD2

CDC2 H19 PALM2-AKAP2 TAGLN

CDCA7 PCCA TBX3

CDHI l HNT PCSKl TDGFl

CDH3 HOXA9 PDZKlIPl TESC

CKS2 HOXB6 PFDN4 TGIFl

CLDNl HSPHl PHLDAl THYl

COLlOAl IFITMl PLCB4 TMEM97

COLI lAl ISLR PLOD2 TMEPAI

COL 12Al ITGA6 PSATl TPX2

COL4A2 KCNN4 PUS7 TRIM29

COL6A2 KIAAl 199 RAB31 TYROBP

CSElL KIAA1913 RDHE2 UBD

CTSE KRT23 RFC3 UBE2C

CTSK LlTDl RNF43 UBE2S

CYP3A5 LBH RP5-875H10.1 VIM

CYP3A5P2 LGALSl RPESP WDR51B

DACHl LGR5 RPL22L1 WDR72

DKCl LOC387763 RRM2 ZNRF3

DPEPl

Reference to "nucleic acid molecule" should be understood as a reference to both deoxyribonucleic acid molecules and ribonucleic acid molecules and fragments thereof. The present invention therefore extends to both directly screening for mRNA levels in a biological sample or screening for the complementary cDNA which has been reverse-transcribed from an mRNA population of interest. It is well within the skill of the person of skill in the art to

design methodology directed to screening for either DNA or RNA. As detailed above, the method of the present invention also extends to screening for the protein product translated from the subject mRNA.

Preferably, the level of gene expression is measured by reference to genes which encode a protein product and, more particularly, said level of expression is measured at the protein level. Accordingly, to the extent that the present invention is directed to screening for markers which are detailed in the preceding table, said screening is preferably directed to the encoded protein.

As detailed hereinbefore, it should be understood that although the present invention is exemplified with respect to the detection of expressed nucleic acid molecules (e.g. mRNA), it also encompasses methods of detection based on screening for the protein product of the subject genes. The present invention should also be understood to encompass methods of detection based on identifying both proteins and/or nucleic acid molecules in one or more biological samples. This may be of particular significance to the extent that some of the neoplastic markers of interest may correspond to genes or gene fragments which do not encode a protein product. Accordingly, to the extent that this occurs it would not be possible to test for a protein and the subject marker would have to be assessed on the basis of transcription expression profiles.

In terms of screening for the upregulation of expression of a marker it would also be well known to the person of skill in the art that changes which are detectable at the DNA level are indicative of changes to gene expression activity and therefore changes to expression product levels. Such changes include but are not limited to, changes to DNA methylation.

Accordingly, reference herein to "screening the level of expression" and comparison of these "levels of expression" to control "levels of expression" should be understood as a reference to assessing DNA factors which are related to transcription, such as gene/DNA methylation patterns.

The term "protein" should be understood to encompass peptides, polypeptides and proteins (including protein fragments). The protein may be glycosylated or unglycosylated and/or may contain a range of other molecules fused, linked, bound or otherwise associated to the protein such as amino acids, lipids, carbohydrates or other peptides, polypeptides or proteins. Reference herein to a "protein" includes a protein comprising a sequence of amino acids as well as a protein associated with other molecules such as amino acids, lipids, carbohydrates or other peptides, polypeptides or proteins.

The proteins encoded by the neoplastic markers of the present invention may be in multimeric

form meaning that two or more molecules are associated together. Where the same protein molecules are associated together, the complex is a homomultimer. An example of a homomultimer is a homodimer. Where at least one marker protein is associated with at least one non-marker protein, then the complex is a heteromultimer such as a heterodimer.

Reference to a "fragment" should be understood as a reference to a portion of the subject nucleic acid molecule or protein. This is particularly relevant with respect to screening for modulated RNA levels in stool samples since the subject RNA is likely to have been degraded or otherwise fragmented due to the environment of the gut. One may therefore actually be detecting fragments of the subject RNA molecule, which fragments are identified by virtue of the use of a suitably specific probe.

Reference to the "onset" of a neoplasm, such as adenoma or adenocarcinoma, should be understood as a reference to one or more cells of that individual exhibiting dysplasia. In this regard, the adenoma or adenocarcinoma may be well developed in that a mass of dysplastic cells has developed. Alternatively, the adenoma or adenocarcinoma may be at a very early stage in that only relatively few abnormal cell divisions have occurred at the time of diagnosis. The present invention also extends to the assessment of an individual's predisposition to the development of a neoplasm, such as an adenoma or adenocarcinoma. Without limiting the present invention in any way, changed levels of the neoplastic markers may be indicative of that individual's predisposition to developing a neoplasia, such as the future development of an adenoma or adenocarcinoma or another adenoma or adenocarcinoma.

In yet another related aspect of the present invention, markers have been identified which enable the characterisation of neoplastic tissue of the large intestine in terms of whether it is an adenoma or a cancer. This development now provides a simple yet accurate means of characterising tissue using means other than the traditional methods which are currently utilised.

According to this aspect of the present invention, there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 200884_at 214234_s_at 226248_s_at

203240_at 214235_at 226302_at

203963 at 214433 s at 227676 at

204508_s_at 215125_s_at 227719_at

204607_at 215867_x_at 227725_at

204811_s_at 217109_at 228232_s_at

204895_x_at 217110_s_at 229070_at

204897_at 21821 l_s_at 231832_at

205259_at 219543_at 232176_at

205765_at 219955_at 23248 l_s_at

205927_s_at 221841_s_at 235976_at

208063_s_at 221874_at 236894_at

208937_s_at 223969_s_at 23752 l_x_at

210107_at 223970_at 24260 l_at

213106_at; and/or

(ϋ) CLCAl CTSE ATP8B1

FCGBP C6orfl05 CACNA2D2

HMGCS2 CKB KLF4

RETNLB ATP8A1 CYP3A5P2

LlTDl MUC4 CAPN9

SLITRK6 UGTlAl NR3C2

VSIG2 SELENBPl PBLD

LOC253012 PTGER4 CA12

ST6GALNAC1 MLPH WDR51B

IDl KIAA1324 FAM3D

CYP3A5

in said cell or cellular population wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer cell level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another aspect there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

200600_at 204006_s_at 213428_s_at 200665_s_at 20405 l_s_at 213524_s_at 200832 s at 204122 at 213869 x at

200974_at 204320_at 213905_x_at

200986_at 204475_at 214247_s_at

201058_s_at 204620_s_at 215049_x_at

201069_at 205479_s_at 215076_s_at

201105_at 205547_s_at 215646_s_at

201141_at 205828_at 216442_x_at

201147_s_at 207173_x_at 217430_x_at

201150_s_at 207191_s_at 217762_s_at

201 162_at 208747_s_at 217763_s_at

201163_s_at 208782_at 217764_s_at

201 185_at 208788_at 218468_s_at

201261_x_at 208850_s_at 218469_at

201289_at 20885 l_s_at 218559_s_at

201426_s_at 209101_at 218638_s_at

201438_at 209156_s_at 219087_at

201616_s_at 209218_at 22101 l_s_at

201645_at 209395_at 221729_at

201667_at 209396_s_at 221730_at

201744_s_at 209596_at 221731_x_at

201792_at 209875_s_at 37892_at

201842_s_at 209955_s_at 223122_s_at

201852_x_at 210095_s_at 223235_s_at

201859_at 210495_x_at 224560_at

201893_x_at 210511_s_at 224694_at

202237_at 210764_s_at 224724_at

202238_s_at 210809_s_at 225664_at

202283_at 211161_s_at 22568 l_at

20229 l_s_at 211571_s_at 225710_at

202310_s_at 211719_x_at 225799_at

202311_s_at 211813_x_at 226237_at

202403_s_at 211896_s_at 22631 l_at

202404_s_at 211959_at 226694_at

202450_s_at 211964_at 226777_at

202620_s_at 211966_at 226930_at

202766_s_at 211980_at 227099_s_at

202859_x_at 211981_at 227140_at

202878_s_at 212077_at 227566_at

202917 s at 212344 at 229218 at

202998_s_at 212353_at 229802_at

203083_at 212354_at 231579_s_at

203325_s_at 212464_s_at 231766_s_at

203382_s_at 212488_at 231879_at

203477_at 212489_at 232458_at

203570_at 212667_at 233555_s_at

203645_s_at 213125_at 234994_at

203878_s_at; and/or

(ϋ) COL1A2 LGALSl SRGN

CTHRCl ELO VL5 LBH

FNl MGP CTGF

POSTN MMP2 TNC

SPPl L0XL2 G0S2

MMPl MYL9 SQLE

SPARC DCN EFEMPl

LUM CALDl APOE

GREMl FBNl MSN

IL8 MMP3 IGFBP3

IGFBP5 IGFBP7 SERPINFl

SFRP2 FSTLl ISLR

SULFl COL4A2 HNT

ASPN VCAN C0L5A1

COL6A3 SMOC2 OLFML2B

COL8A1 HTRAl KIAA1913

COL12A1 CYR61 PALM2-AKAP2

COL5A2 FAP SERPINGl

CDHI l VIM TYROBP

THBS2 TIMP2 ACTA2

COL 15Al SCD COL3A1

COLI lAl TIMP3 PL0D2

S100A8 AEBPl MMPI l

FNDCl GJAl CD163

SFRP4 NNMT FCGR3B

INHBA COLlAl PLAU

COL6A2 SULF2 MAFB

ANTXRl COL6A1 LOC541471

GPNMB SP0N2 LOC387763

BGN CTSK CHI3L1

TAGLN MXRA5 THYl

COL4A1 CIS LOXLl

RAB31 DKK3 CD93

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal adenoma cell level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

Preferably, said gastrointestinal tissue is colorectal tissue.

Reference to an "adenoma control level" or "cancer control level" should be understood as a reference to the level of said gene expression in a population of adenoma or cancer gastrointestinal cells, respectively. As discussed hereinbefore in relation to "normal levels", the subject level may be a discrete level or a range of levels. Accordingly, the definition of "adenoma control level" or "cancer control level" should be understood to have a corresponding definition to "normal level", albeit in the context of the expression of genes by a neoplastic population of large intestine cells.

In terms of this aspect of the present invention, the subject analysis is performed on a population of neoplastic cells. These cells may be derived in any manner, such as sloughed of neoplastic cells which have been collected via an enema wash or from a gastrointestinal sample, such as a stool sample. Alternatively, the subject cells may have been obtained via a biopsy or other surgical technique.

Without limiting this aspect of the invention in any way, several of the markers of this aspect of the present invention have been determined to be expressed at particularly significant levels above those of neoplastic cells. For example, increased expression levels of 3- and 5-fold have been observed in respect of the following markers, when assessed by the method exemplified herein, which are indicative of gastrointestinal adenomas.

In another example, increased expression levels of between 3- and 9-fold have been observed in respect of the following markers which are indicative of gastrointestinal cancers, when assessed by the method herein exemplified:

According to this embodiment, there is therefore provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene or genes detected by Affymetrix probeset IDs: 210107_at; and/or

(») CLCAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer control level is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another embodiment, there is provided a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 203240_at 219955_at 24260 l_at

204607_at 23248 l_s_at 227725_at

223969_s_at 228232_s_at; and/or

(H) FCGBP LlTDl LOC253012

HMGCS2 SLITRK6 ST6GALNAC1

RETNLB VSIG2

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or group (ii) relative to a gastrointestinal cancer control level is

indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

Preferably, said gastrointestinal tissue is colorectal tissue.

Still more preferably, said biological sample is a tissue sample.

In another preferred embodiment the present invention is directed to a method of characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 202404_s_at 210809_s_at 227140_at

22568 l_at 209875_s_at 204475_at

212464_s_at; and/or

(ii) COL1A2 FNl SPPl

CTHRCl POSTN MMPl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

In yet another preferred embodiment the present invention is directed to a method of characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 200665_s_at 218468_s_at 211959_at

201744_s_at 202859_x_at; and/or

(ii) SPARC GREMl IGFBP5

LUM IL8

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

In still yet another preferred embodiment the present invention is directed to a method of

characterising a cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs:

223122_s_at 207173_x_at 21051 l s at

212353_at 203083_at 209156_s_at

219087_at 203477_at 224694_at

201438_at 37892_at 201141_at

226237_at 202917_s_at 213905_x_at

225664_at 226930_at 205547_s_at

221730_at 20405 l_s_at; and/or

(ϋ) SFRP2 CDHI l INHBA

SULFl THBS2 COL6A2

ASPN COL 15Al ANTXRl

COL6A3 COLI lAl GPNMB

COL8A1 S100A8 BGN

COL 12Al FNDCl TAGLN

COL5A2 SFRP4

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to a gastrointestinal adenoma control level is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

Preferably, said gastrointestinal tissue is colorectal tissue.

Even more preferably, said biological sample is a tissue sample.

In still another related aspect it has been determined that a subset of the markers of this aspect of the present invention are useful as qualitative markers of neoplastic tissue characterisation in that these markers, if detectable above background levels in neoplastic tissue are indicative of either adenoma or cancerous tissue.

According to this aspect, the present invention provides a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 235976_at 236894_at; and/or

(ii) SLITRK6 LlTDl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to neoplastic tissue background levels is indicative of an adenoma cell or a cell predisposed to the onset of an adenoma state.

In another aspect the present invention provides a method of characterising a neoplastic cell or cellular population, which cell or cellular population is derived from the large intestine of an individual, said method comprising measuring the level of expression of one or more genes or transcripts selected from:

(i) the gene, genes or transcripts detected by Affymetrix probeset IDs: 202311_s_at 209396_s_at 226237_at

204320_at 215646_s_at 227140_at

204475_at 37892_at 229802_at

209395_at; and/or

(ii) COLlAl VCAN CHI3L 1

MMPl COL8A1 COLI lAl

in a biological sample from said individual wherein a higher level of expression of the genes or transcripts of group (i) and/or (ii) relative to neoplastic tissue background neoplastic cell levels is indicative of a cancer or a cell predisposed to the onset of a cancerous state.

Preferably, said gastrointestinal tissue is colorectal tissue.

Still more preferably, said biological sample is a tissue sample.

In a most preferred embodiment, the methods of the present invention are preferably directed to screening for proteins encoded by the markers of the present invention.

Although the preferred method is to detect the expression products of the neoplastic markers for the purpose of diagnosing neoplasia development or predisposition thereto, the detection of converse changes in the levels of said markers may be desired under certain circumstances, for example, to monitor the effectiveness of therapeutic or prophylactic treatment directed to modulating a neoplastic condition, such as adenoma or adenocarcinoma development. For example, where elevated levels of the subject markers indicate that an individual has

developed a condition characterised by adenoma or adenocarcinoma development, for example, screening for a decrease in the levels of these markers subsequently to the onset of a therapeutic regime may be utilised to indicate reversal or other form of improvement of the subject individual's condition.

The method of the present invention is therefore useful as a one-time test or as an on-going monitor of those individuals thought to be at risk of neoplasia development or as a monitor of the effectiveness of therapeutic or prophylactic treatment regimes directed to inhibiting or otherwise slowing neoplasia development. In these situations, mapping the modulation of neoplastic marker expression levels in any one or more classes of biological samples is a valuable indicator of the status of an individual or the effectiveness of a therapeutic or prophylactic regime which is currently in use. Accordingly, the method of the present invention should be understood to extend to monitoring for increases or decreases in marker expression levels in an individual relative to their normal level (as hereinbefore defined), background control levels, cancer levels, adenoma levels or relative to one or more earlier marker expression levels determined from a biological sample of said individual.

Means of testing for the subject expressed neoplasm markers in a biological sample can be achieved by any suitable method, which would be well known to the person of skill in the art, such as but not limited to:

(i) In vivo detection.

Molecular Imaging may be used following administration of imaging probes or reagents capable of disclosing altered expression of the markers in the intestinal tissues.

Molecular imaging (Moore et al, BBA, 1402:239-249, 1988; Weissleder et al, Nature Medicine 6:351-355, 2000) is the in vivo imaging of molecular expression that correlates with the macro-features currently visualized using "classical" diagnostic imaging techniques such as X-Ray, computed tomography (CT), MRI, Positron

Emission Tomography (PET) or endoscopy.

(ii) Detection of up-regulation of RNA expression in the cells by Fluorescent In Situ

Hybridization (FISH), or in extracts from the cells by technologies such as Quantitative Reverse Transcriptase Polymerase Chain Reaction (QRTPCR) or Flow cytometric qualification of competitive RT-PCR products (Wedemeyer et al, Clinical Chemistry 48:9 1398-1405, 2002).

(iii) Assessment of expression profiles of RNA, for example by array technologies (Alon et

al, Proc. Natl. Acad. ScL USA: 96, 6745-6750, June 1999).

A "microarray" is a linear or multi-dimensional array of preferably discrete regions, each having a defined area, formed on the surface of a solid support. The density of the discrete regions on a microarray is determined by the total numbers of target polynucleotides to be detected on the surface of a single solid phase support. As used herein, a DNA microarray is an array of oligonucleotide probes placed onto a chip or other surfaces used to detect complementary oligonucleotides from a complex nucleic acid mixture. Since the position of each particular group of probes in the array is known, the identities of the target polynucleotides can be determined based on their binding to a particular position in the microarray.

Recent developments in DNA microarray technology make it possible to conduct a large scale assay of a plurality of target nucleic acid molecules on a single solid phase support. U.S. Pat. No. 5,837,832 (Chee et al.) and related patent applications describe immobilizing an array of oligonucleotide probes for hybridization and detection of specific nucleic acid sequences in a sample. Target polynucleotides of interest isolated from a tissue of interest are hybridized to the DNA chip and the specific sequences detected based on the target polynucleotides' preference and degree of hybridization at discrete probe locations. One important use of arrays is in the analysis of differential gene expression, where the profile of expression of genes in different cells or tissues, often a tissue of interest and a control tissue, is compared and any differences in gene expression among the respective tissues are identified. Such information is useful for the identification of the types of genes expressed in a particular tissue type and diagnosis of conditions based on the expression profile.

In one example, RNA from the sample of interest is subjected to reverse transcription to obtain labelled cDNA. See U.S. Pat. No. 6,410,229 (Lockhart et al) The cDNA is then hybridized to oligonucleotides or cDNAs of known sequence arrayed on a chip or other surface in a known order. In another example, the RNA is isolated from a biological sample and hybridised to a chip on which are anchored cDNA probes. The location of the oligonucleotide to which the labelled cDNA hybridizes provides sequence information on the cDNA, while the amount of labelled hybridized RNA or cDNA provides an estimate of the relative representation of the RNA or cDNA of interest. See Schena, et al. Science 270:467-470 (1995). For example, use of a cDNA microarray to analyze gene expression patterns in human cancer is described by DeRisi, et al. {Nature Genetics 14:457-460 (1996)).

In a preferred embodiment, nucleic acid probes corresponding to the subject nucleic

acids are made. The nucleic acid probes attached to the microarray are designed to be substantially complementary to the nucleic acids of the biological sample such that specific hybridization of the target sequence and the probes of the present invention occurs. This complementarity need not be perfect, in that there may be any number of base pair mismatches that will interfere with hybridization between the target sequence and the single stranded nucleic acids of the present invention. It is expected that the overall homology of the genes at the nucleotide level probably will be about 40% or greater, probably about 60% or greater, and even more probably about 80% or greater; and in addition that there will be corresponding contiguous sequences of about 8-12 nucleotides or longer. However, if the number of mutations is so great that no hybridization can occur under even the least stringent of hybridization conditions, the sequence is not a complementary target sequence. Thus, by "substantially complementary" herein is meant that the probes are sufficiently complementary to the target sequences to hybridize under normal reaction conditions, particularly high stringency conditions.

A nucleic acid probe is generally single stranded but can be partly single and partly double stranded. The strandedness of the probe is dictated by the structure, composition, and properties of the target sequence. In general, the oligonucleotide probes range from about 6, 8, 10, 12, 15, 20, 30 to about 100 bases long, with from about 10 to about 80 bases being preferred, and from about 15 to about 40 bases being particularly preferred. That is, generally entire genes are rarely used as probes. In some embodiments, much longer nucleic acids can be used, up to hundreds of bases. The probes are sufficiently specific to hybridize to a complementary template sequence under conditions known by those of skill in the art. The number of mismatches between the probe's sequences and their complementary template (target) sequences to which they hybridize during hybridization generally do not exceed 15%, usually do not exceed 10% and preferably do not exceed 5%, as-determined by BLAST (default settings).

Oligonucleotide probes can include the naturally-occurring heterocyclic bases normally found in nucleic acids (uracil, cytosine, thymine, adenine and guanine), as well as modified bases and base analogues. Any modified base or base analogue compatible with hybridization of the probe to a target sequence is useful in the practice of the invention. The sugar or glycoside portion of the probe can comprise deoxyribose, ribose, and/or modified forms of these sugars, such as, for example, 2'-O-alkyl ribose. In a preferred embodiment, the sugar moiety is 2'-deoxyribose; however, any sugar moiety that is compatible with the ability of the probe to hybridize to a target sequence

can be used.

In one embodiment, the nucleoside units of the probe are linked by a phosphodiester backbone, as is well known in the art. In additional embodiments, internucleotide linkages can include any linkage known to one of skill in the art that is compatible with specific hybridization of the probe including, but not limited to phosphorothioate, methylphosphonate, sulfamate (e.g., U.S. Pat. No. 5,470,967) and polyamide (i.e., peptide nucleic acids). Peptide nucleic acids are described in Nielsen et α/. (1991) Science 254: 1497-1500, U.S. Pat. No. 5,714,331, and Nielsen (1999) Curr. Opin. Biotechnol. 10:71-75.

In certain embodiments, the probe can be a chimeric molecule; i.e., can comprise more than one type of base or sugar subunit, and/or the linkages can be of more than one type within the same primer. The probe can comprise a moiety to facilitate hybridization to its target sequence, as are known in the art, for example, intercalators and/or minor groove binders. Variations of the bases, sugars, and internucleoside backbone, as well as the presence of any pendant group on the probe, will be compatible with the ability of the probe to bind, in a sequence-specific fashion, with its target sequence. A large number of structural modifications, are possible within these bounds. Advantageously, the probes according to the present invention may have structural characteristics such that they allow the signal amplification, such structural characteristics being, for example, branched DNA probes as those described by Urdea et al. {Nucleic Acids Symp. Ser., 24: 197-200 (1991)) or in the European Patent No. EP-0225,807. Moreover, synthetic methods for preparing the various heterocyclic bases, sugars, nucleosides and nucleotides that form the probe, and preparation of oligonucleotides of specific predetermined sequence, are well-developed and known in the art. A preferred method for oligonucleotide synthesis incorporates the teaching of U.S. Pat. No. 5,419,966.

Multiple probes may be designed for a particular target nucleic acid to account for polymorphism and/or secondary structure in the target nucleic acid, redundancy of data and the like. In some embodiments, where more than one probe per sequence is used, either overlapping probes or probes to different sections of a single target gene are used. That is, two, three, four or more probes, are used to build in a redundancy for a particular target. The probes can be overlapping (i.e. have some sequence in common), or are specific for distinct sequences of a gene. When multiple target polynucleotides are to be detected according to the present invention, each probe or probe group corresponding to a particular target polynucleotide is situated in a discrete area of the microarray.

Probes may be in solution, such as in wells or on the surface of a micro-array, or attached to a solid support. Examples of solid support materials that can be used include a plastic, a ceramic, a metal, a resin, a gel and a membrane. Useful types of solid supports include plates, beads, magnetic material, microbeads, hybridization chips, membranes, crystals, ceramics and self-assembling monolayers. One example comprises a two-dimensional or three-dimensional matrix, such as a gel or hybridization chip with multiple probe binding sites (Pevzner et al, J. Biomol. Struc. & Dyn. 9:399-410, 1991; Maskos and Southern, Nuc. Acids Res. 20: 1679-84, 1992). Hybridization chips can be used to construct very large probe arrays that are subsequently hybridized with a target nucleic acid. Analysis of the hybridization pattern of the chip can assist in the identification of the target nucleotide sequence. Patterns can be manually or computer analyzed, but it is clear that positional sequencing by hybridization lends itself to computer analysis and automation. In another example, one may use an Affymetrix chip on a solid phase structural support in combination with a fluorescent bead based approach. In yet another example, one may utilise a cDNA microarray. In this regard, the oligonucleotides described by Lockkart et al. (i.e. Affymetrix synthesis probes in situ on the solid phase) are particularly preferred, that is, photolithography.

As will be appreciated by those in the art, nucleic acids can be attached or immobilized to a solid support in a wide variety of ways. By "immobilized" herein is meant the association or binding between the nucleic acid probe and the solid support is sufficient to be stable under the conditions of binding, washing, analysis, and removal. The binding can be covalent or non-covalent. By "non-covalent binding" and grammatical equivalents herein is meant one or more of either electrostatic, hydrophilic, and hydrophobic interactions. Included in non-covalent binding is the covalent attachment of a molecule, such as streptavidin, to the support and the non-covalent binding of the biotinylated probe to the streptavidin. By "covalent binding" and grammatical equivalents herein is meant that the two moieties, the solid support and the probe, are attached by at least one bond, including sigma bonds, pi bonds and coordination bonds.

Covalent bonds can be formed directly between the probe and the solid support or can be formed by a cross linker or by inclusion of a specific reactive group on either the solid support or the probe or both molecules. Immobilization may also involve a combination of covalent and non-covalent interactions.

Nucleic acid probes may be attached to the solid support by covalent binding such as by conjugation with a coupling agent or by covalent or non-covalent binding such as electrostatic interactions, hydrogen bonds or antibody-antigen coupling, or by

combinations thereof. Typical coupling agents include biotin/avidin, biotin/streptavidin, Staphylococcus aureus protein A/IgG antibody F c fragment, and streptavidin/protein A chimeras (T. Sano and C. R. Cantor, Bio/Technology 9:1378-81 (1991)), or derivatives or combinations of these agents. Nucleic acids may be attached to the solid support by a photocleavable bond, an electrostatic bond, a disulfide bond, a peptide bond, a diester bond or a combination of these sorts of bonds. The array may also be attached to the solid support by a selectively releasable bond such as 4,4'- dimethoxytrityl or its derivative. Derivatives which have been found to be useful include 3 or 4 [bis-(4-methoxyphenyl)]-methyl-benzoic acid, N-succinimidyl-3 or 4 [bis-(4-methoxyphenyl)]-methyl-benzoic acid, N-succinimidyl-3 or 4 [bis-(4- methoxyphenyl)]-hydroxymethyl-benzoic acid, N-succinimidyl-3 or 4 [bis-(4- methoxyphenyl)]-chloromethyl-benzoic acid, and salts of these acids.

In general, the probes are attached to the microarray in a wide variety of ways, as will be appreciated by those in the art. As described herein, the nucleic acids can either be synthesized first, with subsequent attachment to the microarray, or can be directly synthesized on the microarray.

The microarray comprises a suitable solid substrate. By "substrate" or "solid support" or other grammatical equivalents herein is meant any material that can be modified to contain discrete individual sites appropriate for the attachment or association of the nucleic acid probes and is amenable to at least one detection method. The solid phase support of the present invention can be of any solid materials and structures suitable for supporting nucleotide hybridization and synthesis. Preferably, the solid phase support comprises at least one substantially rigid surface on which the primers can be immobilized and the reverse transcriptase reaction performed. The substrates with which the polynucleotide microarray elements are stably associated and may be fabricated from a variety of materials, including plastics, ceramics, metals, acrylamide, cellulose, nitrocellulose, glass, polystyrene, polyethylene vinyl acetate, polypropylene, polymethacrylate, polyethylene, polyethylene oxide, polysilicates, polycarbonates,

Teflon, fluorocarbons, nylon, silicon rubber, polyanhydrides, polyglycolic acid, polylactic acid, polyorthoesters, polypropylfumerate, collagen, glycosaminoglycans, and polyamino acids. Substrates may be two-dimensional or three-dimensional in form, such as gels, membranes, thin films, glasses, plates, cylinders, beads, magnetic beads, optical fibers, woven fibers, etc. A preferred form of array is a three- dimensional array. A preferred three-dimensional array is a collection of tagged beads. Each tagged bead has different primers attached to it. Tags are detectable by signalling means such as color (Luminex, Illumina) and electromagnetic field (Pharmaseq) and

signals on tagged beads can even be remotely detected (e.g., using optical fibers). The size of the solid support can be any of the standard microarray sizes, useful for DNA microarray technology, and the size may be tailored to fit the particular machine being used to conduct a reaction of the invention. In general, the substrates allow optical detection and do not appreciably fluoresce.

In one embodiment, the surface of the microarray and the probe maybe derivatized with chemical functional groups for subsequent attachment of the two. Thus, for example, the microarray is derivatized with a chemical functional group including, but not limited to, amino groups, carboxy groups, oxo groups and thiol groups, with amino groups being particularly preferred. Using these functional groups, the probes can be attached using functional groups on the probes. For example, nucleic acids containing amino groups can be attached to surfaces comprising amino groups, for example using linkers as are known in the art; for example, homo-or hetero-bifunctional linkers as are well known. In addition, in some cases, additional linkers, such as alkyl groups

(including substituted and heteroalkyl groups) may be used.

In this embodiment, the oligonucleotides are synthesized as is known in the art, and then attached to the surface of the solid support. As will be appreciated by those skilled in the art, either the 5' or 3' terminus may be attached to the solid support, or attachment may be via an internal nucleoside. In an additional embodiment, the immobilization to the solid support may be very strong, yet non-covalent. For example, biotinylated oligonucleotides can be made, which bind to surfaces covalently coated with streptavidin, resulting in attachment.

The arrays may be produced according to any convenient methodology, such as preforming the polynucleotide microarray elements and then stably associating them with the surface. Alternatively, the oligonucleotides may be synthesized on the surface, as is known in the art. A number of different array configurations and methods for their production are known to those of skill in the art and disclosed in WO 95/25116 and WO

95/35505 (photolithographic techniques), U.S. Pat. No. 5,445,934 (in situ synthesis by photolithography), U.S. Pat. No. 5,384,261 (in situ synthesis by mechanically directed flow paths); and U.S. Pat. No. 5,700,637 (synthesis by spotting, printing or coupling); the disclosure of which are herein incorporated in their entirety by reference. Another method for coupling DNA to beads uses specific ligands attached to the end of the

DNA to link to ligand-binding molecules attached to a bead. Possible ligand-binding partner pairs include biotin-avidin/streptavidin, or various antibody/antigen pairs such as digoxygenin-antidigoxygenin antibody (Smith et al., Science 258:1122-1126 (1992)). Covalent chemical attachment of DNA to the support can be accomplished by

using standard coupling agents to link the 5'-phosphate on the DNA to coated microspheres through a phosphoamidate bond. Methods for immobilization of oligonucleotides to solid-state substrates are well established. See Pease et al, Proc. Natl. Acad. ScL USA 91(l l):5022-5026 (1994). A preferred method of attaching oligonucleotides to solid-state substrates is described by Guo et al, Nucleic Acids Res.

22:5456-5465 (1994). Immobilization can be accomplished either by in situ DNA synthesis (Maskos and Southern, supra) or by covalent attachment of chemically synthesized oligonucleotides (Guo et al., supra) in combination with robotic arraying technologies.

In addition to the solid-phase technology represented by microarray arrays, gene expression can also be quantified using liquid-phase assays. One such system is kinetic polymerase chain reaction (PCR). Kinetic PCR allows for the simultaneous amplification and quantification of specific nucleic acid sequences. The specificity is derived from synthetic oligonucleotide primers designed to preferentially adhere to single-stranded nucleic acid sequences bracketing the target site. This pair of oligonucleotide primers form specific, non-covalently bound complexes on each strand of the target sequence. These complexes facilitate in vitro transcription of double- stranded DNA in opposite orientations. Temperature cycling of the reaction mixture creates a continuous cycle of primer binding, transcription, and re-melting of the nucleic acid to individual strands. The result is an exponential increase of the target dsDNA product. This product can be quantified in real time either through the use of an intercalating dye or a sequence specific probe. SYBR(r) Green 1, is an example of an intercalating dye, that preferentially binds to dsDNA resulting in a concomitant increase in the fluorescent signal. Sequence specific probes, such as used with TaqMan technology, consist of a fluorochrome and a quenching molecule covalently bound to opposite ends of an oligonucleotide. The probe is designed to selectively bind the target DNA sequence between the two primers. When the DNA strands are synthesized during the PCR reaction, the fluorochrome is cleaved from the probe by the exonuclease activity of the polymerase resulting in signal dequenching. The probe signalling method can be more specific than the intercalating dye method, but in each case, signal strength is proportional to the dsDNA product produced. Each type of quantification method can be used in multi-well liquid phase arrays with each well representing primers and/or probes specific to nucleic acid sequences of interest. When used with messenger RNA preparations of tissues or cell lines, an array of probe/primer reactions can simultaneously quantify the expression of multiple gene products of interest. See Germer et al, Genome Res. 10:258-266 (2000); Heid et al, Genome Res. 6:986-994 (1996).

(iv) Measurement of altered neoplastic marker protein levels in cell extracts, for example by immunoassay.

Testing for proteinaceous neoplastic marker expression product in a biological sample can be performed by any one of a number of suitable methods which are well known to those skilled in the art. Examples of suitable methods include, but are not limited to, antibody screening of tissue sections, biopsy specimens or bodily fluid samples.

To the extent that antibody based methods of diagnosis are used, the presence of the marker protein may be determined in a number of ways such as by Western blotting, ELISA or flow cytometry procedures. These, of course, include both single-site and two-site or "sandwich" assays of the non-competitive types, as well as in the traditional competitive binding assays. These assays also include direct binding of a labelled antibody to a target.

Sandwich assays are among the most useful and commonly used assays. A number of variations of the sandwich assay technique exist, and all are intended to be encompassed by the present invention. Briefly, in a typical forward assay, an unlabelled antibody is immobilized on a solid substrate and the sample to be tested brought into contact with the bound molecule. After a suitable period of incubation, for a period of time sufficient to allow formation of an antibody-antigen complex, a second antibody specific to the antigen, labelled with a reporter molecule capable of producing a detectable signal is then added and incubated, allowing time sufficient for the formation of another complex of antibody-antigen-labelled antibody. Any unreacted material is washed away, and the presence of the antigen is determined by observation of a signal produced by the reporter molecule. The results may either be qualitative, by simple observation of the visible signal, or may be quantitated by comparing with a control sample. Variations on the forward assay include a simultaneous assay, in which both sample and labelled antibody are added simultaneously to the bound antibody.

These techniques are well known to those skilled in the art, including any minor variations as will be readily apparent.

In the typical forward sandwich assay, a first antibody having specificity for the marker or antigenic parts thereof, is either covalently or passively bound to a solid surface. The solid surface is typically glass or a polymer, the most commonly used polymers being cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene. The solid supports may be in the form of tubes, beads, discs of microplates, or any other surface suitable for conducting an immunoassay. The binding processes are well-

known in the art and generally consist of cross-linking, covalently binding or physically adsorbing, the polymer-antibody complex is washed in preparation for the test sample. An aliquot of the sample to be tested is then added to the solid phase complex and incubated for a period of time sufficient (e.g. 2-40 minutes) and under suitable conditions (e.g. 25°C) to allow binding of any subunit present in the antibody.

Following the incubation period, the antibody subunit solid phase is washed and dried and incubated with a second antibody specific for a portion of the antigen. The second antibody is linked to a reporter molecule which is used to indicate the binding of the second antibody to the antigen.

An alternative method involves immobilizing the target molecules in the biological sample and then exposing the immobilized target to specific antibody which may or may not be labelled with a reporter molecule. Depending on the amount of target and the strength of the reporter molecule signal, a bound target may be detectable by direct labelling with the antibody. Alternatively, a second labelled antibody, specific to the first antibody is exposed to the target-first antibody complex to form a target-first antibody-second antibody tertiary complex. The complex is detected by the signal emitted by the reporter molecule.

By "reporter molecule" as used in the present specification, is meant a molecule which, by its chemical nature, provides an analytically identifiable signal which allows the detection of antigen-bound antibody. Detection may be either qualitative or quantitative. The most commonly used reporter molecules in this type of assay are either enzymes, fluorophores or radionuclide containing molecules (i.e. radioisotopes) and chemiluminescent molecules.

In the case of an enzyme immunoassay, an enzyme is conjugated to the second antibody, generally by means of glutaraldehyde or periodate. As will be readily recognized, however, a wide variety of different conjugation techniques exist, which are readily available to the skilled artisan. Commonly used enzymes include horseradish peroxidase, glucose oxidase, beta-galactosidase and alkaline phosphatase, amongst others. The substrates to be used with the specific enzymes are generally chosen for the production, upon hydrolysis by the corresponding enzyme, of a detectable color change. Examples of suitable enzymes include alkaline phosphatase and peroxidase. It is also possible to employ fluorogenic substrates, which yield a fluorescent product rather than the chromogenic substrates noted above. In all cases, the enzyme-labelled antibody is added to the first antibody hapten complex, allowed to bind, and then the excess reagent is washed away. A solution containing the appropriate substrate is then added to the complex of antibody-antigen-antibody. The substrate will react with the enzyme linked

to the second antibody, giving a qualitative visual signal, which may be further quantitated, usually spectrophotometrically, to give an indication of the amount of antigen which was present in the sample. "Reporter molecule" also extends to use of cell agglutination or inhibition of agglutination such as red blood cells on latex beads, and the like.

Alternately, fluorescent compounds, such as fluorecein and rhodamine, may be chemically coupled to antibodies without altering their binding capacity. When activated by illumination with light of a particular wavelength, the fluorochrome- labelled antibody adsorbs the light energy, inducing a state to excitability in the molecule, followed by emission of the light at a characteristic color visually detectable with a light microscope. As in the EIA, the fluorescent labelled antibody is allowed to bind to the first antibody-hapten complex. After washing off the unbound reagent, the remaining tertiary complex is then exposed to the light of the appropriate wavelength the fluorescence observed indicates the presence of the hapten of interest.

Immunofluorescence and EIA techniques are both very well established in the art and are particularly preferred for the present method. However, other reporter molecules, such as radioisotope, chemiluminescent or bioluminescent molecules, may also be employed.

(v) Without limiting the present invention to any one theory or mode of action, during development gene expression is regulated by processes that alter the availability of genes for expression in different cell lineages without any alteration in gene sequence, and these states can be inherited through a cell division - a process called epigenetic inheritance. Epigenetic inheritance is determined by a combination of DNA methylation (modification of cytosine to give 5-methyl cytosine, 5meC) and by modifications of the histone chromosomal proteins that package DNA. Thus methylation of DNA at CpG sites and modifications such as deacetylation of histone H3 on lysine 9, and methylation on lysine 9 or 27 are associated with inactive chromatin, while the converse state of a lack of DNA methylation, acetylation of lysine 9 of histone H3 is associated with open chromatin and active gene expression. In cancer, this epigenetic regulation of gene expression is frequently found to be disrupted (Esteller & Herman, 2000; Jones & Baylin, 2002). Genes such as tumour suppressor or metastasis suppressor genes are often found to be silenced by DNA methylation, while other genes may be hypomethylated and inappropriately expressed. Thus, among genes that elevated or inappropriate expression in cancer, this in some instances is characterised by a loss of methylation of the promoter or regulatory region of the gene.

A variety of methods are available for detection of aberrantly methylated DNA of a

specific gene, even in the presence of a large excess of normal DNA (Clark 2007). Thus, elevated expression of certain genes may be detected through detection of the presence of hypomethylated sequences in tissue, bodily fluid or other patient samples.

Epigenetic alterations and chromatin changes in cancer are also evident in the altered association of modified histones with specific genes(Esteller, 2007); for example activated genes are often found associated with histone H3 that is acetylated on lysine 9 and methylated on lysine 4. The use of antibodies targeted to altered histones allows for the isolation of DNA associated with particular chromatin states and has potential use in cancer diagnosis.

(vi) Determining altered expression of protein neoplastic markers on the cell surface, for example by immunohistochemistry.

(vii) Determining altered protein expression based on any suitable functional test, enzymatic test or immunological test in addition to those detailed in points (iv) and (v) above.

A person of ordinary skill in the art could determine, as a matter of routine procedure, the appropriateness of applying a given method to a particular type of biological sample.

Without limiting the present invention in any way, and as detailed above, gene expression levels can be measured by a variety of methods known in the art. For example, gene transcription or translation products can be measured. Gene transcription products, i.e., RNA, can be measured, for example, by hybridization assays, run-off assays., Northern blots, or other methods known in the art.

Hybridization assays generally involve the use of oligonucleotide probes that hybridize to the single-stranded RNA transcription products. Thus, the oligonucleotide probes are complementary to the transcribed RNA expression product. Typically, a sequence-specific probe can be directed to hybridize to RNA or cDNA. A "nucleic acid probe", as used herein, can be a DNA probe or an RNA probe that hybridizes to a complementary sequence. One of skill in the art would know how to design such a probe such that sequence specific hybridization will occur. One of skill in the art will further know how to quantify the amount of sequence specific hybridization as a measure of the amount of gene expression for the gene was transcribed to produce the specific RNA.

The hybridization sample is maintained under conditions that are sufficient to allow specific hybridization of the nucleic acid probe to a specific gene expression product. "Specific hybridization", as used herein, indicates near exact hybridization (e.g., with few if any

mismatches). Specific hybridization can be performed under high stringency conditions or moderate stringency conditions. In one embodiment, the hybridization conditions for specific hybridization are high stringency. For example, certain high stringency conditions can be used to distinguish perfectly complementary nucleic acids from those of less complementarity. "High stringency conditions", "moderate stringency conditions" and "low stringency conditions" for nucleic acid hybridizations are explained on pages 2.10.1-2.10.16 and pages 6.3.1-6.3.6 in Current Protocols in Molecular Biology (Ausubel, F. et al, "Current Protocols in Molecular Biology", John Wiley & Sons, (1998), the entire teachings of which are incorporated by reference herein). The exact conditions that determine the stringency of hybridization depend not only on ionic strength (e.g., 0.2.times.SSC, 0.1. times. SSC), temperature (e.g., room temperature, 42 0 C, 68 0 C.) and the concentration of destabilizing agents such as formamide or denaturing agents such as SDS, but also on factors such as the length of the nucleic acid sequence, base composition, percent mismatch between hybridizing sequences and the frequency of occurrence of subsets of that sequence within other non- identical sequences. Thus, equivalent conditions can be determined by varying one or more of these parameters while maintaining a similar degree of identity or similarity between the two nucleic acid molecules. Typically, conditions are used such that sequences at least about 60%, at least about 70%, at least about 80%, at least about 90% or at least about 95% or more identical to each other remain hybridized to one another. By varying hybridization conditions from a level of stringency at which no hybridization occurs to a level at which hybridization is first observed, conditions that will allow a given sequence to hybridize (e.g., selectively) with the most complementary sequences in the sample can be determined.

Exemplary conditions that describe the determination of wash conditions for moderate or low stringency conditions are described in Kraus, M. and Aaronson, S., 1991. Methods Enzymol., 200:546-556; and in, Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, (1998). Washing is the step in which conditions are usually set so as to determine a minimum level of complementarity of the hybrids. Generally, starting from the lowest temperature at which only homologous hybridization occurs, each 0 C. by which the final wash temperature is reduced (holding SSC concentration constant) allows an increase by 1% in the maximum mismatch percentage among the sequences that hybridize. Generally, doubling the concentration of SSC results in an increase in T m of about 17 0 C. Using these guidelines, the wash temperature can be determined empirically for high, moderate or low stringency, depending on the level of mismatch sought. For example, a low stringency wash can comprise washing in a solution containing 0.2.times.SSC/0.1% SDS for 10 minutes at room temperature; a moderate stringency wash can comprise washing in a pre-warmed solution (42 0 C) solution containing 0.2.times.SSC/0.1% SDS for 15 minutes at 42 0 C; and a high stringency wash can comprise washing in pre-warmed (68 0 C.) solution containing

0.1. times. SSC/0.1% SDS for 15 minutes at 68 0 C. Furthermore, washes can be performed repeatedly or sequentially to obtain a desired result as known in the art. Equivalent conditions can be determined by varying one or more of the parameters given as an example, as known in the art, while maintaining a similar degree of complementarity between the target nucleic acid molecule and the primer or probe used (e.g., the sequence to be hybridized).

A related aspect of the present invention provides a molecular array, which array comprises a plurality of:

(i) nucleic acid molecules comprising a nucleotide sequence corresponding to any one or more of the neoplastic marker genes hereinbefore described or a sequence exhibiting at least 80% identity thereto or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(ii) nucleic acid molecules comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iii) nucleic acid probes or oligonucleotides comprising a nucleotide sequence capable of hybridising to any one or more of the sequences of (i) under medium stringency conditions or a functional derivative, fragment, variant or homologue of said nucleic acid molecule; or

(iv) probes capable of binding to any one or more of the proteins encoded by the nucleic acid molecules of (i) or a derivative, fragment or, homologue thereof

wherein the level of expression of said marker genes of (i) or proteins of (iv) is indicative of the neoplastic state of a cell or cellular subpopulation derived from the large intestine.

Preferably, said percent identity is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%.

Low stringency includes and encompasses from at least about 1% v/v to at least about 15% v/v formamide and from at least about IM to at least about 2M salt for hybridisation, and at least about IM to at least about 2M salt for washing conditions. Alternative stringency conditions may be applied where necessary, such as medium stringency, which includes and encompasses from at least about 16% v/v at least about 30% v/v formamide and from at least about 0.5M to at least about 0.9M salt for hybridisation, and at least about 0.5M to at least

about 0.9M salt for washing conditions, or high stringency, which includes and encompasses from at least about 31% v/v to at least about 50% v/v formamide and from at least about 0.01M to at least about 0.15M salt for hybridisation, and at least about 0.01M to at least about 0.15M salt for washing conditions. In general, washing is carried out at T n , = 69.3 + 0.41 (G + C) % [ 19] = - 12°C. However, the T m of a duplex DNA decreases by 1 °C with every increase of 1% in the number of mismatched based pairs (Bonner et al (1973) J. MoI. Biol. 81:123).

Preferably, the subject probes are designed to bind to the nucleic acid or protein to which they are directed with a level of specificity which minimises the incidence of non-specific reactivity. However, it would be appreciated that it may not be possible to eliminate all potential cross-reactivity or non-specific reactivity, this being an inherent limitation of any probe based system.

In terms of the probes which are used to detect the subject proteins, they may take any suitable form including antibodies and aptamers.

A library or array of nucleic acid or protein probes provides rich and highly valuable information. Further, two or more arrays or profiles (information obtained from use of an array) of such sequences are useful tools for comparing a test set of results with a reference, such as another sample or stored calibrator. In using an array, individual probes typically are immobilized at separate locations and allowed to react for binding reactions. Primers associated with assembled sets of markers are useful for either preparing libraries of sequences or directly detecting markers from other biological samples.

A library (or array, when referring to physically separated nucleic acids corresponding to at least some sequences in a library) of gene markers exhibits highly desirable properties. These properties are associated with specific conditions, and may be characterized as regulatory profiles. A profile, as termed here refers to a set of members that provides diagnostic information of the tissue from which the markers were originally derived. A profile in many instances comprises a series of spots on an array made from deposited sequences.

A characteristic patient profile is generally prepared by use of an array. An array profile may be compared with one or more other array profiles or other reference profiles. The comparative results can provide rich information pertaining to disease states, developmental state, receptiveness to therapy and other information about the patient.

Another aspect of the present invention provides a diagnostic kit for assaying biological samples comprising an agent for detecting one or more neoplastic markers and reagents useful

for facilitating the detection by said agent. Further means may also be included, for example, to receive a biological sample. The agent may be any suitable detecting molecule.

The present invention is further described by the following non-limiting examples:

EXAMPLE 1

Methods and Materials

Affymetrix GeneChip data

Gene expression profiling data and accompanying clinical data was purchased from GeneLogic Inc (Gaithersburg, MD USA). For each tissue analyzed, oligonucleotide microarray data for 44,928 probesets (Affymetrix HGU133A & HGU133B, combined), experimental and clinical descriptors, and digitally archived microscopy images of histological preparations were received. A quality control analysis was performed to remove arrays not meeting essential quality control measures as defined by the manufacturer.

Transcript expression levels were calculated by both Microarray Suite (MAS) 5.0 (Affymetrix) and the Robust Multichip Average (RMA) normalization techniques (Affymetrix. GeneChip expression data analysis fundamentals. Affymetrix, Santa Clara, CA USA, 2001; Hubbell et al. Bioinformatics, 18: 1585-1592, 2002; Irizarry et al. Nucleic Acid Research, 31, 2003)MAS normalized data was used for performing standard quality control routines and the final data set was normalized with RMA for all subsequent analyses.

Univariate differential expression

Differentially expressed gene transcripts were identified using a moderated t-test implemented in the limma library downloaded from the Bioconductor repository for R. (G. K. Smyth.

Statistical Applications in Genetics and Molecular Biology, 3(l):Article 3, 2004; G K Smyth.

Bioinformatics and Computational Biology Solutions using R and Bioconductor. Springer, New York, 2005). Significance estimates (p-values) were corrected to adjust for multiple hypothesis testing using the Bonferonni correction.

Tissue specific expression patterns

To construct a filter for hypothetically 'turned on' gene expression the mean expression level for all 44,928 probesets across the full range of 454 tissues was first estimated. To estimate an expression on/off threshold, the 44,928 mean values were ranked and the expression value equivalent to the 30th percentile across the dataset calculated. This arbitrary threshold was chosen because it was theorized that the majority of transcripts (and presumably more than 30%) in a given specimen should be transcriptionally silenced. Thus this threshold represents

a conservative upper bound for what is estimated as non-specific, or background, signal.

Gene symbol annotations

To map Affymetrix probeset names to official gene symbols the annotation metadata available from Bioconductor was used. hgul33plus2 library version 1.16.0, which was assembled using Entrez Gene data downloaded on 15 March 2007, was used.

Estimates of performance characteristics

Diagnostic utility for each table of markers shown herein was estimated including: sensitivity, specificity, positive predictive value, negative predictive value, likelihood ratio positive, likelihood ratio negative. These estimates were calculated in the same data used to discover the markers and will therefore potentially overestimate the performance characteristics in future tissue samples. To improve the generalisabilty of the estimates a modified jackknife resampling technique was used to calculate a less biased value for each characteristic.

Results

A range of univariate statistical tests were applied on Affymetrix oligonucleotide microarray data to reveal human genes that could be used to discriminate colorectal neoplastic tissues from non-neoplastic tissues. There were further identified a number of gene transcripts that appear to be useful for differentiating colorectal adenomas from colorectal carcinoma. Also identified were a subset of these transcripts that may have particular diagnostic utility because due to the protein products being either secreted or displayed on the cell surface of epithelial cells. Finally, there were identified a further subset of transcripts expressed specifically in neoplastic tissues and at low- or near-background levels in non-neoplastic tissues.

Genes differentially expressed in neoplastic tissues

From a total GeneChip set of 44,928 probesets it was determined that over 11,000 probesets were differentially expressed by moderated t-test using the limma package in BioConductor (G. K. Smyth, 2004 supra) employing conservative (Bonferroni) multiple test correction. When this list was further filtered to include only those probesets demonstrating a 2-fold or greater mean expression change between the neoplastic and non-neoplastic tissues, 206 probesets were found to be expressed higher in neoplasias relative to normals.

These 205 probesets were annotated using the most recent metadata and annotation packages available for the chips. The 205 overexpressed probesets were mapped to 174 gene symbols.

Hypothetical markers specific for colorectal neoplasia

While differential gene expression patterns are useful for diagnostic purposes, this project also seeks to identify diagnostic proteins shed into the lumen of the gut by neoplastic colorectal epithelia. To discover candidate proteins the list of differentially expressed transcripts were filtered with a selection criteria aimed at identifying markers specifically expressed in colorectal neoplasia tissues. This filter criteria is based on a theoretical assumption that most genes on the GeneChip will be turned 'off and that any microarray signals for such 'off transcripts will reflect technical assay background and non-specific oligonucleotide binding. Accordingly, to select genes specifically expressed in neoplastic tumours (i.e. 'on') the nonneoplastic signals were compared with a hypothetical background signal threshold from across all genes on the chip. By design, all transcripts in the candidate pool from which the 'on' transcripts are chosen are at least two fold overexpressed in the diseased tissues. Combined, it is hypothesized that these criteria yield the subset of differentially expressed genes that are specifically expressed in neoplasia. The expression profile for a representative 'on' transcript is shown in Figure 1.

Genes differentially expressed between adenomas and cancer tissues

There were 33 transcripts observed that were differentially expressed at least two-fold higher in adenoma tissues relative to cancer tissues. In particular, there were identified several transcripts that exhibit an expression pattern specific for adenomas, including SLITRK6 and LlTDl, shown in Figure 2.

Further, there were also identified cancer specific transcripts. The expression profile of one such transcript, COLl IAl is shown in Figure 3.

EXAMPLE 2

Probesets Elevated In Neoplasia

Differential expression analysis was applied to Affymetrix gene chip data measuring RNA concentration in 454 colorectal tissues including 161 adenocarcinoma specimens, 29 adenoma specimens, 42 colitis specimens and 222 non-diseased tissues. Using conservative corrections for multiple hypothesis testing, it was determined that over 25% of the 44,928 probesets measured in each tissue experiment were differentially expressed between the 190 neoplasia specimens and 264 non-neoplasia controls. To identify robust biomarkers for colorectal neoplasia the list of putative probeset biomarkers were further filtered to include only those probesets shown to be expressed at least 2-fold higher in neoplastic vs. non-neoplastic tissues.

205 probesets hybridising to approximately 157 putative genes were observed to be expressed at a statistically significant higher level in neoplastic tissues relative to non-neoplastic controls.

Validation/Hypothesis testing

To validate these discovery results the hybridisation of 199 candidate probesets were measured against RNA extracts from 68 clinical specimens comprising 19 adenomas, 19 adenocarcinomas, and 30 non-diseased controls using a custom-designed 'Adenoma Gene Chip 1 . Six (6) probesets were not tested as they were not included on the custom design. It was confirmed that 186/199 (88%) of the target probesets or probesets which also hybridise to the target locus were likewise differentially expressed (P < 0.05) in these independently- derived tissues. The results of testing these probesets in 68 independently collected clinical specimens is shown in Table 1.

We further tested the 142 of the 157 unique gene loci to which the 205 probesets are understood to hybridise. We note the remaining 15 gene symbols were not represented in the validation data. We observed that 133 of 142 gene symbols were represented in the validation data by at least one differentially expressed probeset and many symbols included multiple probesets against regions across the putative locus. A complete list of probesets that bind to target loci is shown in Table 2.

Conclusion

The candidate probesets shown in Tables 1 and 2 are differentially expressed in neoplastic colorectal tissues compared to non-neoplastic controls.

EXAMPLE 3

Probesets Demonstrating A Neoplasia-Specific Profile

During analysis of the data, a novel expression profile was observed between neoplastic and non-neoplastic phenotypes. It was hypothesized that a subset of quantitatively differentially expressed probesets are furthermore qualitatively differentially expressed. Such probesets show evidence of a neoplasia-specific gene expression profile, i.e. these probesets appear to be expressed above background levels in neoplastic tissues only. This observation and the resulting hypothesis are based on two principles:

1. That the majority of human transcripts that are present on a genome-wide GeneChip (e.g. U 133) will not likely be expressed in the colorectal mucosa; and

2. That microarray binding intensity for such 'off probesets to labelled cRNA will reflect technical assay background, i.e. non-specific oligonucleotide binding.

To generate a list of neoplasia specific probesets the non-neoplastic intensity of differentially expressed probesets was compared with a hypothetical background signal threshold from across all probesets on the chip. Bydesign, all probesets in the candidate pool from which the

'on' transcripts are chosen are at least two fold over-expressed in the diseased tissues.

Combined, these criteria yield the subset of differentially expressed transcript species that are specifically expressed in neoplasia.

Validation/Hypothesis Testing

The custom gene chip design precludes testing the hypothetically neoplasia-specific probesets using the same principles as used for discovery. In particular, the custom gene chip (by design) does not contain a large pool of probesets anticipated to hybridise to hypothetically 'off /'non-transcribed' gene transcripts. This is because the custom gene chip design is biased toward differentially expressed transcripts in colorectal neoplastic tissues.

The usual differential expression testing (limma) was therefore to these candidate probesets for neoplasia-specific transcripts. Of the 33 probesets on the custom gene chip, 32 probesets (or probesets which bind to the same locus) were differentially expressed between the 38 neoplastic tissues (adenoma & cancer) and non-neoplastic controls. The results of these validation experiments is shown in Table 3.

All probesets which are known to hybridise to the gene loci to which the 33 probesets claimed herein were tested. Of the 32 putative gene loci targeted by the probesets, 29 were present in the validation data. Twenty-eight (28) of these 29 gene symbols demonstrated at least one hybridising probeset which was differentially expressed in the neoplastic tissues. Results for these experiments, including all probesets that bind to each target locus in a differentially expressed manner are shown in Table 4.

EXAMPLE 4

Probesets Useful For Characterizing Neoplastic Tissues

Differential expression analysis was applied to Affymetrix gene chip data measuring RNA concentration in neoplastic tissues including 161 adenocarcinoma specimens and 29 adenoma specimens. It was observed that 43 probesets hybridizing to approximately 33 putative gene symbols were expressed higher (P < 0.05) in adenoma tissues relative to cancer tissues.

Conversely, 145 probesets (104 gene symbols) were identified to be expressed higher in cancer relative to adenomas.

Validation/Hypothesis testing 188 (43 + 145) of these probesets were then measured in a set of independent clinical specimens including 19 adenoma tissues and 19 cancer tissues. It was confirmed that 158 (30 + 128) of the target probesets (or probesets against the same gene locus) were likewise differentially expressed (P < 0.05) in these independently-derived tissues. Probesets elevated in adenoma and cancers relative to each other are shown in Table 5 and Table 6 respectively.

It was further observed that 137 (33+104) gene loci are diagnostically useful for discriminating colorectal adenomas and cancers relative to each phenotype. The validation data included probesets designed to hybridise to 128 of these candidate gene symbols. It was observed that 21 of the 31 genes elevated in adenomas relative to cancers were likewise differentially expressed by at least one probeset. Of the 97 gene symbols elevated in cancer relative to adenoma it was confirmed that 89 gene symbols demonstrated at least one probeset in the validation data to be likewise differentially expressed. The validation testing of the adenoma and cancer elevated gene loci is shown in Table 7 and Table 8, respectively.

Conclusion

It was concluded that the candidate probesets shown in FIXME are differentially expressed between adenomatous and adenocarcinoma tissues and thus useful for distinguishing these tissues. Gene transcripts that hybridise to these probesets are thus diagnostically informative in a clinical setting to classify such neoplastic tissues.

EXAMPLE 5

Materials And Methods For Examples 2 To 3

Gene expression profiling data measured in 454 colorectal tissue specimens including neoplastic, normal and non-neoplastic disease controls was purchased from GeneLogic Inc (Gaithersburg, MD USA). For each tissue specimen an Affymetrix (Santa Clara, CA USA) oligonucleotide microarray data totalling 44,928 probesets (HGU133A & HGU133B, combined), experimental and clinical descriptors, and digitally archived microscopy images of histological preparations was received. Prior to applying discovery methods to these data extensive quality control methods were carried out, including statistical exploration, review of clinical records for consistency and histopathology audit of a random sample of arrays. Microarrays that did not meet acceptable quality criteria were removed from the analysis.

Hypothesis testing

Candidate transcription biomarkers were tested using a custom oligonucleotide microarray of 25-mer oligonucleotide probesets designed to hybridise to candidate RNA transcripts identified during discovery. Differential expression hypotheses were tested using RNA extracts derived from independently collected clinical samples comprising 30 normal colorectal tissues, 19 colorectal adenoma tissues, and 19 colorectal adenocarcinoma tissues. Each RNA extract was confirmed to meet strict quality control criteria.

Colorectal tissue specimens

All tissues used for hypothesis testing were obtained from a tertiary referral hospital tissue bank in metropolitan Adelaide, Australia (Repatriation General Hospital and Flinders Medical Centre). Access to the tissue bank for this research was approved by the Research and Ethics Committee of the Repatriation General Hospital and the Ethics Committee of Flinders Medical Centre. Informed patient consent was received for each tissue studied.

Following surgical resection, specimens were placed in a sterile receptacle and collected from theatre. The time from operative resection to collection from theatre was variable but not more than 30 minutes. Samples, approximately 125mm3 (5x5x5mm) in size, were taken from the macroscopically normal tissue as far from pathology as possible, defined both by colonic region as well as by distance either proximal or distal to the pathology. Tissues were placed in cryo vials, then immediately immersed in liquid nitrogen and stored at -150C until processing.

RNA extraction

RNA extractions were performed using Trizol(R)reagent (Invitrogen, Carlsbad, CA, USA) as per manufacturer's instructions. Each sample was homogenised in 300μL of Trizol reagent using a modified Dremel drill and sterilised disposable pestles. An additional 200μL of Trizol reagent was added to the homogenate and samples were incubated at RT for 10 minutes. lOOμL of chloroform was then added, samples were shaken vortexed for 15 seconds, and incubated at RT for 3 further minutes. The aqueous phase containing target RNA was obtained by centrifugation at 12,000 rpm for 15 min, 4OC. RNA was then precipitated by incubating samples at RT for 10 min with 250μL of isopropanol. Purified RNA precipitate was collected by centrifugation at 12,000 rpm for 10 minutes, 4OC and supernatants were discarded. Pellets were then washed with ImL 75% ethanol, followed by vortexing and centrifugation at 7,500g for 8 min, 4OC. Finally, pellets were air-dried for 5 min and resuspended in 80μL of RNase free water. To improve subsequent solubility samples were incubated at 55 0 C for 10 min. RNA was quantified by measuring the optical density at

A260/280 nm. RNA quality was assessed by electrophoresis on a 1.2% agarose formaldehyde gel.

Gene Chip processing To test hypotheses related to biomarker candidates for colorectal neoplasia RNA extracts were assayed using a custom GeneChip designed in collaboration with Affymetrix (Santa Clara, CA USA). These custom GeneChips were processed using the standard Affymetrix protocol developed for the HU Gene ST 1.0 array described in (Affy:WTAssay).

Statistical software and data processing

The R statistics environment R and BioConductor libraries (BioConductor, www.bioconductor.org) (BIOC) were used for most analyses. To map probeset IDs to gene symbol on the Custom GeneChip, hgul33plus2 library version 2.2.0, which was assembled using Entrez Gene data downloaded on Apr 18 12:30:55 2008 (BIOC) was used.

Hypothesis testing of differentially expressed biomarkers

To assess differential expression between tissue classes, the Student's t test for equal means between two samples or the robust variant provided by the limma library (Smyth)(limma) was used. The impact of false discovery due to multiple hypothesis testing was mitigated by applying a Bonferroni adjustment to P values in the discovery process (MHT:Bonf). For hypotheses testing the slightly less conservative multiple hypothesis testing correction of Benjamini & Hochberg, which aims to control the false discovery rate of solutions(MHT:BH) was applied.

Discovery of tissue-specific gene expression patterns

Discovery methods using gene expression data often yield numerous candidates, many of which are not suitable for commercial products because they involve subtle gene expression differences that would be difficult to detect in laboratory practice. Pepe et al. note that the 'ideal' biomarker is detectable in tumor tissue but not detectable (at all) in non-tumour tissue (Pepe:biomarker:development.) To bias toward candidates that meet this criterion, an analysis method was developed that aims to enrich the candidates for biomarkers whose qualitative' absence or presence measurement is diagnostic for the phenotype of interest. This method attempts to select candidates that show a prototypical 'turned-on' or 'turned-off pattern relative to an estimate of the background/noise expression across the chip. Such RNA transcripts are more likely to correlate with downstream translated proteins with diagnostic potential or to predict upstream genomic changes (e.g. methylation status) that can be used diagnostically. This focus on qualitative rather than quantitative outcomes may simplify the product development process for such biomarkers.

The method is based on the assumption that the pool of extracted RNA species in any given tissue (e.g. colorectal mucosae) will specifically bind to a relatively small subset of the full set of probesets on a GeneChip designed to measure the whole genome. On this assumption, it is estimated that most probesets on a full human gene chip will not exhibit specific, high- intensity signals.

This observation is utilised to approximate the background or 'non-specific binding' across the chip by choosing a theoretical level equal to the value of e.g. lowest 30% quantile of the ranked mean values. This quantile can be arbitrarily set to some level below which a reasonable assumption is made that the signals do not represent above-background RNA binding. Finally, this background estimate is used as a threshold to estimate the 'OFF' probesets in an experiment for, say, the non-neoplastic tissue specimens.

Conversely, probesets which are 1) expressed above this theoretical threshold level and 2) at differentially higher levels in the tumour specimens may be a tumour specific candidate biomarker. In this case the concept of 'fold-change' thresholds can also be conveniently applied to further emphasize the concept of absolute expression increases in a putatively 'ON' probeset.

Given the assumption of low background binding for a sizeable fraction of the measured probesets, this method was only used in the large GeneLogic data and discovery. To construct a filter for hypothetically 'turned on' biomarkers in the GeneLogic discovery data, the mean expression level for all 44,928 probesets was first estimated across the full range of 454 tissues. The 44,928 mean values were then ranked and the expression value equivalent to the 25th percentile across the dataset calculated. This arbitrary threshold was chosen because it was theorized that the majority of transcripts (and presumably more than 25%) in a given specimen should exhibit low concentration which effectively transcriptional silence. Thus this threshold represents a conservative upper bound for what is estimated as non-specific, or background, expression.

EXAMPLE 6

Determine Gene Identity of a Nucleic Acid Sequence of Interest which is Define by an Affymetrix Probeset

BLAST the sequence of interest using online available Basic Local Alignment Search Tools [BLAST], e.g. NCBI/BLAST

(http://blast.ncbi.nlm.nih.gov/Blast.cgi)

(a) Select "Human" in BLAST ASSEMBLED GENOMES on the web page http://blast.ncbi.nlm.nih.gov/Blast.cgi

(b) Leave the default settings, i.e.: • Database: Genome (all assemblies)

• Program: megaBLAST: compare highly related nucleotide sequences

• Optional parameters: Expect: 0.01, Filter: default, Descriptions: 100, Alignments: 100

(c) Copy/Paste Sequence into the "BLAST" window (d) Click "Begin Search"

(e) Click "View Report"

Assessment of the Open BLAST Search Results

Multiple significant sequence alignments may be identified when "blasting" the sequence.

Identify gene nomenclature of the identified sequence match

(a) Click the link to one of the identified hits

(b) The new page will schematically depict the position of the hit on one chromosome. It will be apparent which gene is hit. (c) Retrieve the "hit" sequence clicking on the link

(d) Do a search for the gene in the provided "search" window. This provides the gene nucleotide coordinates for the gene.

Determine promiscuity of Sequence (a) Open the NCBI/BLAST tool, (http://blast.ncbi.nlm.nih.gov/Blast.cgi)

(b) Click on "nucleotide Blast" under "basic BLAST"

(c) Copy/paste the sequence of interest into the "Query Sequence" window

(d) Click "Blast".

Assessment of the nBLAST Search Results of the Sequence

(a) The nBLAST exercise with the Sequence may result in multiple Blast hits of which some accession entry numbers are listed in "Description".

(b) These hits should be reviewed.

Determine location of the Sequence in the Gene

The Ensembl database is an online database, which produces and maintains automatic annotation selected eukaryotic genomes (www.ensembl.orq/index.html ' )

Identify location of the Sequence in the Gene

(a) Set "Search" to Homo Sapiens, Type "the gene name" in the provided Search Field Ensemble.org/index.html)

(b) Click "Go" (c) Click the "vega protein coding Gene: OTTHUMG000000144184" link to get an annotation report

(d) Click on "Gene DAS Report" to retrieve information regarding Alternative splice site database: Type "the gene name" in search field

• Click on "the gene entry" • Scroll down to "evidence"

• Review alternative splice sites

• Click "Confirmed intron/exons" to get a list of coordinates for the exons & introns.

Alternative splicing and/or transcription

The AceView Database provides curated and non-redundant sequence representation of all public mRNA sequences. The database is available through NCBI: http://www.ncbi.nlm. nih.gov/IEB/Research/Acembly/

Further investigation of the Gene mRNA transcripts

(a) Type "the gene name" into the provided "search" field

(b) Click "Go"

(c) The following information is available from the resulting entry in AceView:

• The number of cDNA clones from which the gene is constructed (ie originated- from experimental work involving isolation of mRNA)

• The mRNAs predicted to be produced by the gene

• The existence of non-overlapping alternative exons and validated alternative polyadenylation sites

• The existence of truncations • The possibility of regulated alternate expression

• Introns recorded as participating in alternatively splicing of the gene

(d) Classic splice site motives

Application of Method to LOC6439U/hCG_1815491

Materials and Methods

Extraction of RNA

RNA extractions were performed using Trizol(R) reagent (Invitrogen, Carlsbad, CA, USA) as per manufacturer's instructions. Each sample was homogenised in 300μL of Trizol reagent using a modified dremel drill and sterilised disposable pestles. Additional 200μL of Trizol reagent was added to the homogenate and samples were incubated at RT for 10 minutes. lOOμL of chloroform was then added, samples were shaken vortexed for 15 seconds, and incubated at RT for 3 further minutes. The aqueous phase containg target RNA was obtained by centrifugation at 12,000 rpm for 15 min, 40° C. RNA was then precipitated by incubating samples at RT for 10 min with 250μL of isopropanol. Purified RNA precipitate was collected by centrifugation at 12,000 rpm for 10 minutes, 40° C and supernatants were discarded.

Pellets were then washed with ImL 75% ethanol, followed by vortexing and centrifugation at 7,500g for 8 min, 40° C . Finally, pellets were air-dried for 5 min and resuspended in 80μL of RNase free water. To improve subsequent solubility samples were incubated at 55 ° C for 10 min. RNA was quantified by measuring the optical density at A260/280 nm. RNA quality was assessed by electrophoresis on a 1.2% agarose formaldehyde gel.

Gene Chip processing

RNA samples to analyze on Human Exon 1.0 ST GeneChips were processed using the Affymetrix WT target labeling and control kit (part# 900652) following the protocol described in (Affymetrix 2007 P/N 701880 Rev.4). Briefly: First cycle cDNA was synthesized from lOOng ribosomal reduced RNA using random hexamer primers tagged with T7 promoter sequence and Superscript II (Invitrogen, Carlsbad CA), this was followed by DNA Polymerase I synthesis of the second strand cDNA. Anti-sense cRNA was then synthesized using T7 polymerase. Second cycle sense cDNA was then synthesised using Superscript II, dNTP+ dUTP, and random hexamers to produce sense strand cDNA incorporating uracil. This single stranded uracil containing cDNA was then fragmented using a combination of uracil DNA glycosylase (UDG) and apurinic/ apyrimidinic endonucleasel (APE 1). Finally the DNA was biotin labelled using terminal deoxynucleotidyl transferase (TdT) and the Affymetrix proprietary DNA Labeling reagent. Hybridization to the arrays was carried out at 45°C for 16-18hours.

Washing and staining of the hybridized GeneChips was carried out using the Affymetrix

Fluidics Station 450 and scanned with the Affymetrix Scanner 3000 following recommended protocols.

SYBR green basedjQuantitative Real Time-PCR

Quantitative real time polymerase chain reaction was performed on RNA isolated from clinical samples for the amplification and detection of the various hCG_1815491 transcripts.

Firstly cDNA was synthesized from 2ug of total RNA using the Applied Biosystems High Capacity Reverse transcription Kit (P/N 4368814). After synthesis the reaction was diluted 1 :2 with water to obtain a final volume of 40ul and IuI of this diluted cDNA used in subsequent PCR reactions.

PCR was performed in a 25ul volume using 12.5ul Promega 2x PCR master mix (P/N M7502), 1.5ul 5uM forward primer, 1.5ul 5uM reverse primer, 7.875ul water, 0.625ul of a 1:3000 dilution of 10,000x stock of SYBR green 1 pure dye (Invitrogen P/N S7567), and IuI ofcDNA.

Cycling conditions for amplification were 95° for 2minutes xl cycle, 95° for 15 seconds and 60° for 1 minute x40 cycles. The amplification reactions were performed in a Corbett

Research Rotor-Gene RG3000 or a Roche LightCycler480 real-time PCR machine. When the Roche LightCycler480 real-time PCR machine was used for amplification the reaction volume was reduced to lOul and performed in a 384 well plate but the relative ratios between all the components remained the same. Final results were calculated using the δδCt method with the expression levels of the various hCG_1815491 transcripts being calculated relative to the expression level of the endogenous house keeping gene HPRT.

End-point PCR

End point PCR was performed on RNA isolated from clinical samples for the various hCG_1815491 transcripts. Conditions were identical to those described for the SYBR green assay above but with the SYBR green dye being replaced with water. The amplification reactions were performed in a MJ Research PTC-200 thermal cycler. 2.5μl of the amplified products were analysed on 2% agarose E-gel (Invitrogen) along with a 100-base pair DNA Ladder Marker.

Results

The nucleotide structure and expression levels of transcripts related to hCG_1815491 was analysed based on the identification of diagnostic utility of Affymetrix probesets 23802 l s at and 238022_at from the gene chip analysis.

The gene hCG_1815491 is currently represented in NCBI as a single RefSeq sequence, XM 93911. The RefSeq sequence of hCG_1815491 is based on 89 GenBank accessions from 83 cDNA clones. Prior to March 2006, these clones were predicted to represent two overlapping genes, LOC388279 and LOC650242 (the latter also known as LOC643911). In March 2006, the human genome database was filtered against clone rearrangements, co- aligned with the genome and clustered in a minimal non-redundant way. As a result, LOC388272 and LOC650242 were merged into one gene named hCG_1815491 (earlier references to hCG_1815491 are: LOC388279, LOC64391 1, LOC650242, XM 944116, AF275804, XM_373688).

It has been determined that the Ref Sequence, which is defined by the genomic coordinates 8579310 to 8562303 on human chromosome 16 as defined by the NCBI contig reference NT_010498.15 | Hsl6_10655, NCBI 36 March 2006 genome encompasses hCG_1815491. The 10 predicted RNA variants derived from this gene have been aligned with the genomic nucleotide sequence residing in the map region 8579310 to 8562303. This alignment analysis revealed the existence of at least 6 exons of which several are alternatively spliced. The identified exons are in contrast to the just 4 exons specified in the NCBI hCG_1815491 RefSeq XM 93911. Two additional putative exons were also identified in the Ref Sequence by examination of included probesets on Affymetrix Genechip HuGene Exon 1.0 that target nucleotide sequences embedded in the Ref Sequence. The identified and expanded exon- intron structure of hCG_1815491 have been used to design specific oligonucleotide primers, which allowed measurement of the expression of RNA variants generated from the Ref Sequence by using PCR-based methodology (Figure 4)

EXAMPLE 7

Immunohistochemistry is a useful method for evaluating changes in local expression of up or down-regulated markers in human tissue.

Materials and Methods:

Four micrometre sections were incubated in a universal decloaking buffer for 75 minutes at

80μL to expose masked epitopes. Protein expression was determined using an antibody

targeting the C-terminal domain of Mesothelin (MSLN) on colonic biopsies from 30 patients (10 normals, 10 cancers, 10 adenomas). Antibodies were applied for one hour at room temperature. After washing, sections were incubated with polymeric horse- radish peroxidase. Antibody localization was visualized using 3 '3' diaminobenzidine.

Result:

There was a marked upregulation of MSLN in the adenoma and cancer tissues compared to the normal controls. The normal tissues showed mild staining for MSLN in the cytoplasm of the colonic epithelium but the cancer and particularly the adenomas tissues shows significant upregulation of MSLN in their multilayered epithelium. This upregulation was observed in all 10 adenomas tissue and in 9 out of the 10 cancer tissues. These patterns of staining are illustrated in Figures 4, 5 and 6.

Conclusion: Elevated expression of MSLN has been detected in colon neoplasia, confirming the upregulation observed in the mRNA expression data and verifying the diagnostic utility of both the MSLN mRNA and protein for detection of colorectal neoplasia.

EXAMPLE 8

EVIDENCE OF MMP3 PROTEIN EXPRESSION IN STOOLS OF PATIENTS WITH

COLORECTAL NEOPLASIA.

Affymetrix probeset designated 205828_at was identified to be expressed higher in 190 neoplastic tissue specimens relative to 264 non-neoplastic specimens. The probeset

205828_at hybridizes to RNA transcribed from the gene encoding Matrix Metalloproteinase 3 (MMP3) NM 002422. The differential expression profile of probeset 205828_at was further demonstrated by profiling RNA collected from 68 independent clinical specimens comprising 19 adenomas, 19 adenocarcinomas and 30 non-disease controls, Figure 7.

Materials and Methods

A commercially available bead-suspension immunoassay targeting the protein MMP3 was purchased from R&D Systems (MMP Kit reagents LMPOOO and LMP513) to measure MMP3 concentration in stools of human patients diagnosed with colorectal neoplasia. Proteins were extracted from stool specimens using a phosphate buffered saline wash from 6 non-disease controls, 10 adenoma and 11 adenocarcinoma subjects. The resulting protein extracts were analyzed using the Luminex bead-based suspension MMP3 assay as recommended by manufacturer.

Results

An elevated endogenous expression of MMP3 was observed in stool specimens from patients diagnosed with colon adenomas or adenocarcinomas relative to non-neoplastic controls (Figure 8).

Conclusion

Measurement of MMP3 protein in bodily fluids such as stool samples is useful for diagnosing colorectal neoplasia.

TABLES

Probeset designations include both HG-133plus2 probeset IDs and Human Gene LOST array probe ids. The latter can be conveniently mapped to Transcript Cluster ID using the Human Gene 1.OST probe tab file provided by Affymetrix (http://www.affymetrix.com/Auth/analysis/downloads/na22/wtge ne/HuGene- 1 0-st- yl.probe.tab.zip). Using publicly available software such as NetAffx (provided by Affymetrix), the Transcript Cluster ID may be further mapped to gene symbol, chromosomal location, etc.

Table 1.

Probesets demonstrated to be expressed higher in neoplastic tissues relative to non-neoplastic controls. TargetPS: Affymetrix HG-U133plus2 probeset id; Symbol: putative gene symbol corresponding to target probeset id - multiple symbol names indicate the possibility of probeset hybridisation to multiple gene targets; Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of neoplasia vs. non-neoplasia; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 2.

Evidence of multiple probesets which correspond to gene symbols claimed herein exhibiting RNA concentration differences between neoplasia and non-neoplastic controls. Symbol: gene

symbol; ValidPS UP: Affymetrix probeset IDs demonstrating statistically significant overexpression in neoplastic RNA extracts relative to non-neoplastic controls. Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and nonneoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of neoplasia vs. non-neoplasia; Sens- Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Example 2 Table 3.

Probesets which demonstrate a qualitatively (in addition to quantitative) elevated profile in neoplastic tissues relative to non-neoplastic controls. TargetPS: Affymetrix HG-Ul 33plus2 probeset id; Symbol: putative gene symbol corresponding to target probeset id - multiple symbol names indicate the possibility of probeset hybridisation to multiple gene targets; Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of neoplasia vs. non-neoplasia; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 4.

Evidence of multiple probesets which correspond to gene symbols claimed herein exhibiting qualitative changes in RNA concentration in neoplastic tissues. Symbol: gene symbol; ValidPS UP: Affymetrix probeset IDs demonstrating statistically significant overexpression in neoplastic RNA extracts relative to non-neoplastic controls. Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter

estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of neoplasia vs. non-neoplasia; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 5

Probesets demonstrated to be expressed higher in adenoma tissues relative to cancer tissues. TargetPS: Affymetrix HG-U133plus2 probeset id; Symbol: putative gene symbol corresponding to target probeset id - multiple symbol names indicate the possibility of probeset hybridisation to multiple gene targets; Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Beηjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of adenomas vs. cancers; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 6

Probesets demonstrated to be expressed higher in cancer tissues relative to adenoma tissues. TargetPS: Affymetrix HG-U133plus2 probeset id; Symbol: putative gene symbol corresponding to target probeset id - multiple symbol names indicate the possibility of probeset hybridisation to multiple gene targets; Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of cancer tissues vs. adenoma tissues; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 7

Evidence of multiple probesets which correspond to gene symbols claimed herein exhibiting RNA concentration differences between adenoma and cancer tissues. Symbol: gene symbol ; ValidPS UP: Affymetrix probeset IDs demonstrating statistically significant overexpression in neoplastic RNA extracts relative to non-neoplastic controls. Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of adenoma tissues vs. cancer tissues; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Table 8

Evidence of multiple probesets which correspond to gene symbols claimed herein exhibiting RNA concentration differences between cancer and adenoma tissues. Symbol: gene symbol ; ValidPS UP: Affymetrix probeset IDs demonstrating statistically significant overexpression in neoplastic RNA extracts relative to non-neoplastic controls. Signif. FDR: Adjusted p-value for mean difference testing between RNA extracted from neoplasia and non-neoplastic tissues. Adjustment is made using Benjamini & Hochberg correction for multiple hypothesis testing (Benjamini and Hochberg, 1995); D.value50: Diagnostic effectiveness parameter estimate corresponding to the area of a receiver operator characteristic ROC. This parameter provides a convenient estimate of diagnostic utility and is described in (Saunders, 2006); FC: fold change between mean expression level of cancer tissues vs. adenoma tissues; Sens-Spec: Estimate of diagnostic performance corresponding to the ROC curve point demonstrating equal sensitivity and specificity; CI (95): 95% confidence interval of sensitivity and specificity estimates.

Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features.

TABLE 1

TABLE 2

VO

Os

OO

MD

O O

O K)

O

O

O

O as

O

O oo

O

KJ

TABLE 3

TABLE 4

TACSTD2 100461 l-HuGene_st:755233-HuGene;st:815649- γACSTD 3.13E 2.3693 15.71 88.280.9-

HuGene_st:567105-HuGene_st:8281- 2 14 93.3

HuGene_st: 1041491-HuGene_st:339450-

HuGene_st:227128_s_at:653539-HuGene_st: 1079680-

HuGene_st:958592-HuGene_st:202286_s_at:620831-

HuGene_st:53075-HuGene_st: 1009541-

HuGene_st: 861600-HuGene st: 181597-

HuGene st: 130895-HuGene st:849592-HuGene st

IL8 1044664-HuGene_st:714746-HuGene_st:442029- AHNAK: 3.28E 2.3273 1.73 87.880.3-

HuGene_st: 119200-HuGene_st:550444- IGHGl 12 93

HuGene_st:493978-HuGene_st:713906-

HuGene_st:943156-HuGene_st:504843-

HuGene_st:501902-HuGene_st:497219-

HuGene_st:23149-HuGene_st:387164-

HuGene_st:562654-HuGene_st: 1047492-

HuGene_st:538558-HuGene_st:239300-

HuGene_st:211430_s_at:390695-HuGene_st: 1081759-

HuGene_st: 144630-HuGene_st:661013-

HuGene_st:393191 -HuGene_st:55017-

HuGene_st:1005159-HuGene_st:359116-

HuGene_st:373132-HuGene_st:689050-

HuGene_st:611041 -HuGene_st:715689-

HuGene_st:289354-HuGene_st:820633-

HuGene_st:555788-HuGene_st:988945-

HuGene_st:307991 -HuGene_st:211506_s_at:924368-

HuGene_st:47911-HuGene_st:929972-

HuGene_st:561100-HuGene_st:68522-

HuGene_st:217039_x_at:231668_x_at: 1080400-

HuGene_st: 141662-HuGene_st:765200-

HuGene_st:648539-HuGene_st:274868-

HuGene st:233969 at

LGR5 784585-HuGene_st:777519-HuGene_st:937559- .GR5 1.1 OE-I 2.0645 7.89 84.976.9-

HuGene_st:297045-HuGene_st:796093- 1 1 90.8

HuGene_st:783411-HuGene_st:876648-

HuGene_st:842003-HuGene_st: 102017-

HuGene_st:562251-HuGene_st:295997-

HuGene_st:747520-HuGene_st:81 1 121-

HuGene_st:677407-HuGene_st:522834-

HuGene_st:475414-HuGene_st:802999-

HuGene_st: 1066925-HuGene_st:216864-

HuGene_st: 890928-HuGene_st: 179502-

HuGene_st:79633-

HuGene st:210393 at:241266 at:213880 at

QPCT 261837-HuGenejst: 108729-HuGene_st:942514- QPCT 3.46E-I 2.0415 3.03 84.676.7-

HuGene_st: 184578-HuGene_st:454020- 1 1 90.6

HuGene_st:202482-HuGene_st: 1012784-

HuGene_st:854999-HuGene_st:62841-

HuGene_st: 271540-HuGene_st: 524919-

HuGene_st:78507-HuGene_st:89785-HuGene_st:728309-

HuGene_st:625205-HuGene_st: 170279-

HuGene_st:561083-HuGene_st:322236-

HuGene st:205174 s at:303448-HuGene st

WDR72 829740-HuGene_st:1063796-HuGene_st:725768- WDR72 5.22E-I 1.9856 14.11 8475.9- HuGene_st:227174_at:36564- 11 90 HuGene_st:236741_at:604794-HuGene_st:1040675- HuGene_st: 527199-HuGene_st: 1052168- HuGene_st: 1563874_at:177868-HuGene_st:667158- HuGene st:542462-HuGene st: 136243-

TABLE 5

TABLE 6

TABLE 7

TABLE 8

RAB31 784521-HuGene_st:805481-HuGene_st:936297- RAB31 0.0003 1.4857 2.08 77.165-86.6 HuGene_st:576193-HuGene_st:321915- HuGene_st:282618-HuGene_st:930765- HuGene_st:397177-HuGene_st:861778- HuGene_st:411822-HuGene_st:24057 l_at: 816751- HuGene_st:1028297-HuGene_st:291722- HuGene_st:722575-HuGene_st:904649- HuGene_st:991710-HuGene_st: 1032281- HuGene st:217764 s at:252384-HuGene st

LGALSl 540889-HuGene_st:882911-HuGene_st:289133- LGALSl 0.0004 1.4863 1.61 77.164.8- HuGene_st:775286-HuGene_st: 179375- 86.5 HuGene_st:877452-HuGene_st:697650- HuGene st: 1720-HuGene_st: 107624- HuGene_st:810235-HuGene_st: 190535- HuGene_st:529287-HuGene_st:725969- HuGene_st:976579-HuGene_st: 1075722- HuGene_st:453861-HuGene_st:216405_at:624673- HuGene st:724499-HuGene st

ELOVL5 559433-HuGene_st:42298 l-HuGene_st:211457- ELOVL5 0.0004 1.542 1.86 7865.8-

HuGene_st:551828-HuGene_st:412588- 87.2

HuGene_st:912245-HuGene_st:287185-

HuGene_st:103066-HuGene_st:399004-

HuGene_st:528360-HuGene_st:938564-

HuGene_st:1000247-HuGene_st:620190-

HuGene_st:336885-HuGene_st: 165821-

HuGene_st:214153_at:731356-HuGene_st:601072-

HuGene_st: 755480-HuGene_st: 8873-

HuGene_st:1567222_x_at:208788_at:1093316-

HuGene st

ANTXRl 203217-HuGene_st:501276-HuGene_st:799524- ANTXRl 0.0005 1.5167 77.665.4-

HuGene_st:85875-HuGene_st: 12099- 86.9

HuGene_st:230429-HuGene_st:368173-

HuGene_st:369297-HuGene_st:227660_at:86683-

HuGene_st:682518-HuGene_st: 129277-

HuGene_st:827509-HuGene_st:901442-

HuGene_st:408237-HuGene_st:414321-

HuGene_st:330209-

HuGene_st:234430_at:241549_at:755570-

HuGene st:220093 at

ACTA2 479447-HuGene_st: 183075-HuGene_st: 1070983- ACTA2 6.00E-04 1.4242 1.75 76.263.8-

HuGene_st:548186-HuGene_st:529704- 85.8

HuGene_st:530590-HuGene_st:630797-

HuGene_st:404109-HuGene_st:237159-

HuGene_st:90357-HuGene_st:293590-

HuGene_st: 1078299-HuGene_st: 120087-

HuGene_st:855050-HuGene_st:757648-

HuGene_st:980364-HuGene_st:785232-

HuGene st:313690-HuGene st:670805-HuGene st

IGFBP3 242422-HuGene_st: 1046998-HuGene_st:585931- IGFBP3 0.0006 1.552 1.38 78.166-87.3 HuGene_st: 145671 -HuGene_st:71362- HuGene_st:719466-HuGene_st:412306- HuGene_st:922332-HuGene_st:491408- HuGene_st:929740-HuGene_st:276131- HuGene st:201952-HuGene st

VIM 1093618-HuGene_st:398387-HuGene_st:837477- VIM 0.0008 1.4804 1.48 7764.9- HuGene_st: 1079757-HuGene_st:339807- 86.5 HuGene_st:302722-HuGene_st:139661- HuGene_st:436158-HuGene_st:564251- HuGene_st:234475-HuGene_st:770617- HuGene_st: 155888-HuGene_st:319318- HuGene_st:527110-HuGene_st:994975- HuGene st: 192324-HuGene st

ASPN 720939-HuGene_st:96944- ASPN 0.0009 1.4011 2.75 75.863.5-

HuGene_st:224396_s_at:640448- 85.6

HuGene_st: 1055545-HuGene_st:447994-

HuGene_st:835521-HuGene_st:567513-

HuGene_st:546047-HuGene_st:219087_at:673620-

HuGene st

HNT 291569-HuGene_st: 1099035-HuGene_st:237480- EINT 0.001 1.425 2.26 76.263.8- HuGene_st:222020_s_at:959054-HuGene_st:941200- 85.8 HuGene st: 715332-HuGene_st: 761810- HuGene_st:673027-HuGene_st:653559- HuGene_st:241934_at:403764-HuGene_st:719722- HuGene st:227566 at

3JAl 210697-HuGene_st:348617-HuGene_st: 100706- JJAl 0.0013 1.4083 1.63 75.963.7- HuGene_st:202715-HuGene_st:817684- 85.6 HuGene_st:851453-HuGene_st:317083- HuGene_st:852817-HuGene_st:150947- HuGene_st:491742-HuGene_st:612542- HuGene st:352375-HuGene st:684816-HuGene st

MSN 77554-HuGene_st: 103225-HuGene_st:1038630- MSN 1.40E-03 1.4927 1.36 77.265.1-

HuGene_st:555838-HuGene_st:250445- 86.7

HuGene_st:770842-HuGene_st:240960_at:905037-

HuGene_st:693942-HuGene_st:788076-

HuGene st:102934-HuGene st:686613-HuGene st

SFRP4 965138-HuGene_st:512201-HuGene_st:321583- SFRP4 0.0015 1.407 2.2 75.963.5-

HuGene_st:784119-HuGene_st:4568- 85.5

HuGene_st:1073348-HuGene_st:606783-

HuGene_st:203475-HuGene_st:779727-

HuGene_st:236875-HuGene_st:674172-

HuGene st:204051 s at:1084814-HuGene st

:TGF 418229-HuGene_st:547481 -HuGene st: 661089- :TGF 0.0015 1.334 1.78 74.862.4- HuGene_st:338622-HuGene_st:353624- 84.6 HuGene_st:466254-HuGene_st:691815- HuGene st: 156933-HuGene st: 1031177- HuGene_st:265466-HuGene_st: 1079612- HuGene st: 1073900-HuGene st: 1080114-HuGene st

FCGR3B 775320-HuGene_st :483078-HuGene_st:897088- FCGR3B 0.0026 1.232 2.14 73.1 HuGene_st:891504- ■HuGene_st:288407- HuGene_st:367930- ■HuGene_st:512343- HuGene_st:524627- ■HuGene_st:290249- HuGene_st:204007_ at:993144-HuGene_st: 1084295- HuGene_st:606544- ηuGene_st: 160442- HuGene_st:974942- ■HuGene_st:582410- HuGene st: 117174- ■HuGene st

LOXLl 244262-HuGene_st:240421-HuGene_st:999775- LOXLl 0.0027 1.4045 1.33 75.963.7- HuGene st 85.5

TABLE 9

BIBLIOGRAPHY

Affymetrix. GeneChip expression data analysis fundamentals. Affymetrix, Santa Clara, CA USA, 2001.

Alon et al, Proc. Natl. Acad. ScL USA: 96, 6745-6750, June 1999

Ausubel, F. et al, "Current Protocols in Molecular Biology", John Wiley & Sons, (1998)

Bonner et al (1973) J. MoI. Biol. 81 : 123

DeRisi, et al, Nature Genetics 14:457-460 (1996)

Germer e/ α/., Genome Res. 10:258-266 (2000)

Guo et al., Nucleic Acids Res. 22:5456-5465 (1994)

Heid et al, Genome Res. 6:986-994 (1996)

Hubbell E.W., W. M. Liu, and R. Mei. Robust estimators for expression analysis. Bioinformatics, 18:1585-1592, 2002.

Irizarry R.W., B. M. Bolstad, F. Collin, L. M. Cope, B. Hobbs, and T. P. Speed. Summaries of affymetrix genechip probe level data. Nucleic Acid Research, 31, 2003.

Kraus, M. and Aaronson, S., 1991. Methods Enzymol, 200:546-556

Maskos and Southern, Nuc. Acids Res. 20:1679-84, 1992

Moore et al, BBA, 1402:239-249, 1988

Nielsen (1999) Curr. Opin. Biotechnol 10:71-75

Nielsen et al. (1991) Science 254: 1497-1500

Pease et al, Proc. Natl. Acad. ScL USA 91(11):5022-5026 (1994)

Pevzner et al, J. Biomol Struc. & Dyn. 9:399-410, 1991

Schena, et al Science 270:467-470 (1995)

Smith et al, Science 258:1122-1126 (1992)

Smyth G.K. Bioinformatics and Computational Biology Solutions using R and Bioconductor. Springer, New York, 2005.

Smyth G.K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Statistical Applications in Genetics and Molecular Biology, 3(l):Article 3, 2004.

T. Sano and C. R. Cantor, Bio/Technology 9: 1378-81 (1991) Urdea et al, Nucleic Acids Symp. Ser., 24: 197-200 (1991) Wedemeyer et al., Clinical Chemistry 48:9 1398-1405, 2002) Weissleder et al., Nature Medicine 6:351-355, 2000