GENE EXPRESSION IN BIOLOGICAL CONDITIONS

Information

  • Patent Application
  • 20090170097
  • Publication Number
    20090170097
  • Date Filed
    July 25, 2008
    16 years ago
  • Date Published
    July 02, 2009
    15 years ago
Abstract
The present invention relates to a method of predicting the prognosis of a biological condition in animal tissue, wherein the expression of genes is examined and correlated to standards. The invention further relates to the treatment of the biological condition and an assay for predicting the prognosis. In particular, the invention concerns gene expression in epithelial tissue, such as urinary bladder under both normal and abnormal conditions.
Description
TECHNICAL FIELD OF THE INVENTION

The present invention relates to a method of predicting the prognosis of a biological condition in animal tissue, wherein the expression of genes is examined and correlated to standards. The invention further relates to the treatment of the biological condition and an assay for predicting the prognosis.


BACKGROUND

The building of large databases containing human genome sequences is the basis for studies of gene expressions in various tissues during normal physiological and pathological conditions. Constantly (constitutively) expressed sequences as well as sequences whose expression is altered during disease processes are important for our understanding of cellular properties, and for the identification of candidate genes for future therapeutic intervention. As the number of known genes and ESTs build up in the databases, array-based simultaneous screening of thousands of genes is necessary to obtain a profile of transcriptional behaviour, and to identify key genes that either alone or in combination with other genes, control various aspects of cellular life. One cellular behaviour that has been a mystery for many years is the malignant behaviour of cancer cells. It is now known that for example defects in DNA repair can lead to cancer but the cancer-creating mechanism in heterozygous individuals is still largely unknown as is the malignant cell's ability to repeat cell cycles to avoid apoptosis to escape the immune system to invade and metastasize and to escape therapy. There are indications in these areas and excellent progress has been made, both the myriad of genes interacting with each other in a highly complex multidimensional network is making the road to insight long and contorted.


Similar appearing tumors—morphologically, histochemically, microscopically—can be profoundly different. They can have different invasive and metastasizing properties, as well as respond differently to therapy. There is thus a need in the art for methods which distinguish tumors and tissues on factors different than those currently in clinical use. The malignant transformation from normal tissue to cancer is believed to be a multistep process, in which tumorsuppressor genes, that normally repress cancer growth show reduced gene expression and in which other genes that encode tumor promoting proteins (oncogenes) show an increased expression level. Several tumor suppressor genes have been identified up till now, as e.g. p16, Rb, p53 (Nesrin Özören and Wafik S. El-Deiry, Introduction to cancer genes and growth control, In: DNA alterations in cancer, genetic and epigenetic changes, Eaton publishing, Melanie Ehrlich (ed) p. 1-43, 2000; and references therein). They are usually identified by their lack of expression or their mutation in cancer tissue.


Other examinations have shown this downregulation of transcripts to be partly due to loss of genomic material (loss of heterozygosity), partly to methylation of promotorregions, and partly due to unknown factors (Nesrin Özören and Wafik S. El-Deiry, Introduction to cancer genes and growth control, In: DNA alterations in cancer, genetic and epigenetic changes, Eaton publishing, Melanie Ehrlich (ed) p. 1-43, 2000; and references therein).


Several oncogenes are known, e.g. cyclinD1/PRAD1/BCL1, FGFs, c-MYC, BCL-2 all of which are genes that are amplified in cancer showing an increased level of transcript (Nesrin Özören and Wafik S. El-Deiry, Introduction to cancer genes and growth control, In: DNA alterations in cancer, genetic and epigenetic changes, Eaton publishing, Melanie Ehrlich (ed) p. 1-43, 2000; and references therein). Many of these genes are related to cell growth and directs the tumor cells to uninhibited growth. Others may be related to tissue degradation as they e.g. encode enzymes that break down the surrounding connective tissue.


Bladder cancer is the fourth most common malignancy in males in the western countries (Pisani). The disease basically takes two different courses: one where patients have multiple recurrences of superficial tumors (Ta and T1), and one where the disease from the beginning is muscle invasive (T2+) and leads to metastasis. About 5-10% of patients with Ta tumors and 20-30% of the patients with T1 tumors will eventually develop a higher stage tumor (Wolf). Patients with superficial bladder tumors represent 75% of all bladder cancer patients and no clinical useful markers identifying patients with a poor prognosis exists at present.


The patients presenting isolated or concomitant Carcinoma in situ (CIS) lesions have a high risk of disease progression to a muscle invasive stage (Althausen). The CIS lesions may have a widespread manifestation in the bladder (field disease) and are believed to be the most common precursors of invasive carcinomas (Spruck, Rosin). The ability to predict which tumours are likely to recur or progress would have great impact on the clinical management of patients with superficial disease, as it would be possible to treat high-risk patients more aggressively (e.g. radical cystectomy or adjuvant therapy). This approach is currently not possible, as no clinical useful markers exist that identify these patients.


Although many prognostic markers have been investigated, the most important prognostic factors are still disease stage, dysplasia grade and especially the presence of areas with CIS (Anderstrom, Cummings, Cheng). The gold standard for detection of CIS is urine cytology and histopathologic analysis of a set of selected site biopsies removed during routine cytsocopy examinations; however these procedures are not sufficient sensitive.


Implementing routine cytoscopy examinations with 5-ALA fluorescence imaging of the tumours and pre-cancerous lesions (CIS lesions and moderate dysplasia lesions) may increase the sensitivity of the procedure (Kriegmar), however, increased detection sensitivity is still necessary in order to offer better treatment regiments to the individual patients.


SUMMARY OF THE INVENTION

The present invention relates to prediction of prognosis of a biological condition, in particular to the prognosis of cancer such as bladder cancer. It is known that individuals suffering from cancer, although their tumors macroscopically and microscopically are identical, may have very different outcome. The present inventors have identified new predictor genes to classify macroscopically and microscopically identical tumors into two or more groups, wherein in each group has a separate risk profile of recurrence, invasive growth, metastasis etc. as compared to the other group(s). The present invention relates to genotyping of the tissue, and correlating the result to standard expression level(s) to predict the prognosis of the biological condition.


Accordingly, in one aspect the present invention relates to a method of predicting the prognosis of a biological condition in animal tissue,

    • comprising collecting a sample comprising cells from the tissue and/or expression products from the cells,
    • determining an expression level of at least one gene in said sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562,
    • correlating the expression level to at least one standard expression level to predict the prognosis of the biological condition in the animal tissue.


The genes No. 1—gene No. 562 are found in table A described below herein.


Animal tissue may be tissue from any animal, preferably from a mammal, such as a horse, a cow, a dog, a cat, and more preferably the tissue is human tissue. The biological condition may be any condition exhibiting gene expression different from normal tissue. In particular the biological condition relates to a malignant or premalignant condition, such as a tumor or cancer, in particular bladder cancer. By the term “collecting a sample comprising cells” is meant the sample is provided in a manner, so that the expression level of the genes may be determined.


Furthermore, the invention relates to a method of determining the stage of a biological condition in animal tissue,

    • comprising collecting a sample comprising cells from the tissue,
    • determining an expression level of at least one gene in said sample, said gene being selected from the group of genes consisting of gene No 1 to gene No. 562,
    • correlating the expression level of the assessed genes to at least one standard level of expression determining the stage of the condition.


The determination of the stage of the biological condition may be conducted prior to the method of predicting the method, or the stage of the biological condition may as such contain the information about the prognosis.


The methods above may be used for determining single gene expressions, however the invention also relates to a method of determining an expression pattern of a bladder cell sample, comprising:

    • collecting sample comprising bladder cells and/or expression products from bladder cells,
    • determining the expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562, and obtaining an expression pattern of the bladder cell sample.


Further, the invention relates to a method of determining an expression pattern of a bladder cell sample independent of the proportion of submucosal, muscle, or connective tissue cells present, comprising:

    • determining the expression of one or more genes in a sample comprising cells, wherein the one or more genes exclude genes which are expressed in the submucosal, muscle, or connective tissue, whereby a pattern of expression is formed for the sample which is independent of the proportion of submucosal, muscle, or connective tissue cells in the sample.


The expression pattern may be used in a method according to this information, and accordingly, the invention also relates to a method of predicting the prognosis a biological condition in human bladder tissue comprising, collecting a sample comprising cells from the tissue, determining an expression pattern of the cells as defined in any of claims 43-54, correlating the determined expression pattern to a standard pattern, predicting the prognosis of the biological condition of said tissue as well as a method for determining the stage of a biological condition in animal tissue, comprising collecting a sample comprising cells from the tissue, determining an expression pattern of the cells as defined above, correlating the determined expression pattern to a standard pattern, determining the stage of the biological condition is said tissue.


The invention further relates to a method for reducing cell tumorigenicity or malignancy of a cell, said method comprising contacting a tumor cell with at least one peptide expressed by at least one gene selected from the group of genes consisting of gene Nos. 200-214, 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, 446, 453, 460, 461, 463, 464, 465, 466, 467, 469, 470, 471, 472, 473, 475, 476, 477, 479, 480, 481, 482, 483, 485, 486, 487, 488, 490, 492, 494, 496, 497, 498, 499, 503, 515, 516, 517, 521, 526, 527, 528, 530, 532, 533, 537, 539, 540, 541, 542, 543, 545, 554, 557, 560 or obtaining at least one gene selected from the group of genes consisting of gene Nos 200-214, 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, 446, 453, 460, 461, 463, 464, 465, 466, 467, 469, 470, 471, 472, 473, 475, 476, 477, 479, 480, 481, 482, 483, 485, 486, 487, 488, 490, 492, 494, 496, 497, 498, 499, 503, 515, 516, 517, 521, 526, 527, 528, 530, 532, 533, 537, 539, 540, 541, 542, 543, 545, 554, 557, 560, and introducing said at least one gene into the tumor cell in a manner allowing expression of said gene(s), or


obtaining at least one nucleotide probe capable of hybridising with at least one gene of a tumor cell, said at least one gene being selected from the group of genes consisting of gene Nos. 1-199, 215-232, 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 369, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, 444, 447, 448, 449, 450, 451, 452, 454, 455, 456, 457, 458, 459, 462, 468, 474, 478, 484, 489, 491, 493, 495, 500, 501, 502, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 518, 519, 520, 522, 523, 524, 525, 529, 531, 534, 535, 536, 538, 544, 546, 547, 548, 549, 550, 551, 552, 553, 555, 556, 558, 559, 561, 562, and introducing said at least one nucleotide probe into the tumor cell in a manner allowing the probe to hybridise to the at least one gene, thereby inhibiting expression of said at least one gene.


In a further aspect the invention relates to a method for producing antibodies against an expression product of a cell from a biological tissue, said method comprising the steps of


obtaining expression product(s) from at least one gene said gene being expressed as defined above,


immunising a mammal with said expression product(s) obtaining antibodies against the expression product.


The antibodies produced may be used for producing a pharmaceutical composition. Further, the invention relates to a vaccine capable of eliciting an immune response against at least one expression product from at least one gene said gene being expressed as defined above.


The invention furthermore relates to the use of any of the methods discussed above for producing an assay for diagnosing a biological condition in animal tissue.


Also, the invention relates to the use of a peptide as defined above as an expression product and/or the use of a gene as defined above and/or the use of a probe as defined above for preparation of a pharmaceutical composition for the treatment of a biological condition in animal tissue.


In yet a further aspect the invention relates to an assay for determining the presence or absence of a biological condition in animal tissue, comprising

    • at least one first marker capable of detecting an expression level of at least one gene selected from the group of genes consisting of gene No. 1 to gene No. 562,


In another aspect the invention relates to an assay for determining an expression pattern of a bladder cell, comprising at least a first marker and/or a second marker, wherein the first marker is capable of detecting a gene from a first gene group as defined above, and the second marker is capable of detecting a gene from a second gene group as defined above.





DRAWINGS

Description of figures:



FIG. 1 Hierarchical cluster analysis of tumor samples based on 3,197 genes that show large variation across all tumor samples. Samples with progression are marked Prog.



FIG. 2 Delineation of the 200 best marker genes. Genes that show higher levels of expression in the non-progression group are shown in the top and genes that show higher levels of expression in the progression group is shown in the bottom. Each column in the diagram represents a tumor sample and each row a gene. The 13 non-progressing samples are shown to the left and the 16 progressing samples are shown to the right in the diagram. The color saturation indicates differences in gene expression across the tumor samples; light color indicates up regulation compared the median expression and down regulation compared to the median expression of the gene is shown in dark color. Gene names of particular interesting genes are listed. Notable, non-group expression patterns were observed for two tumors (arrows). The tumor in the no progression group (150-6) showed a solid growth pattern, which is associated with a poor prognosis. No special tumor characteristics can help explain the gene expression pattern observed for the tumor in the progression group (825-3).



FIG. 3. Cross-validation performance using from 1 to 200 genes.



FIG. 4. Predicting progression in early stage bladder tumors. a, The 45-gene expression signature found to be optimal for progression prediction. Genes showing high expression in progressing samples are show in the top and genes showing high expression in the non-progressing samples are shown in the bottom. Genes are listed according to how many cross-validation loops included the genes. b, The 45-gene expression signature in the 19 tumor test-set. The samples are listed according to the correlation to the average non-progression signature from the training set samples. The read punctuated line separates samples with positive (left) and negative (right) correlation values. The white lines separates samples above and below the correlation cutoff values of 0.1 and −0.1. The sample legend indicates no-progression (N) samples and progression (P) samples.



FIG. 5 Hierarchical cluster analysis of the metachronous tumor samples. Tight clustering tumors of different stage from the same patients are colored in grey.



FIG. 6 Two-way hierarchical clustering and multidimensional scaling analysis of gene expression data from 40 bladder tumour biopsies. a, Tumour cluster dendrogram based on the 1767 gene-set. CIS annotations following the sample names indicate concomitant carcinoma in situ. Tumour recurrence rates are shown to the right of the dendrogram as + and ++ indicating moderate and high recurrence rates, respectively, while no sign indicates no or moderate recurrence. b, Tumour cluster dendrogram based on 88 cancer related genes. c, 2D plot of multidimensional scaling analysis of the 40 tumours based on the 1767 gene-set. The colour code identifies the tumour samples from the cluster dendrogram (FIG. 1a). d, Two-way cluster analysis diagram of the 1767 gene-set. Each row in the diagram represents a gene and each column a tumour sample. The colour saturation represents differences in gene expression across the tumour samples; light color indicates higher expression of the gene compared to the median expression and lower expression of the gene compared to the median expression shown in dark color. The colour intensities indicate degrees of gene-regulation. The sidebars to the right of the diagram represent gene clusters a-j and normal 1-3 in the left side indicate the three normal biopsies and normal 4 indicates the pool of biopsies from 37 patients.



FIG. 7 Enlarged view of the gene clusters a, c, f, and g. The dendrogram at the top is identical to FIG. 6a. a, Cluster of transcription factors and other nuclear associated genes. c, Cluster of genes involved in proliferation and cell cycle control. f, Gene expression pattern and corresponding area with squamous metaplasia in urothelial carcinoma. The light colour indicates genes up-regulated in samples 1178-1 and 875-1, the only two samples with squamous cell metaplasia. g, Cluster of genes involved in angiogenesis and matrix remodelling.



FIG. 8. Hierarchical Cluster Analysis Results


Here we show expanded views of clusters a-j as identified in the 1767 gene-cluster. The tumour cluster dendrogram and colour bars on top of the clusters represents the same tumour cluster as shown in the paper. The four samples to the left are normal biopsies (normal 1-3) and a pool of 37 normal biopsies (normal 4).



FIG. 8
a. Molecular classification of tumour samples using 80 predictive genes in each cross-validation loop. Each classification is based on the closeness to the mean in the three classes. Samples marked with * were not used to build the classifier. The scale indicates the distance from the samples to the classes in the classifier, measured in weighted squared Euclidean distance.



FIG. 9 Number of classification errors vs. number of genes used in cross-validation loops.



FIG. 10 Expression profiles of the 71 genes used in the final classifier model. The tumors shown are the 33 tumors used in the cross validation scheme. The Ta tumors are shown to the left, the T1 tumors in the middle, and the T2 tumors to the right.



FIG. 11 Number of prediction errors vs. number of genes used in cross-validation loops.



FIG. 12 The expression profiles of the 26 genes that constitute our final prediction model. The genes are listed according to the degree of correlation with the recurrence and non-recurrence groups. Genes with highest correlations are found in the top and the bottom of the list.



FIG. 13. Hierarchical cluster analysis of the gene expression in 41 TCC, 9 normal samples and 10 samples from cystectomy specimens with CIS lesions. a, Cluster dendrogram of all 41 TCC biopsies based on the expression of 5,491 genes. b, Cluster dendrogram of all superficial TCC biopsies based on the expression of 5,252 genes. c, Two-way cluster analysis diagram of the 41 TCC biopsies together with gene expressions in the normal and cystectomy samples (left columns). Each row represents a gene and each column represent a biopsy sample. Yellow indicates up-regulation compared to the median expression (black) of the gene and blue indicates down-regulation compared to the median expression. The colour saturation indicates degree of gene regulation. The sidebars to the right of the diagram represent gene-clusters 1-4; enlarged views of cluster 1 and 4 are shown to the right, with all gene symbols listed.



FIG. 14. Delineation of the 100 best markers that separate TCC without CIS from TCC with concomitant CIS. a, The 50 best up-regulated marker genes in TCC without CIS are shown in the top and the 50 best up-regulated marker genes in TCC with CIS are shown in the bottom. The gene symbols are listed to the right of the diagram. b, Expression profiles of the 100 marker genes in 9 normal biopsies (left column), 5 histologically normal samples adjacent to CIS lesions (middle column), and 5 biopsies with CIS lesions detected. (right column).



FIG. 15 Cross validation performance using all samples



FIG. 16 Expression profiles of the 16 genes in the CIS classifier. a, the expression of the 16 classifier genes in TCC with no surrounding CIS (left) and in TCC with surrounding CIS (right). The gene symbols of the classifier genes are listed together with the number of the times used in cross-validation loops. b, the expression of the 16 classifier genes in normal samples, in histologically normal samples adjacent to CIS lesions, and in biopsies with CIS lesions. The top dendrogram shows the sample clustering from hierarchical cluster analysis based on the 16 classifier genes. The genes appear in the same order as in 3a.



FIG. 17 Cross validation performance using half of the samples



FIG. 18 shows table B



FIG. 19 shows table C



FIG. 20 shows table D



FIG. 21 shows table E



FIG. 22 shows table F



FIG. 23 shows table G



FIG. 24 shows table H





DETAILED DESCRIPTION OF THE INVENTION

As discussed above the present invention relates to the finding that it is possible to predict the prognosis of a biological condition by determining the expression level of one or more genes from a specified group of genes and comparing the expression level to at least one standard for expression levels. The present inventors have identified 562 genes relevant for predicting the prognosis of a biological condition, in particular a cancer disease, such as bladder cancer.


The following table A shows the genes relevant in this context. Whenever a gene is cited herein with reference to a gene No. the numbering refers to the genes of Table A.















TABLE A








Unigene





Gene #
GeneChip
Probeset
Build
Unigene
description
Classifier





















1
HUGeneFL
AB000220_at
168
Hs.171921
sema domain, immunoglobulin domain
stage







(Ig), short basic domain, secreted,







(semaphorin) 3C


2
HUGeneFL
AF000231_at
168
Hs.75618
RAB11A, member RAS oncogene
stage







family


3
HUGeneFL
D10922_s_at
168
Hs.99855
formyl peptide receptor-like 1
stage


4
HUGeneFL
D10925_at
168
Hs.301921
chemokine (C-C motif) receptor 1
stage


5
HUGeneFL
D11086_at
168
Hs.84
interleukin 2 receptor, gamma (severe
stage







combined immunodeficiency)


6
HUGeneFL
D11151_at
168
Hs.211202
endothelin receptor type A
stage


7
HUGeneFL
D13435_at
168
Hs.426142
phosphatidylinositol glycan, class F
stage


8
HUGeneFL
D13666_s_at
168
Hs.136348
osteoblast specific factor 2 (fasciclin I-
stage







like)


9
HUGeneFL
D14520_at
168
Hs.84728
Kruppel-like factor 5 (intestinal)
stage


10
HUGeneFL
D21878_at
168
Hs.169998
bone marrow stromal cell antigen 1
stage


11
HUGeneFL
D26443_at
168
Hs.371369
solute carrier family 1 (glial high affinity
stage







glutamate transporter), member 3


12
HUGeneFL
D42046_at
168
Hs.194665
DNA2 DNA replication helicase 2-like
stage







(yeast)


13
HUGeneFL
D45370_at
168
Hs.74120
adipose specific 2
stage


14
HUGeneFL
D49372_s_at
168
Hs.54460
chemokine (C-C motif) ligand 11
stage


15
HUGeneFL
D50495_at
168
Hs.224397
transcription elongation factor A (SII), 2
stage


16
HUGeneFL
D63135_at
168
Hs.27935
tweety homolog 2 (Drosophila)
stage


17
HUGeneFL
D64053_at
168
Hs.198288
protein tyrosine phosphatase, receptor
stage







type, R


18
HUGeneFL
D83920_at
168
Hs.440898
ficolin (collagen/fibrinogen domain
stage







containing) 1


19
HUGeneFL
D85131_s_at
168
Hs.433881
MYC-associated zinc finger protein
stage







(purine-binding transcription factor)


20
HUGeneFL
D86062_s_at
168
Hs.413482
chromosome 21 open reading frame
stage







33


21
HUGeneFL
D86479_at
168
Hs.439463
AE binding protein 1
stage


22
HUGeneFL
D86957_at
168
Hs.307944
likely ortholog of mouse septin 8
stage


23
HUGeneFL
D86959_at
168
Hs.105751
Ste20-related serine/threonine kinase
stage


24
HUGeneFL
D86976_at
168
Hs.196914
minor histocompatibility antigen HA-1
stage


25
HUGeneFL
D87433_at
168
Hs.301989
stabilin 1
stage


26
HUGeneFL
D87443_at
168
Hs.409862
sorting nexin 19
stage


27
HUGeneFL
D87682_at
168
Hs.134792
KIAA0241 protein
stage


28
HUGeneFL
D89077_at
168
Hs.75367
Src-like-adaptor
stage


29
HUGeneFL
D89377_at
168
Hs.89404
msh homeo box homolog 2 (Drosophila)
stage


30
HUGeneFL
D90279_s_at
168
Hs.433695
collagen, type V, alpha 1
stage


31
HUGeneFL
HG1996-HT2044_at
168


stage


32
HUGeneFL
HG2090-HT2152_s_at
168


stage


33
HUGeneFL
HG2463-HT2559_at
168


stage


34
HUGeneFL
HG3044-HT3742_s_at
168


stage


35
HUGeneFL
HG3187-HT3366_s_at
168


stage


36
HUGeneFL
HG3342-HT3519_s_at
168


stage


37
HUGeneFL
HG371-HT26388_s_at
168


stage


38
HUGeneFL
HG4069-HT4339_s_at
168


stage


39
HUGeneFL
HG67-HT67_f_at
168


stage


40
HUGeneFL
HG907-HT907_at
168


stage


41
HUGeneFL
J02871_s_at
168
Hs.436317
cytochrome P450, family 4, subfamily
stage







B, polypeptide 1


42
HUGeneFL
J03040_at
168
Hs.111779
secreted protein, acidic, cysteine-rich
stage







(osteonectin)


43
HUGeneFL
J03060_at
168


stage


44
HUGeneFL
J03068_at
168


stage


45
HUGeneFL
J03241_s_at
168
Hs.2025
transforming growth factor, beta 3
stage


46
HUGeneFL
J03278_at
168
Hs.307783
platelet-derived growth factor receptor,
stage







beta polypeptide


47
HUGeneFL
J03909_at
168


stage


48
HUGeneFL
J03925_at
168
Hs.172631
integrin, alpha M (complement component
stage







receptor 3, alpha; also known as







CD11b (p170), macrophage antigen







alpha polypeptide)


49
HUGeneFL
J04056_at
168
Hs.88778
carbonyl reductase 1
stage


50
HUGeneFL
J04058_at
168
Hs.169919
electron-transfer-flavoprotein, alpha
stage







polypeptide (glutaric aciduria II)


51
HUGeneFL
J04130_s_at
168
Hs.75703
chemokine (C-C motif) ligand 4
stage


52
HUGeneFL
J04152_rna1_s_at
168


stage


53
HUGeneFL
J04162_at
168
Hs.372679
Fc fragment of IgG, low affinity IIIa,
stage







receptor for (CD16)


54
HUGeneFL
J04456_at
168
Hs.407909
lectin, galactoside-binding, soluble, 1
stage







(galectin 1)


55
HUGeneFL
J05032_at
168
Hs.32393
aspartyl-tRNA synthetase
stage


56
HUGeneFL
J05070_at
168
Hs.151738
matrix metalloproteinase 9 (gelatinase
stage







B, 92 kDa gelatinase, 92 kDa type IV







collagenase)


57
HUGeneFL
J05448_at
168
Hs.79402
polymerase (RNA) II (DNA directed)
stage







polypeptide C, 33 kDa


58
HUGeneFL
K01396_at
168
Hs.297681
serine (or cysteine) proteinase inhibitor,
stage







clade A (alpha-1 antiproteinase,







antitrypsin), member 1


59
HUGeneFL
K03430_at
168


stage


60
HUGeneFL
L06797_s_at
168
Hs.421986
chemokine (C—X—C motif) receptor 4
stage


61
HUGeneFL
L10343_at
168
Hs.112341
protease inhibitor 3, skin-derived
stage







(SKALP)


62
HUGeneFL
L13391_at
168
Hs.78944
regulator of G-protein signalling 2,
stage







24 kDa


63
HUGeneFL
L13698_at
168
Hs.65029
growth arrest-specific 1
stage


64
HUGeneFL
L13720_at
168
Hs.437710
growth arrest-specific 6
stage


65
HUGeneFL
L13923_at
168
Hs.750
fibrillin 1 (Marfan syndrome)
stage


66
HUGeneFL
L15409_at
168
Hs.421597
von Hippel-Lindau syndrome
stage


67
HUGeneFL
L17325_at
168
Hs.195825
RNA binding protein with multiple
stage







splicing


68
HUGeneFL
L19872_at
168
Hs.170087
aryl hydrocarbon receptor
stage


69
HUGeneFL
L27476_at
168
Hs.75608
tight junction protein 2 (zona occludens
stage







2)


70
HUGeneFL
L33799_at
168
Hs.202097
procollagen C-endopeptidase enhancer
stage


71
HUGeneFL
L40388_at
168
Hs.30212
thyroid receptor interacting protein 15
stage


72
HUGeneFL
L40904_at
168
Hs.387667
peroxisome proliferative activated
stage







receptor, gamma


73
HUGeneFL
L41919_rna1_at
168


stage


74
HUGeneFL
M11433_at
168
Hs.101850
retinol binding protein 1, cellular
stage


75
HUGeneFL
M11718_at
168
Hs.283393
collagen, type V, alpha 2
stage


76
HUGeneFL
M12125_at
168
Hs.300772
tropomyosin 2 (beta)
stage


77
HUGeneFL
M14218_at
168
Hs.442047
argininosuccinate lyase
stage


78
HUGeneFL
M15395_at
168
Hs.375957
integrin, beta 2 (antigen CD18 (p95),
stage







lymphocyte function-associated antigen







1; macrophage antigen 1 (mac-1)







beta subunit)


79
HUGeneFL
M16591_s_at
168
Hs.89555
hemopoietic cell kinase
stage


80
HUGeneFL
M17219_at
168
Hs.203862
guanine nucleotide binding protein (G
stage







protein), alpha inhibiting activity poly-







peptide 1


81
HUGeneFL
M20530_at
168


stage


82
HUGeneFL
M23178_s_at
168
Hs.73817
chemokine (C-C motif) ligand 3
stage


83
HUGeneFL
M28130_rna1_s_at
168


stage


84
HUGeneFL
M29550_at
168
Hs.187543
protein phosphatase 3 (formerly 2B),
stage







catalytic subunit, beta isoform (calcineurin







A beta)


85
HUGeneFL
M31165_at
168
Hs.407546
tumor necrosis factor, alpha-induced
stage







protein 6


86
HUGeneFL
M32011_at
168
Hs.949
neutrophil cytosolic factor 2 (65 kDa,
stage







chronic granulomatous disease, autosomal







2)


87
HUGeneFL
M33195_at
168
Hs.433300
Fc fragment of IgE, high affinity I,
stage







receptor for; gamma polypeptide


88
HUGeneFL
M37033_at
168
Hs.443057
CD53 antigen
stage


89
HUGeneFL
M37766_at
168
Hs.901
CD48 antigen (B-cell membrane
stage







protein)


90
HUGeneFL
M55998_s_at
168
Hs.172928
collagen, type I, alpha 1
stage


91
HUGeneFL
M57731_s_at
168
Hs.75765
chemokine (C—X—C motif) ligand 2
stage


92
HUGeneFL
M62840_at
168
Hs.82542
acyloxyacyl hydrolase (neutrophil)
stage


93
HUGeneFL
M63262_at
168


stage


94
HUGeneFL
M68840_at
168
Hs.183109
monoamine oxidase A
stage


95
HUGeneFL
M69203_s_at
168
Hs.75703
chemokine (C-C motif) ligand 4
stage


96
HUGeneFL
M72885_rna1_s_at
168


stage


97
HUGeneFL
M77349_at
168
Hs.421496
transforming growth factor, beta-
stage







induced, 68 kDa


98
HUGeneFL
M82882_at
168
Hs.124030
E74-like factor 1 (ets domain transcription
stage







factor)


99
HUGeneFL
M83822_at
168
Hs.209846
LPS-responsive vesicle trafficking,
stage







beach and anchor containing


100
HUGeneFL
M92934_at
168
Hs.410037
connective tissue growth factor
stage


101
HUGeneFL
M95178_at
168
Hs.119000
actinin, alpha 1
stage


102
HUGeneFL
S69115_at
168
Hs.10306
natural killer cell group 7 sequence
stage


103
HUGeneFL
S77393_at
168
Hs.145754
kruppel-like factor 3 (basic)
stage


104
HUGeneFL
S78187_at
168
Hs.153752
cell division cycle 25B
stage


105
HUGeneFL
U01833_at
168
Hs.81469
nucleotide binding protein 1 (MinD
stage







homolog, E. coli)


106
HUGeneFL
U07231_at
168
Hs.309763
G-rich RNA sequence binding factor 1
stage


107
HUGeneFL
U09278_at
168
Hs.436852
fibroblast activation protein, alpha
stage


108
HUGeneFL
U09937_rna1_s_at
168


stage


109
HUGeneFL
U10550_at
168
Hs.79022
GTP binding protein overexpressed in
stage







skeletal muscle


110
HUGeneFL
U12424_s_at
168
Hs.108646
glycerol-3-phosphate dehydrogenase
stage







2 (mitochondrial)


111
HUGeneFL
U16306_at
168
Hs.434488
chondroitin sulfate proteoglycan 2
stage







(versican)


112
HUGeneFL
U20158_at
168
Hs.2488
lymphocyte cytosolic protein 2 (SH2
stage







domain containing leukocyte protein of







76 kDa)


113
HUGeneFL
U20536_s_at
168
Hs.3280
caspase 6, apoptosis-related cysteine
stage







protease


114
HUGeneFL
U24266_at
168
Hs.77448
aldehyde dehydrogenase 4 family,
stage







member A1


115
HUGeneFL
U28249_at
168
Hs.301350
FXYD domain containing ion transport
stage







regulator 3


116
HUGeneFL
U28488_s_at
168
Hs.155935
complement component 3a receptor 1
stage


117
HUGeneFL
U29680_at
168
Hs.227817
BCL2-related protein A1
stage


118
HUGeneFL
U37143_at
168
Hs.152096
cytochrome P450, family 2, subfamily
stage







J, polypeptide 2


119
HUGeneFL
U38864_at
168
Hs.108139
zinc finger protein 212
stage


120
HUGeneFL
U39840_at
168
Hs.163484
forkhead box A1
stage


121
HUGeneFL
U41315_rna1_s_at
168


stage


122
HUGeneFL
U44111_at
168
Hs.42151
histamine N-methyltransferase
stage


123
HUGeneFL
U47414_at
168
Hs.13291
cyclin G2
stage


124
HUGeneFL
U49352_at
168
Hs.414754
2,4-dienoyl CoA reductase 1, mitochondrial
stage


125
HUGeneFL
U50708_at
168
Hs.1265
branched chain keto acid dehydrogenase
stage







E1, beta polypeptide (maple







syrup urine disease)


126
HUGeneFL
U52101_at
168
Hs.9999
epithelial membrane protein 3
stage


127
HUGeneFL
U59914_at
168
Hs.153863
MAD, mothers against decapentaplegic
stage







homolog 6 (Drosophila)


128
HUGeneFL
U60205_at
168
Hs.393239
sterol-C4-methyl oxidase-like
stage


129
HUGeneFL
U61981_at
168
Hs.42674
mutS homolog 3 (E. coli)
stage


130
HUGeneFL
U64520_at
168
Hs.66708
vesicle-associated membrane protein
stage







3 (cellubrevin)


131
HUGeneFL
U65093_at
168
Hs.82071
Cbp/p300-interacting transactivator,
stage







with Glu/Asp-rich carboxy-terminal







domain, 2


132
HUGeneFL
U66619_at
168
Hs.444445
SWI/SNF related, matrix associated,
stage







actin dependent regulator of chromatin,







subfamily d, member 3


133
HUGeneFL
U68019_at
168
Hs.288261
MAD, mothers against decapentaplegic
stage







homolog 3 (Drosophila)


134
HUGeneFL
U68385_at
168
Hs.380923
likely ortholog of mouse myeloid ecotropic
stage







viral integration site-related gene 2


135
HUGeneFL
U68485_at
168
Hs.193163
bridging integrator 1
stage


136
HUGeneFL
U74324_at
168
Hs.90875
RAB interacting factor
stage


137
HUGeneFL
U77970_at
168
Hs.321164
neuronal PAS domain protein 2
stage


138
HUGeneFL
U83303_cds2_at
168
Hs.164021
chemokine (C—X—C motif) ligand 6
stage







(granulocyte chemotactic protein 2)


139
HUGeneFL
U88871_at
168
Hs.79993
peroxisomal biogenesis factor 7
stage


140
HUGeneFL
U90549_at
168
Hs.236774
high mobility group nucleosomal binding
stage







domain 4


141
HUGeneFL
U90716_at
168
Hs.79187
coxsackie virus and adenovirus receptor
stage


142
HUGeneFL
V00594_at
168
Hs.118786
metallothionein 2A
stage


143
HUGeneFL
V00594_s_at
168
Hs.118786
metallothionein 2A
stage


144
HUGeneFL
X02761_s_at
168
Hs.418138
fibronectin 1
stage


145
HUGeneFL
X04011_at
168
Hs.88974
cytochrome b-245, beta polypeptide
stage







(chronic granulomatous disease)


146
HUGeneFL
X04085_rna1_at
168


stage


147
HUGeneFL
X07438_s_at
168


stage


148
HUGeneFL
X07743_at
168
Hs.77436
pleckstrin
stage


149
HUGeneFL
X13334_at
168
Hs.75627
CD14 antigen
stage


150
HUGeneFL
X14046_at
168
Hs.153053
CD37 antigen
stage


151
HUGeneFL
X14813_at
168
Hs.166160
acetyl-Coenzyme A acyltransferase 1
stage







(peroxisomal 3-oxoacyl-Coenzyme A







thiolase)


152
HUGeneFL
X15880_at
168
Hs.415997
collagen, type VI, alpha 1
stage


153
HUGeneFL
X15882_at
168
Hs.420269
collagen, type VI, alpha 2
stage


154
HUGeneFL
X51408_at
168
Hs.380138
chimerin (chimaerin) 1
stage


155
HUGeneFL
X53800_s_at
168
Hs.89690
chemokine (C—X—C motif) ligand 3
stage


156
HUGeneFL
X54489_rna1_at
168


stage


157
HUGeneFL
X57351_s_at
168
Hs.174195
interferon induced transmembrane
stage







protein 2 (1-8D)


158
HUGeneFL
X57579_s_at
168


stage


159
HUGeneFL
X58072_at
168
Hs.169946
GATA binding protein 3
stage


160
HUGeneFL
X62048_at
168
Hs.249441
WEE1 homolog (S. pombe)
stage


161
HUGeneFL
X64072_s_at
168
Hs.375957
integrin, beta 2 (antigen CD18 (p95),
stage







lymphocyte function-associated antigen







1; macrophage antigen 1 (mac-1)







beta subunit)


162
HUGeneFL
X65614_at
168
Hs.2962
S100 calcium binding protein P
stage


163
HUGeneFL
X66945_at
168
Hs.748
fibroblast growth factor receptor 1
stage







(fms-related tyrosine kinase 2, Pfeiffer







syndrome)


164
HUGeneFL
X67491_f_at
168
Hs.355697
glutamate dehydrogenase 1
stage


165
HUGeneFL
X68194_at
168
Hs.80919
synaptophysin-like protein
stage


166
HUGeneFL
X73882_at
168
Hs.254605
microtubule-associated protein 7
stage


167
HUGeneFL
X78520_at
168
Hs.372528
chloride channel 3
stage


168
HUGeneFL
X78549_at
168
Hs.51133
PTK6 protein tyrosine kinase 6
stage


169
HUGeneFL
X78565_at
168
Hs.98998
tenascin C (hexabrachion)
stage


170
HUGeneFL
X78669_at
168
Hs.79088
reticulocalbin 2, EF-hand calcium
stage







binding domain


171
HUGeneFL
X83618_at
168
Hs.59889
3-hydroxy-3-methylglutaryl-Coenzyme
stage







A synthase 2 (mitochondrial)


172
HUGeneFL
X84908_at
168
Hs.78060
phosphorylase kinase, beta
stage


173
HUGeneFL
X90908_at
168
Hs.147391
fatty acid binding protein 6, ileal (gastrotropin)
stage


174
HUGeneFL
X91504_at
168
Hs.389277
ADP-ribosylation factor related protein 1
stage


175
HUGeneFL
X95632_s_at
168
Hs.387906
abl-interactor 2
stage


176
HUGeneFL
X97267_rna1_s_at
168


stage


177
HUGeneFL
Y00705_at
168
Hs.407856
serine protease inhibitor, Kazal type 1
stage


178
HUGeneFL
Y00787_s_at
168
Hs.624
interleukin 8
stage


179
HUGeneFL
Y00815_at
168
Hs.75216
protein tyrosine phosphatase, receptor
stage







type, F


180
HUGeneFL
Y08374_rna1_at
168


stage


181
HUGeneFL
Z12173_at
168
Hs.334534
glucosamine (N-acetyl)-6-sulfatase
stage







(Sanfilippo disease IIID)


182
HUGeneFL
Z19554_s_at
168
Hs.435800
vimentin
stage


183
HUGeneFL
Z26491_s_at
168
Hs.240013
catechol-O-methyltransferase
stage


184
HUGeneFL
Z29331_at
168
Hs.372758
ubiquitin-conjugating enzyme E2H
stage







(UBC8 homolog, yeast)


185
HUGeneFL
Z35491_at
168
Hs.377484
BCL2-associated athanogene
stage


186
HUGeneFL
Z48199_at
168
Hs.82109
syndecan 1
stage


187
HUGeneFL
Z48605_at
168
Hs.421825
inorganic pyrophosphatase 2
stage


188
HUGeneFL
Z74615_at
168
Hs.172928
collagen, type I, alpha 1
stage


189
HUGeneFL
D87437_at
168
Hs.43660
chromosome 1 open reading frame 16
recurrence


190
HUGeneFL
L49169_at
168
Hs.75678
FBJ murine osteosarcoma viral oncogene
recurrence







homolog B


191
HUGeneFL
AF006041_at
168
Hs.336916
death-associated protein 6
recurrence


192
HUGeneFL
D83780_at
168
Hs.437991
KIAA0196 gene product
recurrence


193
HUGeneFL
D64154_at
168
Hs.90107
adhesion regulating molecule 1
recurrence


194
HUGeneFL
D21337_at
168
Hs.408
collagen, type IV, alpha 6
recurrence


195
HUGeneFL
M16938_s_at
168
Hs.820
homeo box C6
recurrence


196
HUGeneFL
D87258_at
168
Hs.75111
protease, serine, 11 (IGF binding)
recurrence


197
HUGeneFL
U58516_at
168
Hs.3745
milk fat globule-EGF factor 8 protein
recurrence


198
HUGeneFL
U45973_at
168
Hs.178347
skeletal muscle and kidney enriched
recurrence







inositol phosphatase


199
HUGeneFL
U62015_at
168
Hs.8867
cysteine-rich, angiogenic inducer, 61
recurrence


200
HUGeneFL
U94855_at
168
Hs.381255
eukaryotic translation initiation factor 3,
recurrence







subunit 5 epsilon, 47 kDa


201
HUGeneFL
L34155_at
168
Hs.83450
laminin, alpha 3
recurrence


202
HUGeneFL
U70439_s_at
168
Hs.84264
acidic (leucine-rich) nuclear phospho-
recurrence







protein 32 family, member B


203
HUGeneFL
U66702_at
168
Hs.74624
protein tyrosine phosphatase, receptor
recurrence







type, N polypeptide 2


204
HUGeneFL
HG511-HT511_at
168


recurrence


205
HUGeneFL
HG3076-HT3238_s_at
168


recurrence


206
HUGeneFL
M98528_at
168
Hs.79404
DNA segment on chromosome 4
recurrence







(unique) 234 expressed sequence


207
HUGeneFL
M63175_at
168
Hs.295137
autocrine motility factor receptor
recurrence


208
HUGeneFL
D49387_at
168
Hs.294584
leukotriene B4 12-
recurrence







hydroxydehydrogenase


209
HUGeneFL
HG1879-HT1919_at
168


recurrence


210
HUGeneFL
Z23064_at
168
Hs.380118
RNA binding motif protein, X chromosome
recurrence


211
HUGeneFL
X63469_at
168
Hs.77100
general transcription factor IIE, poly-
recurrence







peptide 2, beta 34 kDa


212
HUGeneFL
L38928_at
168
Hs.118131
5,10-methenyltetrahydrofolate synthetase
recurrence







(5-formyltetrahydrofolate







cyclo-ligase)


213
HUGeneFL
U21858_at
168
Hs.60679
TAF9 RNA polymerase II, TATA box
recurrence







binding protein (TBP)-associated







factor, 32 kDa


214
HUGeneFL
M64572_at
168
Hs.405666
protein tyrosine phosphatase, non-
recurrence







receptor type 3


215
HUGeneFL
D83657_at
168
Hs.19413
S100 calcium binding protein A12
SCC







(calgranulin C)


216
HUGeneFL
HG3945-HT4215_at
168


SCC


217
HUGeneFL
J00124_at
168


SCC


218
HUGeneFL
L05187_at
168


SCC


219
HUGeneFL
L42583_f_at
168
Hs.367762
keratin 6A
SCC


220
HUGeneFL
L42601_f_at
168
Hs.367762
keratin 6A
SCC


221
HUGeneFL
L42611_f_at
168
Hs.446417
keratin 6E
SCC


222
HUGeneFL
M19888_at
168
Hs.1076
small proline-rich protein 1B (cornifin)
SCC


223
HUGeneFL
M20030_f_at
168
Hs.505352
Human small proline rich protein (sprII)
SCC







mRNA, clone 930.


224
HUGeneFL
M21005_at
168


SCC


225
HUGeneFL
M21302_at
168
Hs.505327
Human small proline rich protein (sprII)
SCC







mRNA, clone 174N.


226
HUGeneFL
M21539_at
168
Hs.2421
small proline-rich protein 2C
SCC


227
HUGeneFL
M86757_s_at
168
Hs.112408
S100 calcium binding protein A7 (psoriasin
SCC







1)


228
HUGeneFL
S72493_s_at
168
Hs.432448
keratin 16 (focal non-epidermolytic
SCC







palmoplantar keratoderma)


229
HUGeneFL
U70981_at
168
Hs.336046
interleukin 13 receptor, alpha 2
SCC


230
HUGeneFL
V01516_f_at
168
Hs.367762
keratin 6A
SCC


231
HUGeneFL
X53065_f_at
168


SCC


232
HUGeneFL
X57766_at
168
Hs.143751
matrix metalloproteinase 11 (stromelysin
SCC







3)


233
EOS Hu03
400773
133

NM_003105*: Homo sapiens sortilin-
progression







related receptor, L(DLR class) A







repeats-containing (SORL1), mRNA.


234
EOS Hu03
400843
133

NM_003105*: Homo sapiens sortilin-
progression







related receptor, L(DLR class) A







repeats-containing (SORL1), mRNA.


235
EOS Hu03
400844
133

NM_003105*: Homo sapiens sortilin-
progression







related receptor, L(DLR class) A







repeats-containing (SORL1), mRNA.


236
EOS Hu03
400846
133

sortilin-related receptor, L(DLR class)
progression







A repeats-containing (SORL1)


237
EOS Hu03
402328
133

Target Exon
progression


238
EOS Hu03
402384
133

NM_007181*: Homo sapiens mitogen-
progression







activated protein kinase kinase kinase







kinase 1 (MAP4K1), mRNA.


239
EOS Hu03
404208
133

C6001282: gi|4504223|ref|NP_000172.1|
progression







glucuronidase, beta [Homo sapiens]







gi|114963|sp|P082


240
EOS Hu03
404606
133

Target Exon
progression


241
EOS Hu03
404826
133

Target Exon
progression


242
EOS Hu03
404875
133

NM_022819*: Homo sapiens phospholipase
progression







A2, group IIF (PLA2G2F),







mRNA. VERSION NM_020245.2 GI


243
EOS Hu03
404913
133

NM_024408*: Homo sapiens Notch
progression







(Drosophila) homolog 2 (NOTCH2),







mRNA. VERSION NM_024410.1 GI


244
EOS Hu03
404977
133

Insulin-like growth factor 2 (somatomedin
progression







A) (IGF2)


245
EOS Hu03
405036
133

NM_021628*: Homo sapiens arachidonate
progression







lipoxygenase 3 (ALOXE3),







mRNA. VERSION NM_020229.1 GI


246
EOS Hu03
405371
133

NM_005569*: Homo sapiens LIM
progression







domain kinase 2 (LIMK2), transcript







variant 2a, mRNA.


247
EOS Hu03
405667
133

Target Exon
progression


248
EOS Hu03
406002
133

Target Exon
progression


249
EOS Hu03
407955
133
Hs.9343
ESTs
progression


250
EOS Hu03
408049
133
Hs.345588
desmoplakin (DPI, DPII)
progression


251
EOS Hu03
408288
133
Hs.16886
gb: zI73d06.r1 Stratagene colon
progression







(937204) Homo sapiens cDNA clone







5′, mRNA sequence


252
EOS Hu03
409513
133
Hs.54642
methionine adenosyltransferase II,
progression







beta


253
EOS Hu03
409556
133
Hs.54941
phosphorylase kinase, alpha 2 (liver)
progression


254
EOS Hu03
409586
133
Hs.55044
DKFZP586H2123 protein
progression


255
EOS Hu03
409632
133
Hs.55279
serine (or cysteine) proteinase inhibitor,
progression







clade B (ovalbumin), member 5


256
EOS Hu03
410047
133
Hs.379753
zinc finger protein 36 (KOX 18)
progression


257
EOS Hu03
411817
133
Hs.72241
mitogen-activated protein kinase
progression







kinase 2


258
EOS Hu03
412649
133
Hs.74369
integrin, alpha 7
progression


259
EOS Hu03
412841
133
Hs.101395
hypothetical protein MGC11352
progression


260
EOS Hu03
413564
133

gb: 601146990F1 NIH_MGC_19 Homo
progression








sapiens cDNA clone 5′, mRNA sequence



261
EOS Hu03
413786
133
Hs.13500
ESTs
progression


262
EOS Hu03
413840
133
Hs.356228
RNA binding motif protein, X chromosome
progression


263
EOS Hu03
413929
133
Hs.75617
collagen, type IV, alpha 2
progression


264
EOS Hu03
414223
133
Hs.238246
hypothetical protein FLJ22479
progression


265
EOS Hu03
414732
133
Hs.77152
minichromosome maintenance deficient
progression







(S. cerevisiae) 7


266
EOS Hu03
414762
133
Hs.77257
KIAA0068 protein
progression


267
EOS Hu03
414840
133
Hs.23823
hairy/enhancer-of-split related with
progression







YRPW motif-like


268
EOS Hu03
414843
133
Hs.77492
heterogeneous nuclear ribonucleoprotein
progression







A0


269
EOS Hu03
414895
133
Hs.116278

Homo sapiens cDNA FLJ13571 fis,

progression







clone PLACE1008405


270
EOS Hu03
414907
133
Hs.77597
polo (Drosophia)-like kinase
progression


271
EOS Hu03
414918
133
Hs.72222
hypothetical protein FLJ13459
progression


272
EOS Hu03
415200
133
Hs.78202
SWI/SNF related, matrix associated,
Progression







actin dependent regulator of chromatin,







subfamily a, member 4


273
EOS Hu03
416640
133
Hs.79404
neuron-specific protein
Progression


274
EOS Hu03
416815
133
Hs.80120
UDP-N-acetyl-alpha-D-
Progression







galactosamine: polypeptide N-







acetylgalactosaminyltransferase 1







(GalNAc-T1)


275
EOS Hu03
416977
133
Hs.406103
hypothetical protein FKSG44
Progression


276
EOS Hu03
417615
133
Hs.82314
hypoxanthine phosphoribosyltransferase
Progression







1 (Lesch-Nyhan syndrome)


277
EOS Hu03
417839
133
Hs.82712
fragile X mental retardation, autosomal
Progression







homolog 1


278
EOS Hu03
417900
133
Hs.82906
CDC20 (cell division cycle 20, S. cerevisiae,
Progression







homolog)


279
EOS Hu03
417924
133
Hs.82932
cyclin D1 (PRAD1: parathyroid adenomatosis
Progression







1)


280
EOS Hu03
418127
133
Hs.83532
membrane cofactor protein (CD46,
Progression







trophoblast-lymphocyte cross-reactive







antigen)


281
EOS Hu03
418321
133
Hs.84087
KIAA0143 protein
Progression


282
EOS Hu03
418504
133
Hs.85335

Homo sapiens mRNA; cDNA

Progression







DKFZp564D1462 (from clone







DKFZp564D1462)


283
EOS Hu03
418629
133
Hs.86859
growth factor receptor-bound protein 7
Progression


284
EOS Hu03
419602
133
Hs.91521
hypothetical protein
Progression


285
EOS Hu03
419847
133
Hs.184544
Homo sapiens, clone IMAGE: 3355383,
Progression







mRNA, partial cds


286
EOS Hu03
420079
133
Hs.94896
PTD011 protein
Progression


287
EOS Hu03
420116
133
Hs.95231
FH1/FH2 domain-containing protein
Progression


288
EOS Hu03
420307
133
Hs.66219
ESTs
Progression


289
EOS Hu03
420613
133
Hs.406637
ESTs, Weakly similar to A47582 B-cell
Progression







growth factor precursor [H. sapiens]


290
EOS Hu03
420732
133
Hs.367762
ESTs
Progression


291
EOS Hu03
421026
133
Hs.101067
GCN5 (general control of amino-acid
Progression







synthesis, yeast, homolog)-like 2


292
EOS Hu03
421075
133
Hs.101474
KIAA0807 protein
Progression


293
EOS Hu03
421101
133
Hs.101840
major histocompatibility complex, class
Progression







I-like sequence


294
EOS Hu03
421186
133
Hs.270563
ESTs, Moderately similar to T12512
Progression







hypothetical protein DKFZp434G232.1







[H. sapiens]


295
EOS Hu03
421311
133
Hs.283609
hypothetical protein PRO2032 progression


296
EOS Hu03
421475
133
Hs.104640
HIV-1 inducer of short transcripts
progression







binding protein; lymphoma related







factor


297
EOS Hu03
421505
133
Hs.285641
KIAA1111 protein
progression


298
EOS Hu03
421595
133
Hs.301685
KIAA0620 protein
progression


299
EOS Hu03
421628
133
Hs.106210
hypothetical protein FLJ10813
progression


300
EOS Hu03
421649
133
Hs.106415
peroxisome proliferative activated
progression







receptor, delta


301
EOS Hu03
421733
133
Hs.1420
fibroblast growth factor receptor 3
progression







(achondroplasia, thanatophoric dwarfism)


302
EOS Hu03
421782
133
Hs.108258
actin binding protein; macrophin
progression







(microfilament and actin filament







cross-linker protein)


303
EOS Hu03
421989
133
Hs.110457
Wolf-Hirschhorn syndrome candidate 1
progression


304
EOS Hu03
422043
133
Hs.110953
retinoic acid induced 1
progression


305
EOS Hu03
422068
133
Hs.104520

Homo sapiens cDNA FLJ13694 fis,

progression







clone PLACE2000115


306
EOS Hu03
422506
133
Hs.300741
sorcin
progression


307
EOS Hu03
422913
133
Hs.121599
CGI-18 protein
progression


308
EOS Hu03
422929
133
Hs.94011
ESTs, Weakly similar to
progression







MGB4_HUMAN MELANOMA-







ASSOCIATED ANTIGEN B4







[H. sapiens]


309
EOS Hu03
422959
133
Hs.349256
paired immunoglobulin-like receptor
progression







beta


310
EOS Hu03
423138
133

gb: EST385571 MAGE resequences,
progression







MAGM Homo sapiens cDNA, mRNA







sequence


311
EOS Hu03
423185
133
Hs.380062
ornithine decarboxylase antizyme 1
progression


312
EOS Hu03
423599
133
Hs.31731
peroxiredoxin 5
progression


313
EOS Hu03
423810
133
Hs.132955
BCL2/adenovirus E1B 19 kD-
progression







interacting protein 3-like


314
EOS Hu03
423960
133
Hs.136309
SH3-containing protein SH3GLB1
progression


315
EOS Hu03
424244
133
Hs.143601
hypothetical protein hCLA-iso
progression


316
EOS Hu03
424415
133
Hs.146580
enolase 2, (gamma, neuronal)
progression


317
EOS Hu03
424909
133
Hs.153752
cell division cycle 25B
progression


318
EOS Hu03
424959
133
Hs.153937
activated p21cdc42Hs kinase
progression


319
EOS Hu03
425093
133
Hs.154525
KIAA1076 protein
progression


320
EOS Hu03
425097
133
Hs.154545
PDZ domain containing guanine nucleotide
progression







exchange factor(GEF)1


321
EOS Hu03
425205
133
Hs.155106
receptor (calcitonin) activity modifying
progression







protein 2


322
EOS Hu03
425221
133
Hs.155188
TATA box binding protein (TBP)-
progression







associated factor, RNA polymerase II,







F, 55 kD


323
EOS Hu03
425243
133
Hs.155291
KIAA0005 gene product
progression


324
EOS Hu03
425380
133
Hs.32148
AD-015 protein
progression


325
EOS Hu03
426028
133
Hs.172028
a disintegrin and metalloproteinase
progression







domain 10 (ADAM10)


326
EOS Hu03
426125
133
Hs.166994
FAT tumor suppressor (Drosophila)
progression







homolog


327
EOS Hu03
426177
133
Hs.167700

Homo sapiens cDNA FLJ10174 fis,

progression







clone HEMBA1003959


328
EOS Hu03
426252
133
Hs.28917
ESTs
progression


329
EOS Hu03
426468
133
Hs.117558
ESTs
progression


330
EOS Hu03
426469
133
Hs.363039
methylmalonate-semialdehyde dehydrogenase
progression


331
EOS Hu03
426508
133
Hs.170171
glutamate-ammonia ligase (glutamine
progression







synthase)


332
EOS Hu03
426682
133
Hs.2056
UDP glycosyltransferase 1 family,
progression







polypeptide A9


333
EOS Hu03
426799
133
Hs.303154
popeye protein 3
progression


334
EOS Hu03
426982
133
Hs.173091
ubiquitin-like 3
progression


335
EOS Hu03
427239
133
Hs.356512
ubiquitin carrier protein
progression


336
EOS Hu03
427351
133
Hs.123253
hypothetical protein FLJ22009
progression


337
EOS Hu03
427681
133
Hs.284232
tumor necrosis factor receptor super-
progression







family, member 12 (translocating







chain-association membrane protein)


338
EOS Hu03
427722
133
Hs.180479
hypothetical protein FLJ20116
progression


339
EOS Hu03
427747
133
Hs.180655
serine/threonine kinase 12
progression


340
EOS Hu03
427999
133
Hs.181369
ubiquitin fusion degradation 1-like
progression


341
EOS Hu03
428115
133
Hs.300855
KIAA0977 protein
progression


342
EOS Hu03
428284
133
Hs.183435
NM_004545: Homo sapiens NADH
progression







dehydrogenase (ubiquinone) 1 beta







subcomplex, 1 (7 kD, MNLL)







(NDUFB1), mRNA.


343
EOS Hu03
428318
133
Hs.356190
ubiquitin B
progression


344
EOS Hu03
428712
133
Hs.190452
KIAA0365 gene product
progression


345
EOS Hu03
428901
133
Hs.146668
KIAA1253 protein
progression


346
EOS Hu03
429124
133
Hs.196914
minor histocompatibility antigen HA-1
progression


347
EOS Hu03
429187
133
Hs.163872
ESTs, Weakly similar to S65657 alpha-
progression







1C-adrenergic receptor splice







form 2 [H. sapiens]


348
EOS Hu03
429311
133
Hs.198998
conserved helix-loop-helix ubiquitous
progression







kinase


349
EOS Hu03
429561
133
Hs.250646
baculoviral IAP repeat-containing 6
progression


350
EOS Hu03
429802
133
Hs.5367
ESTs, Weakly similar to I38022 hypothetical
progression







protein [H. sapiens]


351
EOS Hu03
429953
133
Hs.226581
COX15 (yeast) homolog, cytochrome c
progression







oxidase assembly protein


352
EOS Hu03
430604
133
Hs.247309
succinate-CoA ligase, GDP-forming,
progression







beta subunit


353
EOS Hu03
430677
133
Hs.359784
desmoglein 2
progression


354
EOS Hu03
430746
133
Hs.406256
ESTs
progression


355
EOS Hu03
431604
133
Hs.264190
vacuolar protein sorting 35 (yeast
progression







homolog)


356
EOS Hu03
431842
133
Hs.271473
epithelial protein up-regulated in carcinoma,
progression







membrane associated protein







17


357
EOS Hu03
431857
133
Hs.271742
ADP-ribosyltransferase (NAD; poly
progression







(ADP-ribose) polymerase)-like 3


358
EOS Hu03
432258
133
Hs.293039
ESTs
progression


359
EOS Hu03
432327
133
Hs.274363
neuroglobin
progression


360
EOS Hu03
432554
133
Hs.278411
NCK-associated protein 1
progression


361
EOS Hu03
432864
133
Hs.359682
calpastatin
progression


362
EOS Hu03
433052
133
Hs.293003
ESTs, Weakly similar to PC4259
progression







ferritin associated protein [H. sapiens]


363
EOS Hu03
433282
133
Hs.49007
hypothetical protein
progression


364
EOS Hu03
433844
133
Hs.179647

Homo sapiens cDNA FLJ12195 fis,

progression







clone MAMMA1000865


365
EOS Hu03
433914
133
Hs.112160

Homo sapiens DNA helicase homolog

progression







(PIF1) mRNA, partial cds


366
EOS Hu03
434055
133
Hs.3726
x 003 protein
progression


367
EOS Hu03
434263
133
Hs.79187
ESTs
progression


368
EOS Hu03
434547
133
Hs.106124
ESTs
progression


369
EOS Hu03
434831
133
Hs.273397
KIAA0710 gene product
progression


370
EOS Hu03
434978
133
Hs.4310
eukaryotic translation initiation factor
progression







1A


371
EOS Hu03
435158
133
Hs.65588
DAZ associated protein 1
progression


372
EOS Hu03
435320
133
Hs.117864
ESTs
progression


373
EOS Hu03
435521
133
Hs.6361
mitogen-activated protein kinase
progression







kinase 1 interacting protein 1


374
EOS Hu03
436472
133
Hs.46366
KIAA0948 protein
progression


375
EOS Hu03
436576
133
Hs.77542
ESTs
progression


376
EOS Hu03
437223
133
Hs.330716

Homo sapiens cDNA FLJ14368 fis,

progression







clone HEMBA1001122


377
EOS Hu03
437256
133
Hs.97871
Homo sapiens, clone IMAGE: 3845253,
progression







mRNA, partial cds


378
EOS Hu03
437524
133
Hs.385719
ESTs
progression


379
EOS Hu03
438013
133
Hs.15670
ESTs
progression


380
EOS Hu03
438644
133
Hs.129037
ESTs
progression


381
EOS Hu03
438818
133
Hs.30738
ESTs
progression


382
EOS Hu03
438942
133
Hs.6451
PRO0659 protein
progression


383
EOS Hu03
439010
133
Hs.75216

Homo sapiens cDNA FLJ13713 fis,

progression







clone PLACE2000398, moderately







similar to LAR PROTEIN PRECURSOR







(LEUKOCYTE ANTIGEN RELATED)







(EC 3.1.3.48)


384
EOS Hu03
439130
133
Hs.375195
ESTs
progression


385
EOS Hu03
439578
133
Hs.350547
nuclear receptor co-repressor/HDAC3
progression







complex subunit


386
EOS Hu03
439632
133
Hs.334437
hypothetical protein MGC4248
progression


387
EOS Hu03
440014
133
Hs.6856
ash2 (absent, small, or homeotic,
progression








Drosophila, homolog)-like



388
EOS Hu03
440100
133
Hs.158549
ESTs, Weakly similar to
progression







T2D3_HUMAN TRANSCRIPTION







INITIATION FACTOR TFIID 135 KDA







SUBUNIT [H. sapiens]


389
EOS Hu03
440197
133
Hs.317714
pallid (mouse) homolog, pallidin
progression


390
EOS Hu03
440357
133
Hs.20950
phospholysine phosphohistidine inorganic
progression







pyrophosphate phosphatase


391
EOS Hu03
441650
133
Hs.132545
ESTs
progression


392
EOS Hu03
442220
133
Hs.8148
selenoprotein T
progression


393
EOS Hu03
442549
133
Hs.8375
TNF receptor-associated factor 4
progression


394
EOS Hu03
443407
133
Hs.348514
ESTs, Moderately similar to 2109260A
progression







B cell growth factor [H. sapiens]


395
EOS Hu03
443471
133
Hs.398102

Homo sapiens clone FLB3442

progression







PRO0872 mRNA, complete cds


396
EOS Hu03
443679
133
Hs.9670
hypothetical protein FLJ10948
progression


397
EOS Hu03
443893
133
Hs.115472
ESTs, Weakly similar to 2004399A
progression







chromosomal protein [H. sapiens]


398
EOS Hu03
444037
133
Hs.380932
CHMP1.5 protein
progression


399
EOS Hu03
444312
133
Hs.351142
ESTs
progression


400
EOS Hu03
444336
133
Hs.10882
HMG-box containing protein 1
progression


401
EOS Hu03
444604
133
Hs.11441
chromosome 1 open reading frame 8
progression


402
EOS Hu03
445084
133
Hs.250848
hypothetical protein FLJ14761
progression


403
EOS Hu03
445462
133
Hs.288649
hypothetical protein MGC3077
progression


404
EOS Hu03
445692
133
Hs.182099
ESTs
progression


405
EOS Hu03
445831
133
Hs.13351
LanC (bacterial lantibiotic synthetase
progression







component C)-like 1


406
EOS Hu03
446556
133
Hs.15303
KIAA0349 protein
progression


407
EOS Hu03
446847
133
Hs.82845

Homo sapiens cDNA: FLJ21930 fis,

progression







clone HEP04301, highly similar to







HSU90916 Human clone 23815







mRNA sequence


408
EOS Hu03
447343
133
Hs.236894
ESTs, Highly similar to S02392 alpha-
progression







2-macroglobulin receptor precursor







[H. sapiens]


409
EOS Hu03
447400
133
Hs.18457
hypothetical protein FLJ20315
progression


410
EOS Hu03
448357
133
Hs.108923
RAB38, member RAS oncogene family
progression


411
EOS Hu03
448524
133
Hs.21356
hypothetical protein DKFZp762K2015
progression


412
EOS Hu03
448625
133
Hs.178470
hypothetical protein FLJ22662
progression


413
EOS Hu03
448780
133
Hs.267749
Human DNA sequence from clone
progression







366N23 on chromosome 6q27. Contains







two genes similar to consecutive







parts of the C. elegans UNC-93 (protein







1, C46F11.1) gene, a KIAA0173







and Tubulin-Tyrosine Ligase LIKE







gene, a Mitotic Feedback Control







Protein MADP2 H


414
EOS Hu03
448813
133
Hs.22142
cytochrome b5 reductase b5R.2
progression


415
EOS Hu03
449268
133
Hs.23412
hypothetical protein FLJ20160
progression


416
EOS Hu03
449626
133
Hs.112860
zinc finger protein 258
progression


417
EOS Hu03
450893
133
Hs.25625
hypothetical protein FLJ11323
progression


418
EOS Hu03
450997
133
Hs.35254
hypothetical protein FLB6421
progression


419
EOS Hu03
451164
133
Hs.60659
ESTs, Weakly similar to T46471 hypothetical
progression







protein DKFZp434L0130.1







[H. sapiens]


420
EOS Hu03
451225
133
Hs.57655
ESTs
progression


421
EOS Hu03
451867
133
Hs.27192
hypothetical protein dJ1057B20.2
progression


422
EOS Hu03
451970
133
Hs.211046
ESTs
progression


423
EOS Hu03
452012
133
Hs.279766
kinesin family member 4A
progression


424
EOS Hu03
452170
133
Hs.28285
patched related protein translocated in
progression







renal cancer


425
EOS Hu03
452517
133

gb: RC-BT068-130399-068 BT068
progression








Homo sapiens cDNA, mRNA sequence



426
EOS Hu03
452829
133
Hs.63368
ESTs, Weakly similar to
progression







TRHY_HUMAN TRICHOHYALI







[H. sapiens]


427
EOS Hu03
452929
133
Hs.172816
neuregulin 1
progression


428
EOS Hu03
453395
133
Hs.377915
mannosidase, alpha, class 2A, member 1
progression


429
EOS Hu03
454639
133

gb: RC2-ST0158-091099-011-d05
progression







ST0158 Homo sapiens cDNA, mRNA







sequence


430
EOS Hu03
456332
133
Hs.399939
gb: nc39d05.r1 NCI_CGAP_Pr2 Homo
progression








sapiens cDNA clone, mRNA sequence



431
EOS Hu03
457228
133
Hs.195471
Human cosmid CRI-JC2015 at
progression







D10S289 in 10sp13


432
EOS Hu03
458132
133
Hs.103267
hypothetical protein FLJ22548 similar
progression







to gene trap PAT 12


433
EOS Hu03
408688
133
Hs.152925
KIAA1268 protein
progression


434
EOS Hu03
410691
133
Hs.65450
reticulon 4
progression


435
EOS Hu03
420269
133
Hs.96264
alpha thalassemia/mental retardation
progression







syndrome X-linked (RAD54 (S. cerevisiae)







homolog)


436
EOS Hu03
422119
133
Hs.111862
KIAA0590 gene product
progression


437
EOS Hu03
422765
133
Hs.1578
baculoviral IAP repeat-containing 5
progression







(survivin)


438
EOS Hu03
422984
133
Hs.351597
ESTs
progression


439
EOS Hu03
428016
133
Hs.181461
ariadne homolog, ubiquitin-conjugating
progression







enzyme E2 binding protein, 1 (Drosophila)


440
EOS Hu03
437325
133
Hs.5548
F-box and leucine-rich repeat protein 5
progression


441
EOS Hu03
444773
133
Hs.11923
hypothetical protein DJ167A19.1
progression


442
EOS Hu03
445926
133
Hs.334826
splicing factor 3b, subunit 1, 155 kDa
progression


443
EOS Hu03
452714
133
Hs.30340
KIAA1165: likely ortholog of mouse
progression







Nedd4 WW domain-binding protein 5A


444
EOS Hu03
452866
133
Hs.268016
ESTs
progression


445
EOS Hu03
453963
133
Hs.28959
cDNA FLJ36513 fis, clone
progression







TRACH2001523


446
EOS Hu03
457329
133
Hs.359682
calpastatin
progression


447
U133A
200600_at
168
Hs.170328
NM_001910; cathepsin E isoform a
CIS







preproprotein NM_148964; cathepsin







E isoform b preproprotein


448
U133A
200762_at
168
Hs.173381
NM_019894; transmembrane protease,
CIS







serine 4 isoform 1 NM_183247;







transmembrane protease, serine 4







isoform 2


449
U133A
201088_at
168
Hs.159557
NM_000228; laminin subunit beta 3
CIS







precursor


450
U133A
201291_s_at
168
Hs.156346
NM_030570; uroplakin 3B isoform a
CIS







NM_182683; uroplakin 3B isoform c







NM_182684; uroplakin 3B isoform b


451
U133A
201560_at
168
Hs.25035
NM_005547; involucrin
CIS


452
U133A
201616_s_at
168
Hs.443811
NM_004692; NM_032727; internexin
CIS







neuronal intermediate filament protein,







alpha


453
U133A
201641_at
168
Hs.118110
NM_016233; peptidylarginine deiminase
CIS







type III


454
U133A
201744_s_at
168
Hs.406475
NM_014417; BCL2 binding component 3
CIS


455
U133A
201842_s_at
168
Hs.76224
NM_020142; NADH: ubiquinone oxidoreductase
CIS







MLRQ subunit homolog


456
U133A
201858_s_at
168
Hs.1908
NM_018058; cartilage acidic protein 1
CIS


457
U133A
201859_at
168
Hs.1908
NM_000497; cytochrome P450, sub-
CIS







family XIB (steroid 11-beta-







hydroxylase), polypeptide 1 precursor


458
U133A
202746_at
168
Hs.17109
NM_007193; annexin A10
CIS


459
U133A
202917_s_at
168
Hs.416073
NM_001958; eukaryotic translation
CIS







elongation factor 1 alpha 2


460
U133A
203009_at
168
Hs.155048
NM_005581; Lutheran blood group
CIS







(Auberger b antigen included)


461
U133A
203287_at
168
Hs.18141
NM_005581; Lutheran blood group
CIS







(Auberger b antigen included)


462
U133A
203477_at
168
Hs.409034
NM_030570; uroplakin 3B isoform a
CIS







NM_182683; uroplakin 3B isoform c







NM_182684; uroplakin 3B isoform b


463
U133A
203649_s_at
168
Hs.76422
NM_000300; phospholipase A2, group
CIS







IIA (platelets, synovial fluid)


464
U133A
203759_at
168
Hs.75268
NM_007193; annexin A10
CIS


465
U133A
203792_x_at
168
Hs.371617
NM_007144; ring finger protein 110
CIS


466
U133A
203842_s_at
168
Hs.172740
NM_014417; BCL2 binding component 3
CIS


467
U133A
203980_at
168
Hs.391561
NM_001442; fatty acid binding protein
CIS







4, adipocyte


468
U133A
204141_at
168
Hs.300701
NM_017689; hypothetical protein
CIS







FLJ20151


469
U133A
204380_s_at
168
Hs.1420
NM_007144; ring finger protein 110
CIS


470
U133A
204465_s_at
168
Hs.76888
NM_004692; NM_032727; internexin
CIS







neuronal intermediate filament protein,







alpha


471
U133A
204487_s_at
168
Hs.367809
NM_001248; ectonucleoside triphosphate
CIS







diphosphohydrolase 3


472
U133A
204508_s_at
168
Hs.279916
NM_017689; hypothetical protein
CIS







FLJ20151


473
U133A
204540_at
168
Hs.433839
NM_001958; eukaryotic translation
CIS







elongation factor 1 alpha 2


474
U133A
204688_at
168
Hs.409798
NM_016233; peptidylarginine deiminase
CIS







type III


475
U133A
204952_at
168
Hs.377028
NM_000445; plectin 1, intermediate
CIS







filament binding protein 500 kDa


476
U133A
204990_s_at
168
Hs.85266
NM_000213; integrin, beta 4
CIS


477
U133A
205073_at
168
Hs.152096
NM_019894; transmembrane protease,
CIS







serine 4 isoform 1 NM_183247;







transmembrane protease, serine 4







isoform 2


478
U133A
205382_s_at
168
Hs.155597
NM_000213; integrin, beta 4
CIS


479
U133A
205453_at
168
Hs.290432
NM_002145; homeo box B2
CIS


480
U133A
205455_at
168
Hs.2942
NM_006760; uroplakin 2
CIS


481
U133A
205927_s_at
168
Hs.1355
NM_001910; cathepsin E isoform a
CIS







preproprotein NM_148964; cathepsin







E isoform b preproprotein


482
U133A
206122_at
168
Hs.95582
NM_006942; SRY-box 15
CIS


483
U133A
206191_at
168
Hs.47042
NM_001248; ectonucleoside triphosphate
CIS







diphosphohydrolase 3


484
U133A
206392_s_at
168
Hs.82547
NM_005522; homeobox A1 protein
CIS







isoform a NM_153620; homeobox A1







protein isoform b


485
U133A
206393_at
168
Hs.83760
NM_003282; troponin I, skeletal, fast
CIS


486
U133A
206465_at
168
Hs.277543
NM_015162; lipidosin
CIS


487
U133A
206561_s_at
168
Hs.116724
NM_015162; lipidosin
CIS


488
U133A
206658_at
168
Hs.284211
NM_030570; uroplakin 3B isoform a
CIS







NM_182683; uroplakin 3B isoform c







NM_182684; uroplakin 3B isoform b


489
U133A
207173_x_at
168
Hs.443435
NM_000213; integrin, beta 4
CIS


490
U133A
207862_at
168
Hs.379613
NM_006760; uroplakin 2
CIS


491
U133A
209138_x_at
168
Hs.505407
NM_015162; lipidosin
CIS


492
U133A
209270_at
168
Hs.436983
NM_000228; laminin subunit beta 3
CIS







precursor


493
U133A
209340_at
168
Hs.21293
NM_007144; ring finger protein 110
CIS


494
U133A
209591_s_at
168
Hs.170195
NM_000228; laminin subunit beta 3
CIS







precursor


495
U133A
209732_at
168
Hs.85201
NM_001248; ectonucleoside triphosphate
CIS







diphosphohydrolase 3


496
U133A
210143_at
168
Hs.188401
NM_007193; annexin A10
CIS


497
U133A
210735_s_at
168
Hs.5338
NM_017689; hypothetical protein
CIS







FLJ20151


498
U133A
210761_s_at
168
Hs.86859
NM_020142; NADH: ubiquinone oxidoreductase
CIS







MLRQ subunit homolog


499
U133A
211002_s_at
168
Hs.82237
NM_001958; eukaryotic translation
CIS







elongation factor 1 alpha 2


500
U133A
211161_s_at
168

NM_000300; phospholipase A2, group
CIS







IIA (platelets, synovial fluid)


501
U133A
211430_s_at
168
Hs.413826
NM_001910; cathepsin E isoform a
CIS







preproprotein NM_148964; cathepsin







E isoform b preproprotein


502
U133A
211671_s_at
168
Hs.126608
NM_007144; ring finger protein 110
CIS


503
U133A
211692_s_at
168
Hs.87246
NM_014417; BCL2 binding component 3
CIS


504
U133A
211896_s_at
168
Hs.156316
NM_005581; Lutheran blood group
CIS







(Auberger b antigen included)


505
U133A
212077_at
168
Hs.443811
NM_003282; troponin I, skeletal, fast
CIS


506
U133A
212192_at
168
Hs.109438
NM_020142; NADH: ubiquinone oxidoreductase
CIS







MLRQ subunit homolog


507
U133A
212195_at
168
Hs.71968
NM_000445; plectin 1, intermediate
CIS







filament binding protein 500 kDa


508
U133A
212386_at
168
Hs.359289
NM_005547; involucrin
CIS


509
U133A
212667_at
168
Hs.111779
NM_000299; plakophilin 1
CIS


510
U133A
212671_s_at
168
Hs.387679
NM_002145; homeo box B2
CIS


511
U133A
212998_x_at
168
Hs.375115
NM_000497; cytochrome P450, sub-
CIS







family XIB (steroid 11-beta-







hydroxylase), polypeptide 1 precursor


512
U133A
213891_s_at
168
Hs.359289
NM_007193; annexin A10
CIS


513
U133A
213975_s_at
168
Hs.234734
NM_005522; homeobox A1 protein
CIS







isoform a NM_153620; homeobox A1







protein isoform b


514
U133A
214352_s_at
168
Hs.412107
NM_006760; uroplakin 2
CIS


515
U133A
214599_at
168
Hs.157091
NM_005547; involucrin
CIS


516
U133A
214630_at
168
Hs.184927
NM_000497; cytochrome P450, sub-
CIS







family XIB (steroid 11-beta-







hydroxylase), polypeptide 1 precursor


517
U133A
214639_s_at
168
Hs.67397
NM_005522; homeobox A1 protein
CIS







isoform a NM_153620; homeobox A1







protein isoform b


518
U133A
214651_s_at
168
Hs.127428
NM_002145; homeo box B2
CIS


519
U133A
214669_x_at
168
Hs.377975
NM_001442; fatty acid binding protein
CIS







4, adipocyte


520
U133A
214677_x_at
168
Hs.449601
NM_006942; SRY-box 15
CIS


521
U133A
214752_x_at
168
Hs.195464
NM_006942; SRY-box 15
CIS


522
U133A
215076_s_at
168
Hs.443625
NM_016233; peptidylarginine deiminase
CIS







type III


523
U133A
215121_x_at
168
Hs.356861
NM_018058; cartilage acidic protein 1
CIS


524
U133A
215176_x_at
168
Hs.503443
NM_001248; ectonucleoside triphosphate
CIS







diphosphohydrolase 3


525
U133A
215379_x_at
168
Hs.449601
NM_006760; uroplakin 2
CIS


526
U133A
215812_s_at
168
Hs.499113
NM_018058; cartilage acidic protein 1
CIS


527
U133A
216641_s_at
168
Hs.18141
NM_005547; involucrin
CIS


528
U133A
216971_s_at
168
Hs.79706
NM_000445; plectin 1, intermediate
CIS







filament binding protein 500 kDa


529
U133A
217028_at
168
Hs.421986
NM_003282; troponin I, skeletal, fast
CIS


530
U133A
217040_x_at
168
Hs.95582
NM_001910; cathepsin E isoform a
CIS







preproprotein NM_148964; cathepsin







E isoform b preproprotein


531
U133A
217388_s_at
168
Hs.444471
NM_000228; laminin subunit beta 3
CIS







precursor


532
U133A
217626_at
168
Hs.201967
NM_000299; plakophilin 1
CIS


533
U133A
218484_at
168
Hs.221447
NM_020142; NADH: ubiquinone oxidoreductase
CIS







MLRQ subunit homolog


534
U133A
218656_s_at
168
Hs.93765
NM_001442; fatty acid binding protein
CIS







4, adipocyte


535
U133A
218718_at
168
Hs.43080
NM_000445; plectin 1, intermediate
CIS







filament binding protein 500 kDa


536
U133A
218918_at
168
Hs.8910
NM_000300; phospholipase A2, group
CIS







IIA (platelets, synovial fluid)


537
U133A
218960_at
168
Hs.414005
NM_019894; transmembrane protease,
CIS







serine 4 isoform 1 NM_183247;







transmembrane protease, serine 4







isoform 2


538
U133A
219410_at
168
Hs.104800
NM_004692; NM_032727; internexin
CIS







neuronal intermediate filament protein,







alpha


539
U133A
219922_s_at
168
Hs.289019
NM_030570; uroplakin 3B isoform a
CIS







NM_182683; uroplakin 3B isoform c







NM_182684; uroplakin 3B isoform b


540
U133A
220026_at
168
Hs.227059
NM_001442; fatty acid binding protein
CIS







4, adipocyte


541
U133A
220779_at
168
Hs.149195
NM_016233; peptidylarginine deiminase
CIS







type III


542
U133A
221204_s_at
168
Hs.326444
NM_018058; cartilage acidic protein 1
CIS


543
U133A
221660_at
168
Hs.247831
NM_000300; phospholipase A2, group
CIS







IIA (platelets, synovial fluid)


544
U133A
221671_x_at
168
Hs.377975
NM_000299; plakophilin 1
CIS


545
U133A
221854_at
168
Hs.313068
NM_000299; plakophilin 1
CIS


546
U133A
221872_at
168
Hs.82547
NM_001958; eukaryotic translation
CIS







elongation factor 1 alpha 2


547
U133A
200958_s_at
168
Hs.164067
NM_005625; syndecan binding protein
CIS







(syntenin)


548
U133A
201877_s_at
168
Hs.249955
NM_002719; gamma isoform of regulatory
CIS







subunit B56, protein phosphatase







2A isoform a NM_178586;







gamma isoform of regulatory subunit







B56, protein phosphatase 2A isoform b







NM_178587; gamma isoform of regulatory







subunit B56, protein phosphatase







2A isoform c NM_178588;







gamma isoform of regulatory subunit







B56, protein phosphatase 2A isoform d


549
U133A
201887_at
168
Hs.285115
NM_001560; interleukin 13 receptor,
CIS







alpha 1 precursor


550
U133A
202076_at
168
Hs.289107
NM_001166; baculoviral IAP repeat-
CIS







containing protein 2


551
U133A
202777_at
168
Hs.104315
NM_007373; soc-2 suppressor of clear
CIS







homolog


552
U133A
204640_s_at
168
Hs.129951
NM_003563; speckle-type POZ protein
CIS


553
U133A
209004_s_at
168
Hs.5548
NM_012161; F-box and leucine-rich
CIS







repeat protein 5 isoform 1







NM_033535; F-box and leucine-rich







repeat protein 5 isoform 2


554
U133A
209241_x_at
168
Hs.112028
NM_015716; misshapen/NIK-related
CIS







kinase isoform 1 NM_153827; misshapen/







NIK-related kinase isoform 3







NM_170663; misshapen/NIK-related







kinase isoform 2


555
U133A
209579_s_at
168
Hs.35947
NM_003925; methyl-CpG binding
CIS







domain protein 4


556
U133A
209630_s_at
168
Hs.444354
NM_012164; F-box and WD-40 domain
CIS







protein 2


557
U133A
212784_at
168
Hs.388236
NM_015125; capicua homolog
CIS


558
U133A
212802_s_at
168
Hs.287266

CIS


559
U133A
212899_at
168
Hs.129836
NM_015076; cyclin-dependent kinase
CIS







(CDC2-like) 11


560
U133A
213633_at
168
Hs.97858
NM_018957; SH3-domain binding
CIS







protein 1


561
U133A
217941_s_at
168
Hs.8117
NM_018695; erbb2 interacting protein
CIS


562
U133A
218150_at
168
Hs.342849
NM_012097; ADP-ribosylation factor-
CIS







like 5 isoform 1 NM_177985; ADP-







ribosylation factor-like 5 isoform 2









The expression level of at least one gene in the sample is determined, wherein at least one of said genes is selected from the genes of Table A. The samples according to the present invention may be any tissue sample or body fluid sample, it is however often preferred to conduct the methods according to the invention on epithelial tissue, such as epithelial tissue from the bladder. In particular the epithelial tissue may be mucosa. In another embodiment the sample is a urine sample comprising the tissue cells.


The sample may be obtained by any suitable manner known to the man skilled in the art, such as a biopsy of the tissue, or a superficial sample scraped from the tissue. The sample may be prepared by forming a cell suspension made from the tissue, or by obtaining an extract from the tissue.


In one embodiment it is preferred that the sample comprises substantially only cells from said tissue, such as substantially only cells from mucosa of the bladder.


The methods according to the invention may be used for determining any biological condition, wherein said condition leads to a change in the expression of at least one gene, and preferably a change in a variety of genes.


Thus, the biological condition may be any malignant or premalignant condition, in particular in bladder, such as a tumor or an adenocarcinoma, a carcinoma, a teratoma, a sarcoma, and/or a lymphoma, and/or carcinoma-in-situ, and/or dysplasia-in-situ.


The expression level may be determined as single gene approaches, i.e. wherein the determination of expression from one or two or a few genes is conducted. It is however preferred that information is obtained from several genes, so that an expression pattern is obtained.


In a preferred embodiment expression from at least one gene from a first group is determined, said first gene group representing genes being expressed at a higher level in one type of tissue, i.e. tissue in one stage or one risk group, in combination with determination of expression of at least one gene from a second group, said second group representing genes being expressed at a higher level in tissue from another stage or from another risk group. Thereby the validity of the prediction increases, since expression levels from genes from more than one group are determined.


However, determination of the expression of a single gene whether belonging to the first group or second group is also within the scope of the present invention. In this case it is preferred that the single gene is selected among genes having a high change in expression level from normal cells to biological condition cells.


Another approach is determination of an expression pattern from a variety of genes, wherein the determination of the biological condition in the tissue relies on information from a variety of gene expression, i.e. rather on the combination of expressed genes than on the information from single genes.


The following data presented herein relates to bladder tumors, and therefore the description has focused on the gene expression level as one way of identifying genes that lose or gain function in cancer tissue. Genes showing a remarkable downregulation (or complete loss) or upregulation (gene expression gained de novo) of the expression level—measured as the mRNA transcript, during the malignant progression in bladder from normal mucosa through Ta superficial tumors, and Carcinoa in situ (CIS) to T1, slightly invasive tumors, to T2, T3 and T4 which have spread to muscle or even further into lymph nodes or other organs are within the scope of the invention, as well as genes gaining importance during the differentiation from normal towards malignancy.


The present invention relates to a variety of genes identified either by an EST identification number and/or by a gene identification number. Both type of identification numbers relates to identification numbers of UniGene database, NCBI, build 18.


The various genes have been identified using Affymetrix arrays of the following product numbers:


HUGeneFL (sold in 2000-2002)


EOS Hu03 (customized Affymetric array)


U133A (product #900367 sold in 2003)


Stage of a bladder tumor indicates how deep the tumor has penetrated. Superficial tumors are termed Ta, and Carcinoma in situ (CIS), and T1, T2, T3 and T4 are used to describe increasing degrees of penetration into the muscle. The grade of a bladder tumor is expressed on a scale of I-IV (1-4) according to Bergkvist, A.; Ijungquist, A.; Moberger, B. “Classification of bladder tumours basedf on the cellular pattern. Preliminary report of a clinical-pathological study of 300 cases with a minimum follow-up of eight years”, Acta Chir Scand., 1965, 130(4):371-8). The grade reflects the cytological appearance of the cells. Grade I cells are almost normal. Grade II cells are slightly deviant. Grade III cells are clearly abnormal. And Grade IV cells are highly abnormal. A special form of bladder malignancy is carcinoma-in-situ or dyplasia-in-situ in which the altered cells are located in-situ.


It is important to predict the prognosis of a cancer disease, as superficial tumors may require a less intensive treatment than invasive tumors. According to the invention the expression level of genes may be used to identify genes whose expression can be used to identify a certain stage and/or the prognosis of the disease. These “Classifiers” are divided into those which can be used to identify Ta, Carcinoma in situ (CIS), T1, and T2 stages as well as those identifying risk of recurrence or progression. In one aspect of the invention measuring the transcript level of one or more of these genes may lead to a classifier that can add supplementary information to the information obtained from the pathological classification. For example gene expression levels that signify a T2 stage will be unfavourable to detect in a Ta tumor, as they may signal that the Ta tumor has the potential to become a T2 tumor. The opposite is probably also true, that an expression level that signify Ta will be favorable to have in a T2 tumor. In that way independent information may be obtained from pathological classification and a classification based on gene expression levels is made.


In the present context a standard expression level is the level of expression of a gene in a standard situation, such as a standard Ta tumor or a standard T2 tumor. For use in the pre-sent invention standard expression levels is determined for each stage as well as for each group of progression, recurrence, and other prognostic indices. It is then possible to compare the result of a determination of the expression level from a gene of a given biological condition with a standard for each stage, progression, recurrence and other indices to obtain a classification of the biological condition.


Furthermore, in the present context a reference pattern refers to the pattern of expression levels seen in standard situations as discussed above, and reference patterns may be used as discussed above for standard expression levels.


It is known from the histopathological classification of bladder tumors that some information is obtained from merely classifying into stage and grade of tumor. Accordingly, in one aspect, the invention relates to a method of predicting the prognosis of the biological condition by determining the stage of the biological condition, by determining an expression level of at least one gene, wherein said gene is selected from the group of genes consisting of gene No 1 to gene No. 562. In this aspect information about the stage reveils directly information about the prognosis as well. An example hereof is when a bladder tumor is classified as for example stage T2, then the prognosis for the bladder tumor is obtained directly from the prognosis related generally to stage T2 tumors. In a preferred embodiment the genes for predicting the prognosis by establishing the stage of the tumor may be selected from gene selected from the group of genes consisting of gene No. 1 to gene No. 188. More preferably the genes for predicting the prognosis by establishing the stage of the tumor may be selected from gene selected from the group of genes consisting of gene Nos. 18, 39, 40, 55, 58, 79, 86, 87, 88, 91, 93, 103, 105, 106, 121, 123, 125, 126, 136, 137, 140, 149, 156, 158, 161, 165, 166, 167, 175, 184, 187, 188.


It is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes, such as the expression level of at least 15 genes, such as the expression level of at least 20 genes, such as the expression levels of at least 25 genes, such as the expression levels of at least 30 genes, such as the expression level of 32 genes.


As discussed above, in relation to bladder cancer the stages of a bladder tumor are selected from bladder cancer stages Ta, Carcinoma in situ, T1, T2, T3 and T4. In an embodiment the determination of a stage comprises


assaying at least the expression of Ta stage gene from a Ta stage gene group, at least one expression of a CIS gene, at least one expression of T1 stage gene from a T1 stage gene group, at least the expression of T2 stage gene from a T2 stage gene group, and more preferably assaying at least the expression of Ta stage gene from a Ta stage gene group, at least one expression of a CIS gene, at least one expression of T1 stage gene from a T1 stage gene group, at least the expression of T2 stage gene from a T2 stage gene group, at least the expression of T3 stage gene from a T3 stage gene group, at least the expression of T4 stage gene from a T4 stage gene group wherein at least one gene from each gene group is expressed in a significantly different amount in that stage than in one of the other stages.


Preferably, the genes selected may be a gene from each gene group being expressed in a significantly higher amount in that stage than in one of the other stages as compared to normal controls, see for example Table B below.


The genes selected may be a gene from each gene group being expressed in a significantly lower amount in that stage than in one of the other stages.


In another embodiment the present invention relates to a method of predicting the prognosis of a biological condition by obtaining information in addition to the stage classification as such. As described above, by determining gene expression levels that signify a T2 stage in a tumor otherwise classified as a Ta tumor, the expression levels signal that the Ta tumor has the potential to become a T2 tumor. The opposite is also true, that an expression level that signify Ta will be favorable to have in a T2 tumor. In the present invention the inventors have shown that some genes are relevant for obtaining this additional information.


Also, in one embodiment the present invention relates to a further method of predicting the prognosis of a biological condition by obtaining information in addition to the stage classification as such. Determination of squamous metaplasia in a tumor, in particular in a T2 stage tumor, is indicative of risk of progression. In particular the genes may be selected from gene selected from the group of genes consisting of gene No. 215 to gene No. 232, see also table H.


It is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes, such as the expression level of at least 15 genes, such as the expression level of 18 genes.


In another embodiment the invention relates to genes bearing information of recurrence of the biological condition as such. In particular the genes may be selected from gene selected from the group of genes consisting of gene No. 189 to gene No. 214. It is preferred to determine a first expression level of at least one gene from a first gene group, wherein the gene from the first gene group is selected from the group of genes wherein expression is increased in case of recurrence, genes No. 189 to gene No. 199 (recurrence genes), and determined a second expression level of at least one gene from a second gene group, wherein the second gene group is selected from the group of genes wherein expression is increased in case of no recurrence, genes No. 200 to No. 214 (non-recurrence genes), and correlate the first expression level to a standard expression level for progressors, and/or the second expression level to a standard expression level for non-progressors to predict the prognosis of the biological condition in the animal tissue, see also table C.


It is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes, such as the expression level of at least 15 genes, such as the expression level of at least 20 genes, such as the expression level of at least 25 genes, such as the expression level of 26 genes.


Furthermore, in another embodiment the invention relates to genes bearing information of progression as such. In particular the genes may be selected from the group of genes of table D, more preferably selected from the group of genes consisting of gene No. 233 to gene No. 446. More preferably the genes may be selected from the group of genes Nos. 255, 273, 279, 280, 281, 282, 287, 295, 300, 311, 317, 320, 333, 346, 347, 349, 352, 364, 365, 373, 383, 386, 390, 394, 401, 407, 414, 417, 426, 427, 428, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, see table E.


It is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes, such as the expression level of at least 15 genes, such as the expression level of at least 20 genes, such as the expression levels of at least 25 genes, such as the expression levels of at least 30 genes, such as the expression level of at least 35 genes, such as the expression level of at least 40 genes, such as the expression level of 45 genes.


Furthermore, it is within the scope of the invention to predict the prognosis of a biological condition in animal tissue by determining the expression level of at least two genes, by

    • determining a first expression level of at least one gene from a first gene group, wherein the gene from the first gene group is selected from the group of gene Nos. 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 369, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, 444 (progressor genes), and
    • determining a second expression level of at least one gene from a second gene group, wherein the second gene group is selected from the group of genes Nos. 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, 446 (non-progressor genes), and
    • correlating the first expression level to a standard expression level for progressors, and/or the second expression level to a standard expression level for non-progressors to predict the prognosis of the biological condition in the animal tissue.


In particular the genes of the first group and the second group for predicting the prognosis of a Ta stage tumor may be selected from gene selected from the group of progression/non-progression genes described above.


In yet another embodiment the present invention offers the possibility to predict the presence or absence of Carcinoma in situ in the same organ as the primary biological condition. An example hereof is for a Ta bladder tumor to predict, whether the bladder in addition to the Ta tumor comprises Carcinoma in situ (CIS). The presence of carcinoma in situ in a bladder containing a superficial Ta tumor is a signal that the Ta tumor has the potential of recurrence and invasiveness. Accordingly, by predicting the presence of carcinoma in situ important information about the prognosis is obtained. In the present context, genes for predicting the presence of carcinoma in situ for a Ta stage tumor may be selected from gene selected from the group of genes consisting of gene No. 447 to gene No. 562. More preferably the genes are selected from the group of genes consisting of gene Nos 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, see table F, or from the group of genes consisting of gene Nos. 547, 548, 549, 550, 551, 552, 553, 554, 555, 556, 557, 558, 559, 560, 561, 562, see table G.


It is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes, such as the expression level of at least 15 genes, such as the expression level of at least 20 genes, such as the expression levels of at least 25 genes, such as the expression levels of at least 30 genes, such as the expression level of at least 35 genes, such as the expression level of at least 40 genes, such as the expression level of at least 45 genes, such as the expression level of at least 50 genes, such as 100 genes. In another embodiment the expression level of 16 genes are determined.


It is also preferred to determine a first expression level of at least one gene from a first gene group, wherein the gene from the first gene group is selected from the group of genes wherein expression is increased in case of CIS, genes Nos. 447, 448, 449, 450, 451, 452, 454, 455, 456, 457, 458, 459, 462, 468, 474, 478, 484, 489, 491, 493, 495, 500, 501, 502, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 518, 519, 520, 522, 523, 524, 525, 529, 531, 534, 535, 536, 538, 544, 546, 547, 548, 549, 550, 551, 552, 553, 555, 556, 558, 559, 561, 562 (CIS genes), and determined a second expression level of at least one gene from a second gene group, wherein the second gene group is selected from the group of genes wherein expression is increased in case of no CIS, genes Nos. 453, 460, 461, 463, 464, 465, 466, 467, 469, 470, 471, 472, 473, 475, 476, 477, 479, 480, 481, 482, 483, 485, 486, 487, 488, 490, 492, 494, 496, 497, 498, 499, 503, 515, 516, 517, 521, 526, 527, 528, 530, 532, 533, 537, 539, 540, 541, 542, 543, 545, 554, 557, 560 (non-CIS genes), and correlate the first expression level to a standard expression level for CIS, and/or the second expression level to a standard expression level for non-CIS to predict the prognosis of the biological condition in the animal tissue.


It is preferred when determining the expression level of at least one gene from a first group and at least one gene from a second group that the expression level of more than one genes from each group is determined. Thus, it is preferred that the expression level of more one gene is determined, such as the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of at least six genes, such as the expression level of at least seven genes, such as the expression level of at least eight genes, such as the expression level of at least nine genes, such as the expression level of at least ten genes in each group.


In one embodiment of the invention the stage of the biological condition has been determined before the prediction of prognosis. The stage may be determined by any suitable means such as determined by histological examination of the tissue or by genotyping of the tissue, preferably by genotyping of the tissue such as described herein or as described in WO 02/02804 incorporated herein by reference.


In another aspect the invention relates to a method of determining the stage of a biological condition in animal tissue,

    • comprising collecting a sample comprising cells from the tissue,
    • determining an expression level of at least one gene selected from the group of genes consisting of gene No. 1 to gene No. 562,
    • correlating the expression level of the assessed genes to at least one standard level of expression determining the stage of the condition.


In particular the expression level of at least one gene selected from the group of genes consisting of gene Nos. 1-457 and gene Nos. 459-535 and gene Nos. 537-562.


Specific embodiments of determining the stage is as described above for predicting prognosis by determination of stage.


In a preferred embodiment the expression level of at least two genes is determined by

    • determining the expression of at least a first stage gene from a first stage gene group and at least a second stage gene from a second stage gene group, wherein at least one of said genes is expressed in said first stage of the condition in a higher amount than in said second stage, and the other gene is a expressed in said first stage of the condition in a lower amount than in said second stage of the condition, and
    • correlating the expression level of the assessed genes to a standard level of expression determining the stage of the condition.


In general, genes being downregulated for higher stage tumors as well as for progression and recurrence may be of importance as predictive markers for the disease as loss of one or more of these may signal a poor outcome or an aggressive disease course. Furthermore, they may be important targets for therapy as restoring their expression level, e.g. by gene therapy, or substitution with those peptide products or small molecules with a similar biological effect may suppress the malignant growth.


Genes that are up-regulated (or gained de novo) during the malignant progression of bladder cancer from normal tissue through Ta, T1, T2, T3 and T4 is also within the scope of the invention. These genes are potential oncogenes and may be those genes that create or enhance the malignant growth of the cells. The expression level of these genes may serve as predictive markers for the disease course and treatment response, as a high level may signal an aggressive disease course, and they may serve as targets for therapy, as blocking these genes by e.g. anti-sense therapy, or by biochemical means could inhibit, or slow the tumor growth.


The genes used according to the invention show a sufficient difference in expression from one group to another and/or from one stage to another to use the gene as a classifier for the group and/or stage. Thus, comparison of an expression pattern to another may score a change from expressed to non-expressed, or the reverse. Alternatively, changes in intensity of expression may be scored, either increases or decreases. Any significant change can be used. Typical changes which are more than 2-fold are suitable. Changes which are greater than 5-fold are highly suitable.


The present invention in particular relates to methods using genes wherein at least a two-fold change in expression, such as at least a three-fold change, for example at least a four fold change, such as at least a five fold change, for example at least a six fold change, such as at least a ten fold change, for example at least a fifteen fold change, such as at least a twenty fold change is seen between two groups.


As described above the invention relates to the use of information of expression levels. In one embodiment the expression patterns is obtained, thus, the invention relates to a method of determining an expression pattern of a bladder cell sample, comprising:

    • collecting sample comprising bladder cells and/or expression products from bladder cells,
    • determining the expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562, and obtaining an expression pattern of the bladder cell sample.


The invention preferably include more than one gene in the pattern, according it is preferred to include the expression level of at least two genes, such as the expression level of at least three genes, such as the expression level of at least four genes, such as the expression level of at least five genes, such as the expression level of more than six genes.


The expression pattern preferably relates to one or more of the group of genes discussed above with respect to prognosis relating to stage, SSC, progression, recurrence and/or CIS.


In order to predict prognosis and/or stages it is preferred to determine an expression pattern of a cell sample preferably independent of the proportion of submucosal, muscle and connective tissue cells present. Expression is determined of one or more genes in a sample comprising cells, said genes being selected from the same genes as discussed above and shown in the tables.


It is an object of the present invention that characteristic patterns of expression of genes can be used to characterize different types of tissue. Thus, for example gene expression patterns can be used to characterize stages and grades of bladder tumors. Similarly, gene expression patterns can be used to distinguish cells having a bladder origin from other cells. Moreover, gene expression of cells which routinely contaminate bladder tumor biopsies has been identified, and such gene expression can be removed or subtracted from patterns obtained from bladder biopsies. Further, the gene expression patterns of single-cell solutions of bladder tumor cells have been found to be substantially without interfering expression of contaminating muscle, submucosal, and connective tissue cells than biopsy samples.


The one or more genes exclude genes which are expressed in the submucosal, muscle, and connective tissue. A pattern of expression is formed for the sample which is independent of the proportion of submucosal, muscle, and connective tissue cells in the sample.


In another aspect of the invention a method of determining an expression pattern of a cell sample is provided. Expression is determined of one or more genes in a sample comprising cells. A first pattern of expression is thereby formed for the sample. Genes which are expressed in submucosal, muscle, and connective tissue cells are removed from the first pattern of expression, forming a second pattern of expression which is independent of the proportion of submucosal, muscle, and connective tissue cells in the sample.


Another embodiment of the invention provides a method for determining an expression pattern of a bladder mucosa or bladder cancer cell. Expression is determined of one or more genes in a sample comprising bladder mucosa or bladder cancer cells; the expression determined forms a first pattern of expression. A second pattern of expression which was formed using the one or more genes and a sample comprising predominantly submucosal, muscle, and connective tissue cells, is subtracted from the first pattern of expression, forming a third pattern of expression. The third pattern of expression reflects expression of the bladder mucosa or bladder cancer cells independent of the proportion of submucosal, muscle, and connective tissue cells present in the sample.


In one embodiment the invention provides a method to predict the prognosis of a bladder tumor as described above. A first pattern of expression is determined of one or more genes in a bladder tumor sample. The first pattern is compared to one or more reference patterns of expression determined for bladder tumors at different stages and/or in different groups. The reference pattern which shares maximum similarity with the first pattern is identified. The stage of the reference pattern with the maximum similarity is assigned to the bladder tumor sample.


Yet another embodiment the invention provides a method to determine the stage of a bladder tumor as described above. A first pattern of expression is determined of one or more genes in a bladder tumor sample. The first pattern is compared to one or more reference patterns of expression determined for bladder tumors at different stages. The reference pattern which shares maximum similarity with the first pattern is identified. The stage of the reference pattern with the maximum similarity is assigned to the bladder tumor sample.


Since a biopsy of the tissue often contains more tissue material such as connective tissue than the tissue to be examined, when the tissue to be examined is epithelial or mucosa, the invention also relates to methods, wherein the expression pattern of the tissue is independent of the amount of connective tissue in the sample.


Biopsies contain epithelial cells that most often are the targets for the studies, and in addition many other cells that contaminate the epithelial cell fraction to a varying extent. The contaminants include histiocytes, endothelial cells, leukocytes, nerve cells, muscle cells etc. Micro dissection is the method of choice for DNA examination, but in the case of expression studies this procedure is difficult due to RNA degradation during the procedure. The epithelium may be gently removed and the expression in the remaining submucosa and underlying connective tissue (the bladder wall) monitored. Genes expressed at high or low levels in the bladder wall should be interrogated when performing expression monitoring of the mucosa and tumors. A similar approach could be used for studies of epithelia in other organs.


In one embodiment of the invention normal mucosa lining the bladder lumen from bladders for cancer is scraped off. Then biopsies is taken from the denuded submucosa and connective tissue, reaching approximately 5 mm into the bladder wall, and immediately disintegrated in guanidinium isothiocyanate. Total RNA may be extracted, pooled, and poly(A)+ mRNA may be prepared from the pool followed by conversion to double-stranded cDNA and in vitro transcription into cRNA containing biotin-labeled CTP and UTP.


Genes that are expressed and genes that are not expressed in bladder wall can both interfere with the interpretation of the expression in a biopsy, and should be considered when interpreting expression intensities in tumor biopsies, as the bladder wall component of a biopsy varies in amount from biopsy to biopsy.


When having determined the pattern of genes expressed in bladder wall components said pattern may be subtracted from a pattern obtained from the sample resulting in a third pattern related to the mucosa (epithelial) cells.


In another embodiment of the invention a method is provided for determining an expression pattern of a bladder tissue sample independent of the proportion of submucosal, muscle and connective tissue cells present. A single-cell suspension of disaggregated bladder tumor cells is isolated from a bladder tissue sample comprising bladder tumor cells is isolated from a bladder tissue sample comprising bladder cells, submucosal cells, muscle cells, and connective tissue cells. A pattern of expression is thus formed for the sample which is independent of the proportion of submucosal, muscle, and connective tissue cells in the bladder tissue sample.


Yet another method relates to the elimination of mRNA from bladder wall components before determining the pattern, e.g. by filtration and/or affinity chromatography to remove mRNA related to the bladder wall.


Working with tumor material requires biopsies or body fluids suspected to comprise relevant cells. Working with RNA requires freshly frozen or immediately processed biopsies, or chemical pretreatment of the biopsy. Apart from the cancer tissue, biopsies do inevitably contain many different cell types, such as cells present in the blood, connective and muscle tissue, endothelium etc. In the case of DNA studies, microdissection or laser capture are methods of choice, however the time-dependent degradation of RNA makes it difficult to perform manipulation of the tissue for more than a few minutes. Furthermore, studies of expressed sequences may be difficult on the few cells obtained via microdissection or laser capture, as these cells may have an expression pattern that deviates from the predominant pattern in a tumor due to large intratumoral heterogeneity.


In the present context high density expression arrays may be used to evaluate the impact of bladder wall components in bladder tumor biopsies, and tested preparation of single cell solutions as a means of eliminating the contaminants. The results of these evaluations permit for the design of methods of evaluating bladder samples without the interfering background noise caused by ubiquitous contaminating submucosal, muscle, and connective tissue cells. The evaluating assays of the invention may be of any type.


While high density expression arrays can be used, other techniques are also contemplated. These include other techniques for assaying for specific mRNA species, including RT-PCR and Northern Blotting, as well as techniques for assaying for particular protein products, such as ELISA, Western blotting, and enzyme assays. Gene expression patterns according to the present invention are determined by measuring any gene product of a particular gene, including mRNA and protein. A pattern may be for one or more genes.


RNA or protein can be isolated and assayed from a test sample using any techniques known in the art. They can for example be isolated from a fresh or frozen biopsy, from formalin-fixed tissue, from body fluids, such as blood, plasma, serum, urine, or sputum.


Expression of genes may in general be detected by either detecting mRNA from the cells and/or detecting expression products, such as peptides and proteins.


The detection of mRNA of the invention may be a tool for determining the developmental stage of a cell type which may be definable by its pattern of expression of messenger RNA. For example, in particular stages of cells, high levels of ribosomal RNA are found whereas relatively low levels of other types of messenger RNAs may be found. Where a pattern is shown to be characteristic of a stage, said stage may be defined by that particular pattern of messenger RNA expression. The mRNA population is a good determinant of a developmental stage, and may be correlated with other structural features of the cell. In this manner, cells at specific developmental stages will be characterized by the intracellular environment, as well as the extracellular environment. The present invention also allows the combination of definitions based in part upon antigens and in part upon mRNA expression. In one embodiment, the two may be combined in a single incubation step. A particular incubation condition may be found which is compatible with both hybridization recognition and non-hybridization recognition molecules. Thus, e.g. an incubation condition may be selected which allows both specificity of antibody binding and specificity of nucleic acid hybridization. This allows simultaneous performance of both types of interactions on a single matrix. Again, where developmental mRNA patterns are correlated with structural features, or with probes which are able to hybridize to intracellular mRNA populations, a cell sorter may be used to sort specifically those cells having desired mRNA population patterns.


It is within the general scope of the present invention to provide methods for the detection of mRNA. Such methods often involve sample extraction, PCR amplification, nucleic acid fragmentation and labeling, extension reactions, and transcription reactions.


The nucleic acid (either genomic DNA or mRNA) may be isolated from the sample according to any of a number of methods well known to those of skill in the art. One of skill will appreciate that where alterations in the copy number of a gene are to be detected genomic DNA is preferably isolated. Conversely, where expression levels of a gene or genes are to be detected, preferably RNA (mRNA) is isolated.


Methods of isolating total mRNA are well known to those of skill in the art. In one embodiment, the total nucleic acid is isolated from a given sample using, for example, an acid guanidinium-phenol-chloroform extraction method and polyA.sup. and mRNA is isolated by oligo dT column chromatography or by using (dT)n magnetic beads (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd ed.), Vols. 1-3, Cold Spring Harbor Laboratory, (1989), or Current Protocols in Molecular Biology, F. Ausubel et al., ed. Greene Publishing and Wiley-Interscience, New York (1987)).


The sample may be from tissue and/or body fluids, as defined elsewhere herein. Before analyzing the sample, e.g., on an oligonucleotide array, it will often be desirable to perform one or more sample preparation operations upon the sample. Typically, these sample preparation operations will include such manipulations as extraction of intracellular material, e.g., nucleic acids from whole cell samples, viruses, amplification of nucleic acids, fragmentation, transcription, labeling and/or extension reactions. One or more of these various operations may be readily incorporated into the device of the present invention.


DNA extraction may be relevant under circumstances where possible mutations in the genes are to be determined in addition to the determination of expression of the genes.


For those embodiments where whole cells, or other tissue samples are being analyzed, it will typically be necessary to extract the nucleic acids from the cells or viruses, prior to continuing with the various sample preparation operations. Accordingly, following sample collection, nucleic acids may be liberated from the collected cells, viral coat etc. into a crude extract followed by additional treatments to prepare the sample for subsequent operations, such as denaturation of contaminating (DNA binding) proteins, purification, filtration and desalting.


Liberation of nucleic acids from the sample cells, and denaturation of DNA binding proteins may generally be performed by physical or chemical methods. For example, chemical methods generally employ lysing agents to disrupt the cells and extract the nucleic acids from the cells, followed by treatment of the extract with chaotropic salts such as guanidinium isothiocyanate or urea to denature any contaminating and potentially interfering proteins.


Alternatively, physical methods may be used to extract the nucleic acids and denature DNA binding proteins, such as physical protrusions within microchannels or sharp edged particles piercing cell membranes and extract their contents. Combinations of such structures with piezoelectric elements for agitation can provide suitable shear forces for lysis.


More traditional methods of cell extraction may also be used, e.g., employing a channel with restricted cross-sectional dimension which causes cell lysis when the sample is passed through the channel with sufficient flow pressure. Alternatively, cell extraction and denaturing of contaminating proteins may be carried out by applying an alternating electrical current to the sample. More specifically, the sample of cells is flowed through a microtubular array while an alternating electric current is applied across the fluid flow. Subjecting cells to ultrasonic agitation, or forcing cells through microgeometry apertures, thereby subjecting the cells to high shear stress resulting in rupture are also possible extraction methods.


Following extraction, it will often be desirable to separate the nucleic acids from other elements of the crude extract, e.g. denatured proteins, cell membrane particles and salts. Removal of particulate matter is generally accomplished by filtration or flocculation. Further, where chemical denaturing methods are used, it may be desirable to desalt the sample prior to proceeding to the next step. Desalting of the sample and isolation of the nucleic acid may generally be carried out in a single step, e.g. by binding the nucleic acids to a solid phase and washing away the contaminating salts, or performing gel filtration chromatography on the sample passing salts through dialysis membranes. Suitable solid supports for nucleic acid binding include e.g. diatomaceous earth or silica (i.e., glass wool). Suitable gel exclusion media also well known in the art may be readily incorporated into the devices of the present invention and is commercially available from, e.g., Pharmacia and Sigma Chemical.


Alternatively, desalting methods may generally take advantage of the high electrophoretic mobility and negativity of DNA compared to other elements. Electrophoretic methods may also be utilized in the purification of nucleic acids from other cell contaminants and debris. Upon application of an appropriate electric field, the nucleic acids present in the sample will migrate toward the positive electrode and become trapped on the capture membrane. Sample impurities remaining free of the membrane are then washed away by applying an appropriate fluid flow. Upon reversal of the voltage, the nucleic acids are released from the membrane in a substantially purer form. Further, coarse filters may also be overlaid on the barriers to avoid any fouling of the barriers by particulate matter, proteins or nucleic acids, thereby permitting repeated use.


In a similar aspect, the high electrophoretic mobility of nucleic acids with their negative charges, may be utilized to separate nucleic acids from contaminants by utilizing a short column of a gel or other appropriate matrices or gels which will slow or retard the flow of other contaminants while allowing the faster nucleic acids to pass.


This invention provides nucleic acid affinity matrices that bear a large number of different nucleic acid affinity ligands allowing the simultaneous selection and removal of a large number of preselected nucleic acids from the sample. Methods of producing such affinity matrices are also provided. In general the methods involve the steps of a) providing a nucleic acid amplification template array comprising a surface to which are attached at least 50 oligonucleotides having different nucleic acid sequences, and wherein each different oligonucleotide is localized in a predetermined region of said surface, the density of said oligonucleotides is greater than about 60 different oligonucleotides per 1 cm.sup.2, and all of said different oligonucleotides have an identical terminal 3′ nucleic acid sequence and an identical terminal 5′ nucleic acid sequence. b) amplifying said multiplicity of oligonucleotides to provide a pool of amplified nucleic acids; and c) attaching the pool of nucleic acids to a solid support.


For example, nucleic acid affinity chromatography is based on the tendency of complementary, single-stranded nucleic acids to form a double-stranded or duplex structure through complementary base pairing. A nucleic acid (either DNA or RNA) can easily be attached to a solid substrate (matrix) where it acts as an immobilized ligand that interacts with and forms duplexes with complementary nucleic acids present in a solution contacted to the immobilized ligand. Unbound components can be washed away from the bound complex to either provide a solution lacking the target molecules bound to the affinity column, or to provide the isolated target molecules themselves. The nucleic acids captured in a hybrid duplex can be separated and released from the affinity matrix by denaturation either through heat, adjustment of salt concentration, or the use of a destabilizing agent such as formamide, TWEEN™-20 denaturing agent, or sodium dodecyl sulfate (SDS).


Affinity columns (matrices) are typically used either to isolate a single nucleic acid typically by providing a single species of affinity ligand. Alternatively, affinity columns bearing a single affinity ligand (e.g. oligo dt columns) have been used to isolate a multiplicity of nucleic acids where the nucleic acids all share a common sequence (e.g. a polyA).


The type of affinity matrix used depends on the purpose of the analysis. For example, where it is desired to analyze mRNA expression levels of particular genes in a complex nucleic acid sample (e.g., total mRNA) it is often desirable to eliminate nucleic acids produced by genes that are constitutively overexpressed and thereby tend to mask gene products expressed at characteristically lower levels. Thus, in one embodiment, the affinity matrix can be used to remove a number of preselected gene products (e.g., actin, GAPDH, etc.). This is accomplished by providing an affinity matrix bearing nucleic acid affinity ligands complementary to the gene products (e.g., mRNAs or nucleic acids derived therefrom) or to subsequences thereof. Hybridization of the nucleic acid sample to the affinity matrix will result in duplex formation between the affinity ligands and their target nucleic acids. Upon elution of the sample from the affinity matrix, the matrix will retain the duplexes nucleic acids leaving a sample depleted of the overexpressed target nucleic acids.


The affinity matrix can also be used to identify unknown mRNAs or cDNAs in a sample. Where the affinity matrix contains nucleic acids complementary to every known gene (e.g., in a cDNA library, DNA reverse transcribed from an mRNA, mRNA used directly or amplified, or polymerized from a DNA template) in a sample, capture of the known nucleic acids by the affinity matrix leaves a sample enriched for those nucleic acid sequences that are unknown. In effect, the affinity matrix is used to perform a subtractive hybridization to isolate unknown nucleic acid sequences. The remaining “unknown” sequences can then be purified and sequenced according to standard methods.


The affinity matrix can also be used to capture (isolate) and thereby purify unknown nucleic acid sequences. For example, an affinity matrix can be prepared that contains nucleic acid (affinity ligands) that are complementary to sequences not previously identified, or not previously known to be expressed in a particular nucleic acid sample. The sample is then hybridized to the affinity matrix and those sequences that are retained on the affinity matrix are “unknown” nucleic acids. The retained nucleic acids can be eluted from the matrix (e.g. at increased temperature, increased destabilizing agent concentration, or decreased salt) and the nucleic acids can then be sequenced according to standard methods.


Similarly, the affinity matrix can be used to efficiently capture (isolate) a number of known nucleic acid sequences. Again, the matrix is prepared bearing nucleic acids complementary to those nucleic acids it is desired to isolate. The sample is contacted to the matrix under conditions where the complementary nucleic acid sequences hybridize to the affinity ligands in the matrix. The non-hybridized material is washed off the matrix leaving the desired sequences bound. The hybrid duplexes are then denatured providing a pool of the isolated nucleic acids. The different nucleic acids in the pool can be subsequently separated according to standard methods (e.g. gel electrophoresis).


As indicated above the affinity matrices can be used to selectively remove nucleic acids from virtually any sample containing nucleic acids (e.g. in a cDNA library, DNA reverse transcribed from an mRNA, mRNA used directly or amplified, or polymerized from a DNA template, and so forth). The nucleic acids adhering to the column can be removed by washing with a low salt concentration buffer, a buffer containing a destabilizing agent such as formamide, or by elevating the column temperature.


In one particularly preferred embodiment, the affinity matrix can be used in a method to enrich a sample for unknown RNA sequences (e.g. expressed sequence tags (ESTs)). The method involves first providing an affinity matrix bearing a library of oligonucleotide probes specific to known RNA (e.g., EST) sequences. Then, RNA from undifferentiated and/or unactivated cells and RNA from differentiated or activated or pathological (e.g., transformed) or otherwise having a different metabolic state are separately hybridized against the affinity matrices to provide two pools of RNAs lacking the known RNA sequences.


In a preferred embodiment, the affinity matrix is packed into a columnar casing. The sample is then applied to the affinity matrix (e.g. injected onto a column or applied to a column by a pump such as a sampling pump driven by an autosampler). The affinity matrix (e.g. affinity column) bearing the sample is subjected to conditions under which the nucleic acid probes comprising the affinity matrix hybridize specifically with complementary target nucleic acids. Such conditions are accomplished by maintaining appropriate pH, salt and temperature conditions to facilitate hybridization as discussed above.


For a number of applications, it may be desirable to extract and separate messenger RNA from cells, cellular debris, and other contaminants. As such, the device of the present invention may, in some cases, include a mRNA purification chamber or channel. In general, such purification takes advantage of the poly-A tails on mRNA. In particular and as noted above, poly-T oligonucleotides may be immobilized within a chamber or channel of the device to serve as affinity ligands for mRNA. Poly-T oligonucleotides may be immobilized upon a solid support incorporated within the chamber or channel, or alternatively, may be immobilized upon the surface(s) of the chamber or channel itself. Immobilization of oligonucleotides on the surface of the chambers or channels may be carried out by methods described herein including, e.g., oxidation and silanation of the surface followed by standard DMT synthesis of the oligonucleotides.


In operation, the lysed sample is introduced to a high salt solution to increase the ionic strength for hybridization, whereupon the mRNA will hybridize to the immobilized poly-T. The mRNA bound to the immobilized poly-T oligonucleotides is then washed free in a low ionic strength buffer. The poly-T oligonucleotides may be immobilized upon poroussurfaces, e.g., porous silicon, zeolites silica xerogels, scintered particles, or other solid supports.


Following sample preparation, the sample can be subjected to one or more different analysis operations. A variety of analysis operations may generally be performed, including size based analysis using, e.g., microcapillary electrophoresis, and/or sequence based analysis using, e.g., hybridization to an oligonucleotide array.


In the latter case, the nucleic acid sample may be probed using an array of oligonucleotide probes. Oligonucleotide arrays generally include a substrate having a large number of positionally distinct oligonucleotide probes attached to the substrate. These arrays may be produced using mechanical or light directed synthesis methods which incorporate a combination of photolithographic methods and solid phase oligonucleotide synthesis methods.


The basic strategy for light directed synthesis of oligonucleotide arrays is as follows. The surface of a solid support, modified with photosensitive protecting groups is illuminated through a photolithographic mask, yielding reactive hydroxyl groups in the illuminated regions. A selected nucleotide, typically in the form of a 3′-O-phosphoramidite-activated deoxynucleoside (protected at the 5′ hydroxyl with a photosensitive protecting group), is then presented to the surface and coupling occurs at the sites that were exposed to light. Following capping and oxidation, the substrate is rinsed and the surface is illuminated through a second mask to expose additional hydroxyl groups for coupling. A second selected nucleotide (e.g., 5′-protected, 3′-O-phosphoramidite-activated deoxynucleoside) is presented to the surface. The selective deprotection and coupling cycles are repeated until the desired set of products is obtained. Since photolithography is used the process can be readily miniaturized to generate high density arrays of oligonucleotide probes. Furthermore, the sequence of the oligonucleotides at each site is known. See Pease et al. Mechanical synthesis methods are similar to the light directed methods except involving mechanical direction of fluids for deprotection and addition in the synthesis steps.


For some embodiments, oligonucleotide arrays may be prepared having all possible probes of a given length. The hybridization pattern of the target sequence on the array may be used to reconstruct the target DNA sequence. Hybridization analysis of large numbers of probes can be used to sequence long stretches of DNA or provide an oligonucleotide array which is specific and complementary to a particular nucleic acid sequence. For example, in particularly preferred aspects, the oligonucleotide array will contain oligonucleotide probes which are complementary to specific target sequences, and individual or multiple mutations of these. Such arrays are particularly useful in the diagnosis of specific disorders which are characterized by the presence of a particular nucleic acid sequence.


Following sample collection and nucleic acid extraction, the nucleic acid portion of the sample is typically subjected to one or more preparative reactions. These preparative reactions include in vitro transcription, labeling, fragmentation, amplification and other reactions. Nucleic acid amplification increases the number of copies of the target nucleic acid sequence of interest. A variety of amplification methods are suitable for use in the methods and device of the present invention, including for example, the polymerase chain reaction method or (PCR), the ligase chain reaction (LCR), self sustained sequence replication (3SR), and nucleic acid based sequence amplification (NASBA).


The latter two amplification methods involve isothermal reactions based on isothermal transcription, which produce both single stranded RNA (ssRNA) and double stranded DNA (dsDNA) as the amplification products in a ratio of approximately 30 or 100 to 1, respectively. As a result, where these latter methods are employed, sequence analysis may be carried out using either type of substrate, i.e. complementary to either DNA or RNA.


Frequently, it is desirable to amplify the nucleic acid sample prior to hybridization. One of skill in the art will appreciate that whatever amplification method is used, if a quantitative result is desired, care must be taken to use a method that maintains or controls for the relative frequencies of the amplified nucleic acids.


PCR

Methods of “quantitative” amplification are well known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. The high density array may then include probes specific to the internal standard for quantification of the amplified nucleic acid.


Thus, in one embodiment, this invention provides for a method of optimizing a probe set for detection of a particular gene. Generally, this method involves providing a high density array containing a multiplicity of probes of one or more particular length(s) that are complementary to subsequences of the mRNA transcribed by the target gene. In one embodiment the high density array may contain every probe of a particular length that is complementary to a particular mRNA. The probes of the high density array are then hybridized with their target nucleic acid alone and then hybridized with a high complexity, high concentration nucleic acid sample that does not contain the targets complementary to the probes. Thus, for example, where the target nucleic acid is an RNA, the probes are first hybridized with their target nucleic acid alone and then hybridized with RNA made from a cDNA library (e.g., reverse transcribed polyA.sup.+ mRNA) where the sense of the hybridized RNA is opposite that of the target nucleic acid (to insure that the high complexity sample does not contain targets for the probes). Those probes that show a strong hybridization signal with their target and little or no cross-hybridization with the high complexity sample are preferred probes for use in the high density arrays of this invention.


PCR amplification generally involves the use of one strand of the target nucleic acid sequence as a template for producing a large number of complements to that sequence. Generally, two primer sequences complementary to different ends of a segment of the complementary strands of the target sequence hybridize with their respective strands of the target sequence, and in the presence of polymerase enzymes and nucleoside triphosphates, the primers are extended along the target sequence. The extensions are melted from the target sequence and the process is repeated, this time with the additional copies of the target sequence synthesized in the preceding steps. PCR amplification typically involves repeated cycles of denaturation, hybridization and extension reactions to produce sufficient amounts of the target nucleic acid. The first step of each cycle of the PCR involves the separation of the nucleic acid duplex formed by the primer extension. Once the strands are separated, the next step in PCR involves hybridizing the separated strands with primers that flank the target sequence. The primers are then extended to form complementary copies of the target strands. For successful PCR amplification, the primers are designed so that the position at which each primer hybridizes along a duplex sequence is such that an extension product synthesized from one primer, when separated from the template (complement), serves as a template for the extension of the other primer. The cycle of denaturation, hybridization, and extension is repeated as many times as necessary to obtain the desired amount of amplified nucleic acid.


In PCR methods, strand separation is normally achieved by heating the reaction to a sufficiently high temperature for a sufficient time to cause the denaturation of the duplex but not to cause an irreversible denaturation of the polymerase. Typical heat denaturation involves temperatures ranging from about 80.degree. C. to 105.degree. C. for times ranging from seconds to minutes. Strand separation, however, can be accomplished by any suitable denaturing method including physical, chemical, or enzymatic means. Strand separation may be induced by a helicase, for example, or an enzyme capable of exhibiting helicase activity.


In addition to PCR and IVT reactions, the methods and devices of the present invention are also applicable to a number of other reaction types, e.g., reverse transcription, nick translation, and the like.


The nucleic acids in a sample will generally be labeled to facilitate detection in subsequent steps. Labeling may be carried out during the amplification, in vitro transcription or nick translation processes. In particular, amplification, in vitro transcription or nick translation may incorporate a label into the amplified or transcribed sequence, either through the use of labeled primers or the incorporation of labeled dNTPs into the amplified sequence. Hybridization between the sample nucleic acid and the oligonucleotide probes upon the array is then detected, using, e.g., epifluorescence confocal microscopy. Typically, sample is mixed during hybridization to enhance hybridization of nucleic acids in the sample to nucleic acid probes on the array.


In some cases, hybridized oligonucleotides may be labeled following hybridization. For example, where biotin labeled dNTPs are used in, e.g. amplification or transcription, streptavidin linked reporter groups may be used to label hybridized complexes. Such operations are readily integratable into the systems of the present invention. Alternatively, the nucleic acids in the sample may be labeled following amplification. Post amplification labeling typically involves the covalent attachment of a particular detectable group upon the amplified sequences. Suitable labels or detectable groups include a variety of fluorescent or radioactive labeling groups well known in the art. These labels may also be coupled to the sequences using methods that are well known in the art.


Methods for detection depend upon the label selected. A fluorescent label is preferred because of its extreme sensitivity and simplicity. Standard labeling procedures are used to determine the positions where interactions between a sequence and a reagent take place. For example, if a target sequence is labeled and exposed to a matrix of different probes, only those locations where probes do interact with the target will exhibit any signal. Alternatively, other methods may be used to scan the matrix to determine where interaction takes place. Of course, the spectrum of interactions may be determined in a temporal manner by repeated scans of interactions which occur at each of a multiplicity of conditions. However, instead of testing each individual interaction separately, a multiplicity of sequence interactions may be simultaneously determined on a matrix.


Means of detecting labeled target (sample) nucleic acids hybridized to the probes of the high density array are known to those of skill in the art. Thus, for example, where a colorimetric label is used, simple visualization of the label is sufficient. Where a radioactive labeled probe is used, detection of the radiation (e.g with photographic film or a solid state detector) is sufficient.


In a preferred embodiment, however, the target nucleic acids are labeled with a fluorescent label and the localization of the label on the probe array is accomplished with fluorescent microscopy. The hybridized array is excited with a light source at the excitation wavelength of the particular fluorescent label and the resulting fluorescence at the emission wavelength is detected. In a particularly preferred embodiment, the excitation light source is a laser appropriate for the excitation of the fluorescent label.


The target polynucleotide may be labeled by any of a number of convenient detectable markers. A fluorescent label is preferred because it provides a very strong signal with low background. It is also optically detectable at high resolution and sensitivity through a quick scanning procedure. Other potential labeling moieties include, radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers, magnetic labels, and linked enzymes.


Another method for labeling may bypass any label of the target sequence. The target may be exposed to the probes, and a double strand hybrid is formed at those positions only. Addition of a double strand specific reagent will detect where hybridization takes place. An intercalative dye such as ethidium bromide may be used as long as the probes themselves do not fold back on themselves to a significant extent forming hairpin loops. However, the length of the hairpin loops in short oligonucleotide probes would typically be insufficient to form a stable duplex.


Suitable chromogens will include molecules and compounds which absorb light in a distinctive range of wavelengths so that a color may be observed, or emit light when irradiated with radiation of a particular wave length or wave length range, e.g., fluorescers. Biliproteins, e.g., phycoerythrin, may also serve as labels.


A wide variety of suitable dyes are available, being primarily chosen to provide an intense color with minimal absorption by their surroundings. Illustrative dye types include quinoline dyes, triarylmethane dyes, acridine dyes, alizarine dyes, phthaleins, insect dyes, azo dyes, anthraquinoid dyes, cyanine dyes, phenazathionium dyes, and phenazoxonium dyes.


A wide variety of fluorescers may be employed either by themselves or in conjunction with quencher molecules. Fluorescers of interest fall into a variety of categories having certain primary functionalities. These primary functionalities include 1- and 2-aminonaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminoequilenin, perylene, bis-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigenin, tetracycline, sterophenol, benzimidzaolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, salicylate, strophanthidin, porphyrins, triarylmethanes and flavin. Individual fluorescent compounds which have functionalities for linking or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamineisothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl, N-methyl 2-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioctadecyl oxacarbocyanine; N,N′-dihexyl oxacarbocyanine; merocyanine, 4-(3′pyrenyl)butyrate; d-3-aminodesoxy-equilenin; 12-(9′-anthroyl)stearate; 2-methylanthracene; 9-vinylanthracene; 2,2′-(vinylene-p-phenylene)bisbenzoxazole; p-bis>2-(4-methyl-5-phenyl-oxazolyl)!benzene; 6-dimethylamino-1,2-benzophenazin; retinol; bis(3′-aminopyridinium) 1,10-decandiyl diiodide; sulfonaphthylhydrazone of hellibrienin; chlorotetracycline; N-(7-dimethylamino-4-methyl-2-oxo-3-chromenyl)maleimide; N->p-(2-benzimidazolyl)-phenyl!maleimide; N-(4-fluoranthyl)maleimide; bis(homovanillic acid); resazarin; 4-chloro-7-nitro-2,1,3-benzooxadiazole; merocyanine 540; resorufin; rose bengal; and 2,4-diphenyl-3(2H)-furanone.


Desirably, fluorescers should absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound dye may differ from the unbound dye. Therefore, when referring to the various wavelength ranges and characteristics of the dyes, it is intended to indicate the dyes as employed and not the dye which is unconjugated and characterized in an arbitrary solvent.


Fluorescers are generally preferred because by irradiating a fluorescer with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events.


Detectable signal may also be provided by chemiluminescent and bioluminescent sources. Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectable signal or donates energy to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety of conditions. One family of compounds is 2,3-dihydro-1,-4-phthalazinedione. The most popular compound is luminol, which is the 5-amino compound. Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino>ca!benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base. Another family of compounds is the 2,4,5-triphenylimidazoles, with lophine as the common name for the parent product. Chemiluminescent analogs include para-dimethylamino and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions. Alternatively, luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence.


Spin labels are provided by reporter molecules with an unpaired electron spin which can be detected by electron spin resonance (ESR) spectroscopy. Exemplary spin labels include organic free radicals, transitional metal complexes, particularly vanadium, copper, iron, and manganese, and the like. Exemplary spin labels include nitroxide free radicals.


In addition, amplified sequences may be subjected to other post amplification treatments. For example, in some cases, it may be desirable to fragment the sequence prior to hybridization with an oligonucleotide array, in order to provide segments which are more readily accessible to the probes, which avoid looping and/or hybridization to multiple probes. Fragmentation of the nucleic acids may generally be carried out by physical, chemical or enzymatic methods that are known in the art.


Following the various sample preparation operations, the sample will generally be subjected to one or more analysis operations. Particularly preferred analysis operations include, e.g. sequence based analyses using an oligonucleotide array and/or size based analyses using, e.g. microcapillary array electrophoresis.


In some embodiments it may be desirable to provide an additional, or alternative means for analyzing the nucleic acids from the sample


Microcapillary array electrophoresis generally involves the use of a thin capillary or channel which may or may not be filled with a particular separation medium. Electrophoresis of a sample through the capillary provides a size based separation profile for the sample. Microcapillary array electrophoresis generally provides a rapid method for size based sequencing, PCR product analysis and restriction fragment sizing. The high surface to volume ratio of these capillaries allows for the application of higher electric fields across the capillary without substantial thermal variation across the capillary, consequently allowing for more rapid separations. Furthermore, when combined with confocal imaging methods these methods provide sensitivity in the range of attomoles, which is comparable to the sensitivity of radioactive sequencing methods.


In many capillary electrophoresis methods, the capillaries e.g. fused silica capillaries or channels etched, machined or molded into planar substrates, are filled with an appropriate separation/sieving matrix. Typically, a variety of sieving matrices are known in the art may be used in the microcapillary arrays. Examples of such matrices include, e.g. hydroxyethyl cellulose, polyacrylamide and agarose. Gel matrices may be introduced and polymerized within the capillary channel. However, in some cases this may result in entrapment of bubbles within the channels which can interfere with sample separations. Accordingly, it is often desirable to place a preformed separation matrix within the capillary channel(s), prior to mating the planar elements of the capillary portion. Fixing the two parts, e.g. through sonic welding, permanently fixes the matrix within the channel. Polymerization outside of the channels helps to ensure that no bubbles are formed. Further, the pressure of the welding process helps to ensure a void-free system.


In addition to its use in nucleic acid “fingerprinting” and other sized based analyses the capillary arrays may also be used in sequencing applications. In particular, gel based sequencing techniques may be readily adapted for capillary array electrophoresis.


In addition to detection of mRNA or as the sole detection method expression products from the genes discussed above may be detected as indications of the biological condition of the tissue. Expression products may be detected in either the tissue sample as such, or in a body fluid sample, such as blood, serum, plasma, faeces, mucus, sputum, cerebrospinal fluid, and/or urine of the individual.


The expression products, peptides and proteins, may be detected by any suitable technique known to the person skilled in the art.


In a preferred embodiment the expression products are detected by means of specific antibodies directed to the various expression products, such as immunofluorescent and/or immunohistochemical staining of the tissue.


Immunohistochemical localization of expressed proteins may be carried out by immunostaining of tissue sections from the single tumors to determine which cells expressed the protein encoded by the transcript in question. The transcript levels may be used to select a group of proteins supposed to show variation from sample to sample making a rough correlation between the level of protein detected and the intensity of the transcript on the microarray possible.


For example sections may be cut from paraffin-embedded tissue blocks, mounted, and deparaffinized by incubation at 80° C. for 10 min. followed by immersion in heated oil at 60° C. for 10 min. (Estisol 312, Estichem A/S, Denmark) and rehydration. Antigen retrieval is achieved in TEG (TrisEDTA-Glycerol) buffer using microwaves at 900 W. The tissue sections may be cooled in the buffer for 15 min before a brief rinse in tap water. Endogenous peroxidase activity is blocked by incubating the sections with 1% H2O2 for 20 min. followed by three rinses in tap water, 1 min each. The sections may then be soaked in PBS buffer for 2 min. The next steps can be modified from the descriptions given by Oncogene Science Inc., in the Mouse Immunohistochemistry Detection System, XHCO1 (UniTect, Uniondale, N.Y., USA). Briefly, the tissue sections are incubated overnight at 4° C. with primary antibody (against beta-2 microglobulin (Dako), cytokeratin 8, cystatin-C (both from Europa, US), junB, CD59, E-cadherin, apo-E, cathepsin E, vimentin, IGFII (all from Santa Cruz), followed by three rinses in PBS buffer for 5 min each. Afterwards, the sections are incubated with biotinylated secondary antibody for 30 min, rinsed three times with PBS buffer and subsequently incubated with ABC (avidin-biotinlylated horseradish peroxidase complex) for 30 min. followed by three rinses in PBS buffer.


Staining may be performed by incubation with AEC (3-amino-ethylcarbazole) for 10 min. The tissue sections are counter stained with Mayers hematoxylin, washed in tap water for 5 min. and mounted with glycerol-gelatin. Positive and negative controls may be included in each staining round with all antibodies.


In yet another embodiment the expression products may be detected by means of conventional enzyme assays, such as ELISA methods.


Furthermore, the expression products may be detected by means of peptide/protein chips capable of specifically binding the peptides and/or proteins assessed. Thereby an expression pattern may be obtained.


Assay

In a further aspect the invention relates to an assay for predicting the prognosis of a biological condition in animal tissue, comprising

    • at least one first marker capable of detecting an expression level of at least one gene selected from the group of genes consisting of gene No. 1 to gene No. 562.


Preferably the assay further comprises means for correlating the expression level to at least one standard expression level and/or at least one reference pattern.


The means for correlating preferably includes one or more standard expression levels and/or reference patterns for use in comparing or correlating the expression levels or patterns obtained from a tumor under examination to the standards.


Preferably the invention relates to an assay for determining an expression pattern of a bladder cell, comprising at least a first marker and/or a second marker, wherein the first marker is capable of detecting a gene from a first gene group as defined above, and/or the second marker is capable of detecting a gene from a second gene group as defined above, correlating the first expression level and/or the second expression level to a standard level of the assessed genes to predict the prognosis of a biological condition in the animal tissue. The marker(s) are preferably specifically detecting a gene as identified herein.


As described above, it is preferred to determine the expression level from more than one gene, and correspondingly, it is preferred to include more than one marker in the assay, such as at least two markers, such as at least three markers, such as at least four markers, such as at least five markers, such as at least six markers, such as at least seven markers, such as at least eight markers, such as at least nine markers, such as at least ten markers, such as at least 15 markers.


When using markers for at least two different groups, it is preferred that the above number of markers relate to markers in each group.


As discussed above the marker may be any nucleotide probe, such as a DNA, RNA, PNA, or LNA probe capable of hybridising to mRNA indicative of the expression level. The hybridisation conditions are preferably as described below for probes. In another embodiment the marker is an antibody capable of specifically binding the expression product in question.


Patterns can be compared manually by a person or by a computer or other machine. An algorithm can be used to detect similarities and differences. The algorithm may score and compare, for example, the genes which are expressed and the genes which are not expressed. Alternatively, the algorithm may look for changes in intensity of expression of a particular gene and score changes in intensity between two samples. Similarities may be determined on the basis of genes which are expressed in both samples and genes which are not expressed in both samples or on the basis of genes whose intensity of expression are numerically similar.


Generally, the detection operation will be performed using a reader device external to the diagnostic device. However, it may be desirable in some cases to incorporate the data gathering operation into the diagnostic device itself.


The detection apparatus may be a fluorescence detector, or a spectroscopic detector, or another detector.


Although hybridization is one type of specific interaction which is clearly useful for use in this mapping embodiment antibody reagents may also be very useful.


Gathering data from the various analysis operations, e.g. oligonucleotide and/or microcapillary arrays will typically be carried out using methods known in the art. For example, the arrays may be scanned using lasers to excite fluorescently labeled targets that have hybridized to regions of probe arrays mentioned above, which can then be imaged using charged coupled devices (“CCDs”) for a wide field scanning of the array. Alternatively, another particularly useful method for gathering data from the arrays is through the use of laser confocal microscopy which combines the ease and speed of a readily automated process with high resolution detection.


Following the data gathering operation, the data will typically be reported to a data analysis operation. To facilitate the sample analysis operation, the data obtained by the reader from the device will typically be analyzed using a digital computer. Typically, the computer will be appropriately programmed for receipt and storage of the data from the device, as well as for analysis and reporting of the data gathered, i.e., interpreting fluorescence data to determine the sequence of hybridizing probes, normalization of background and single base mismatch hybridizations, ordering of sequence data in SBH applications, and the like.


The invention also relates to a pharmaceutical composition for treating a biological condition, such as bladder tumors.


In one embodiment the pharmaceutical composition comprises one or more of the peptides being expression products as defined above. In a preferred embodiment, the peptides are bound to carriers. The peptides may suitably be coupled to a polymer carrier, for example a protein carrier, such as BSA. Such formulations are well-known to the person skilled in the art.


The peptides may be suppressor peptides normally lost or decreased in tumor tissue administered in order to stabilise tumors towards a less malignant stage. In another embodiment the peptides are onco-peptides capable of eliciting an immune response towards the tumor cells.


In another embodiment the pharmaceutical composition comprises genetic material, either genetic material for substitution therapy, or for suppressing therapy as discussed below.


In a third embodiment the pharmaceutical composition comprises at least one antibody produced as described above.


In the present context the term pharmaceutical composition is used synonymously with the term medicament. The medicament of the invention comprises an effective amount of one or more of the compounds as defined above, or a composition as defined above in combination with pharmaceutically acceptable additives. Such medicament may suitably be formulated for oral, percutaneous, intramuscular, intravenous, intracranial, intrathecal, intracerebroventricular, intranasal or pulmonal administration. For most indications a localised or substantially localised application is preferred.


Strategies in formulation development of medicaments and compositions based on the compounds of the present invention generally correspond to formulation strategies for any other protein-based drug product. Potential problems and the guidance required to overcome these problems are dealt with in several textbooks, e.g. “Therapeutic Peptides and Protein Formulation. Processing and Delivery Systems”, Ed. A. K. Banga, Technomic Publishing AG, Basel, 1995.


Injectables are usually prepared either as liquid solutions or suspensions, solid forms suitable for solution in, or suspension in, liquid prior to injection. The preparation may also be emulsified. The active ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like, and combinations thereof. In addition, if desired, the preparation may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or which enhance the effectiveness or transportation of the preparation.


Formulations of the compounds of the invention can be prepared by techniques known to the person skilled in the art. The formulations may contain pharmaceutically acceptable carriers and excipients including microspheres, liposomes, microcapsules and nanoparticles.


The preparation may suitably be administered by injection, optionally at the site, where the active ingredient is to exert its effect. Additional formulations which are suitable for other modes of administration include suppositories, and in some cases, oral formulations. For suppositories, traditional binders and carriers include polyalkylene glycols or triglycerides. Such suppositories may be formed from mixtures containing the active ingredient(s) in the range of from 0.5% to 10%, preferably 1-2%. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and generally contain 10-95% of the active ingredient(s), preferably 25-70%.


The preparations are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective. The quantity to be administered depends on the subject to be treated, including, e.g. the weight and age of the subject, the disease to be treated and the stage of disease. Suitable dosage ranges are of the order of several hundred μg active ingredient per administration with a preferred range of from about 0.1 μg to 1000 μg, such as in the range of from about 1 μg to 300 μg, and especially in the range of from about 10 μg to 50 μg. Administration may be performed once or may be followed by subsequent administrations. The dosage will also depend on the route of administration and will vary with the age and weight of the subject to be treated. A preferred dosis would be in the interval 30 mg to 70 mg per 70 kg body weight.


Some of the compounds of the present invention are sufficiently active, but for some of the others, the effect will be enhanced if the preparation further comprises pharmaceutically acceptable additives and/or carriers. Such additives and carriers will be known in the art. In some cases, it will be advantageous to include a compound, which promote delivery of the active substance to its target.


In many instances, it will be necessary to administrate the formulation multiple times. Administration may be a continuous infusion, such as intraventricular infusion or administration in more doses such as more times a day, daily, more times a week, weekly, etc.


Vaccines

In a further embodiment the present invention relates to a vaccine for the prophylaxis or treatment of a biological condition comprising at least one expression product from at least one gene said gene being expressed as defined above.


The term vaccines is used with its normal meaning, i.e preparations of immunogenic material for administration to induce in the recipient an immunity to infection or intoxication by a given infecting agent. Vaccines may be administered by intravenous injection or through oral, nasal and/or mucosal administration. Vaccines may be either simple vaccines prepared from one species of expression products, such as proteins or peptides, or a variety of expression products, or they may be mixed vaccines containing two or more simple vaccines. They are prepared in such a manner as not to destroy the immunogenic material, although the methods of preparation vary, depending on the vaccine.


The enhanced immune response achieved according to the invention can be attributable to e.g. an enhanced increase in the level of immunoglobulins or in the level of T-cells including cytotoxic T-cells will result in immunisation of at least 50% of individuals exposed to said immunogenic composition or vaccine, such as at least 55%, for example at least 60%, such as at least 65%, for example at least 70%, for example at least 75%, such as at least 80%, for example at least 85%, such as at least 90%, for example at least 92%, such as at least 94%, for example at least 96%, such as at least 97%, for example at least 98%, such as at least 98.5%, for example at least 99%, for example at least 99.5% of the individuals exposed to said immunogenic composition or vaccine are immunised.


Compositions according to the invention may also comprise any carrier and/or adjuvant known in the art including functional equivalents thereof. Functionally equivalent carriers are capable of presenting the same immunogenic determinant in essentially the same steric conformation when used under similar conditions. Functionally equivalent adjuvants are capable of providing similar increases in the efficacy of the composition when used under similar conditions.


Therapy

The invention further relates to a method of treating individuals suffering from the biological condition in question, in particular for treating a bladder tumor.


Accordingly, the invention relates to a method for reducing cell tumorigenicity or malignancy of a cell, said method comprising contacting a tumor cell with at least one peptide expressed by at least one gene selected from the group of genes consisting of gene No. 200-214, 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, 446, 453, 460, 461, 463, 464, 465, 466, 467, 469, 470, 471, 472, 473, 475, 476, 477, 479, 480, 481, 482, 483, 485, 486, 487, 488, 490, 492, 494, 496, 497, 498, 499, 503, 515, 516, 517, 521, 526, 527, 528, 530, 532, 533, 537, 539, 540, 541, 542, 543, 545, 554, 557, 560.


In order to increase the effect several different peptides may be used simultaneously, such as wherein the tumor cell is contacted with at least two different peptides.


In one embodiment the invention relates to a method of substitution therapy, ie. administration of genetic material generally expressed in normal cells, but lost or decreased in biological condition cells (tumor suppressors). Thus, the invention relates to a method for reducing cell tumorigenicity or malignancy of a cell, said method comprising


obtaining at least one gene selected from the group of genes consisting of gene No. 200-214, 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, 446, 453, 460, 461, 463, 464, 465, 466, 467, 469, 470, 471, 472, 473, 475, 476, 477, 479, 480, 481, 482, 483, 485, 486, 487, 488, 490, 492, 494, 496, 497, 498, 499, 503, 515, 516, 517, 521, 526, 527, 528, 530, 532, 533, 537, 539, 540, 541, 542, 543, 545, 554, 557, 560,


introducing said at least one gene into the tumor cell in a manner allowing expression of said gene(s).


In one embodiment at least one gene is introduced into the tumor cell. In another embodiment at least two genes are introduced into the tumor cell.


In one aspect of the invention small molecules that either inhibit increased gene expression or their effects or substitute decreased gene expression or their effects, are introduced to the cellular environment or the cells. Application of small molecules to tumor cells may be performed by e.g. local application or intravenous injection or by oral ingestion. Small molecules have the ability to restore function of reduced gene expression in tumor or cancer tissue.


In another aspect the invention relates to a therapy whereby genes (increase and/or decrease) generally are correlated to disease are inhibited by one or more of the following methods:


A method for reducing cell tumorigenicity or malignancy of a cell, said method comprising


obtaining at least one nucleotide probe capable of hybridising with at least one gene of a tumor cell, said at least one gene being selected from the group of genes consisting of gene Nos. 1-199, 215-232, 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 369, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, 444, 447, 448, 449, 450, 451, 452, 454, 455, 456, 457, 458, 459, 462, 468, 474, 478, 484, 489, 491, 493, 495, 500, 501, 502, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 518, 519, 520, 522, 523, 524, 525, 529, 531, 534, 535, 536, 538, 544, 546, 547, 548, 549, 550, 551, 552, 553, 555, 556, 558, 559, 561, 562,


introducing said at least one nucleotide probe into the tumor cell in a manner allowing the probe to hybridise to the at least one gene, thereby inhibiting expression of said at least one gene. This method is preferably based on anti-sense technology, whereby the hybridisation of said probe to the gene leads to a down-regulation of said gene.


In another preferred embodiment, the method for reducing cell tumorigenicity or malignancy of a cell is based on RNA interference, comprising small interfering RNAs (siRNAs) specifically directed against at least one gene being selected from the group of genes consisting of gene Nos. 1-199, 215-232, 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 369, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, 444, 447, 448, 449, 450, 451, 452, 454, 455, 456, 457, 458, 459, 462, 468, 474, 478, 484, 489, 491, 493, 495, 500, 501, 502, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 518, 519, 520, 522, 523, 524, 525, 529, 531, 534, 535, 536, 538, 544, 546, 547, 548, 549, 550, 551, 552, 553, 555, 556, 558, 559, 561, 562.


The down-regulation may of course also be based on a probe capable of hybridising to regulatory components of the genes in question, such as promoters.


The hybridization may be tested in vitro at conditions corresponding to in vivo conditions. Typically, hybridization conditions are of low to moderate stringency. These conditions favour specific interactions between completely complementary sequences, but allow some non-specific interaction between less than perfectly matched sequences to occur as well. After hybridization, the nucleic acids can be “washed” under moderate or high conditions of stringency to dissociate duplexes that are bound together by some non-specific interaction (the nucleic acids that form these duplexes are thus not completely complementary).


As is known in the art, the optimal conditions for washing are determined empirically, often by gradually increasing the stringency. The parameters that can be changed to affect stringency include, primarily, temperature and salt concentration. In general, the lower the salt concentration and the higher the temperature the higher the stringency. Washing can be initiated at a low temperature (for example, room temperature) using a solution containing a salt concentration that is equivalent to or lower than that of the hybridization solution. Sub-sequent washing can be carried out using progressively warmer solutions having the same salt concentration. As alternatives, the salt concentration can be lowered and the temperature maintained in the washing step, or the salt concentration can be lowered and the temperature increased. Additional parameters can also be altered. For example, use of a destabilizing agent, such as formamide, alters the stringency conditions.


In reactions where nucleic acids are hybridized, the conditions used to achieve a given level of stringency will vary. There is not one set of conditions, for example, that will allow duplexes to form between all nucleic acids that are 85% identical to one another; hybridization also depends on unique features of each nucleic acid. The length of the sequence, the composition of the sequence (for example, the content of purine-like nucleotides versus the content of pyrimidine-like nucleotides) and the type of nucleic acid (for example, DNA or RNA) affect hybridization. An additional consideration is whether one of the nucleic acids is immobilized (for example on a filter).


An example of a progression from lower to higher stringency conditions is the following, where the salt content is given as the relative abundance of SSC (a salt solution containing sodium chloride and sodium citrate; 2×SSC is 10-fold more concentrated than 0.2×SSC). Nucleic acids are hybridized at 42° C. in 2×SSC/0.1% SDS (sodium dodecylsulfate; a detergent) and then washed in 0.2×SSC/0.1% SDS at room temperature (for conditions of low stringency); 0.2×SSC/0.1% SDS at 42° C. (for conditions of moderate stringency); and 0.1×SSC at 68° C. (for conditions of high stringency). Washing can be carried out using only one of the conditions given, or each of the conditions can be used (for example, washing for 10-15 minutes each in the order listed above). Any or all of the washes can be repeated. As mentioned above, optimal conditions will vary and can be determined empirically.


In another aspect a method of reducing tumoregeneicity relates to the use of antibodies against an expression product of a cell from the biological tissue. The antibodies may be produced by any suitable method, such as a method comprising the steps of


obtaining expression product(s) from at least one gene said gene being expressed as defined above,


immunising a mammal with said expression product(s) obtaining antibodies against the expression product.


Use

The methods described above may be used for producing an assay for diagnosing a biological condition in animal tissue, or for identification of the origin of a piece of tissue. Further, the methods of the invention may be used for prediction of a disease course and treatment response.


Furthermore, the invention relates to the use of a peptide as defined above for preparation of a pharmaceutical composition for the treatment of a biological condition in animal tissue.


Furthermore, the invention relates to the use of a gene as defined above for preparation of a pharmaceutical composition for the treatment of a biological condition in animal tissue.


Also, the invention relates to the use of a probe as defined above for preparation of a pharmaceutical composition for the treatment of a biological condition in animal tissue.


The genetic material discussed above for may be any of the described genes or functional parts thereof. The constructs may be introduced as a single DNA molecule encoding all of the genes, or different DNA molecules having one or more genes. The constructs may be introduced simultaneously or consecutively, each with the same or different markers.


The gene may be linked to the complex as such or protected by any suitable system normally used for transfection such as viral vectors or artificial viral envelope, liposomes or micellas, wherein the system is linked to the complex.


Numerous techniques for introducing DNA into eukaryotic cells are known to the skilled artisan. Often this is done by means of vectors, and often in the form of nucleic acid encapsidated by a (frequently virus-like) proteinaceous coat. Gene delivery systems may be applied to a wide range of clinical as well as experimental applications.


Vectors containing useful elements such as selectable and/or amplifiable markers, promoter/enhancer elements for expression in mammalian, particularly human, cells, and which may be used to prepare stocks of construct DNAs and for carrying out transfections are well known in the art. Many are commercially available.


Various techniques have been developed for modification of target tissue and cells in vivo. A number of virus vectors, discussed below, are known which allow transfection and random integration of the virus into the host. See, for example, Dubensky et al. (1984) Proc. Natl. Acad. Sci. USA 81:7529-7533; Kaneda et al., (1989) Science 243:375-378; Hiebert et al. (1989) Proc. Natl. Acad. Sci. USA 86:3594-3598; Hatzoglu et al., (1990) J. Biol. Chem. 265:17285-17293; Ferry et al. (1991) Proc. Natl. Acad. Sci. USA 88:8377-8381. Routes and modes of administering the vector include injection, e.g intravascularly or intramuscularly, inhalation, or other parenteral administration.


Advantages of adenovirus vectors for human gene therapy include the fact that recombination is rare, no human malignancies are known to be associated with such viruses, the adenovirus genome is double stranded DNA which can be manipulated to accept foreign genes of up to 7.5 kb in size, and live adenovirus is a safe human vaccine organisms.


Another vector which can express the DNA molecule of the present invention, and is useful in gene therapy, particularly in humans, is vaccinia virus, which can be rendered non-replicating (U.S. Pat. Nos. 5,225,336; 5,204,243; 5,155,020; 4,769,330).


Based on the concept of viral mimicry, artificial viral envelopes (AVE) are designed based on the structure and composition of a viral membrane, such as HIV-1 or RSV and used to deliver genes into cells in vitro and in vivo. See, for example, U.S. Pat. No. 5,252,348, Schreier H. et al., J. Mol. Recognit., 1995, 8:59-62; Schreier H et al., J. Biol. Chem., 1994, 269:9090-9098; Schreier, H., Pharm. Acta Helv. 1994, 68:145-159; Chander, R et al. Life Sci., 1992, 50:481-489, which references are hereby incorporated by reference in their entirety. The envelope is preferably produced in a two-step dialysis procedure where the “naked” envelope is formed initially, followed by unidirectional insertion of the viral surface glycoprotein of interest. This process and the physical characteristics of the resulting AVE are described in detail by Chander et al., (supra). Examples of AVE systems are (a) an AVE containing the HIV-1 surface glycoprotein gp160 (Chander et al., supra; Schreier et al., 1995, supra) or glycosyl phosphatidylinositol (GPI)-linked gp120 (Schreier et al., 1994, supra), respectively, and (b) an AVE containing the respiratory syncytial virus (RSV) attachment (G) and fusion (F) glycoproteins (Stecenko, A. A. et al., Pharm. Pharmacol. Lett. 1:127-129 (1992)). Thus, vesicles are constructed which mimic the natural membranes of enveloped viruses in their ability to bind to and deliver materials to cells bearing corresponding surface receptors.


AVEs are used to deliver genes both by intravenous injection and by instillation in the lungs. For example, AVEs are manufactured to mimic RSV, exhibiting the RSV F surface glycoprotein which provides selective entry into epithelial cells. F-AVE are loaded with a plasmid coding for the gene of interest, (or a reporter gene such as CAT not present in mammalian tissue).


The AVE system described herein in physically and chemically essentially identical to the natural virus yet is entirely “artificial”, as it is constructed from phospholipids, cholesterol, and recombinant viral surface glycoproteins. Hence, there is no carry-over of viral genetic information and no danger of inadvertant viral infection. Construction of the AVEs in two independent steps allows for bulk production of the plain lipid envelopes which, in a separate second step, can then be marked with the desired viral glycoprotein, also allowing for the preparation of protein cocktail formulations if desired.


Another delivery vehicle for use in the present invention are based on the recent description of attenuated Shigella as a DNA delivery system (Sizemore, D. R. et al., Science 270:299-302 (1995), which reference is incorporated by reference in its entirety). This approach exploits the ability of Shigellae to enter epithelial cells and escape the phagocytic vacuole as a method for delivering the gene construct into the cytoplasm of the target cell. Invasion with as few as one to five bacteria can result in expression of the foreign plasmid DNA delivered by these bacteria.


A preferred type of mediator of nonviral transfection in vitro and in vivo is cationic (ammonium derivatized) lipids. These positively charged lipids form complexes with negatively charged DNA, resulting in DNA charged neutralization and compaction. The complexes endocytosed upon association with the cell membrane, and the DNA somehow escapes the endosome, gaining access to the cytoplasm. Cationic lipid:DNA complexes appear highly stable under normal conditions. Studies of the cationic lipid DOTAP suggest the complex dissociates when the inner layer of the cell membrane is destabilized and anionic lipids from the inner layer displace DNA from the cationic lipid. Several cationic lipids are available commercially. Two of these, DMRI and DC-cholesterol, have been used in human clinical trials. First generation cationic lipids are less efficient than viral vectors. For delivery to lung, any inflammatory responses accompanying the liposome administration are reduced by changing the delivery mode to aerosol administration which distributes the dose more evenly.


Drug Screening

Genes identified as changing in various stages of bladder cancer can be used as markers for drug screening. Thus by treating bladder cancer cells with test compounds or extracts, and monitoring the expression of genes identified as changing in the progression of bladder cancers, one can identify compounds or extracts which change expression of genes to a pattern which is of an earlier stage or even of normal bladder mucosa.


It is also within the scope of the invention to use small molecules in drug screening.


The following are non-limiting examples illustrating the present invention.


EXAMPLES
Example 1
Identification of a Molecular Signature Defining Disease Progression in Patients with Superficial Bladder Carcinoma
Patient Samples

Bladder tumor biopsies were obtained directly from surgery after removal of the necessary amount of tissue for routine pathology examination. The tumors were frozen at −80° C. in a guanidinium thiocyanate solution for preservation of the RNA. Informed consent was obtained in all cases, and the protocols were approved by the scientific ethical committee of Aarhus County. The samples for the no progression group were selected by the following criteria: a) Ta or T1 tumors with no prior higher stage tumors; b) a minimum follow up period of 12 months to the most recent routine cystoscopy examination of the bladder with no occurrence of tumors of higher stage. The samples for the progression group were selected by two criteria: a) Ta or T1 tumors with no prior higher stage tumors; b) subsequent progression to a higher stage tumor, see Table 1.









TABLE 1







Clinical data on all patients involved in the study

















Follow-





Progressed
Time to
up time


Group
Sample
Hist.
to:
progression
months










Training set












No prog.
150-6
Ta gr3


44


No prog.
997-1
Ta gr2


24


No prog.
833-2
Ta gr3


35


No prog.
1070-1 
Ta gr3


33


No prog.
968-1
Ta gr2


26


No prog.
625-1
T1 gr3


12


No prog.
880-1
T1 gr3


47


No prog.
815-1
Ta gr2


49


No prog.
861-1
Ta gr2


45


No prog.
669-1
Ta gr2


55


No prog.
368-4
Ta gr2


16


No prog.
898-1
Ta gr2


17


No prog.
576-6
Ta gr2


36


Prog.
747-3
Ta gr2
T1 gr3
6


Prog.
956-2
Ta gr3
T1 gr3
27



Prog.
1083-1 
Ta gr2
T1 gr3
1



Prog.
686-3
Ta gr2
T1 gr2
6



Prog.
 795-13
Ta gr2
T1 gr3
4



Prog.
865-1
Ta gr2
T1 gr2
5



Prog.
112-2
Ta gr3
T1 gr3
7



Prog.
825-3
Ta gr3
T1 gr3
6



Prog.
679-2
Ta gr2
T2+ gr3
31



Prog.
941-4
Ta gr3
T2+ gr3
10



Prog.
607-1
T1 gr2
T2+ gr3
3



Prog.
1017-1 
T1 gr3
T2+ gr3
8



Prog.
1276-1 
T1 gr3
T2+ gr3
7



Prog.
501-1
T1 gr3
T2+ gr3
26



Prog.
744-1
T1 gr3
T2+ gr3
14



Prog.
839-1
T1 gr3
T2+ gr3
12








Test set












No prog.
1008-1 
Ta gr2


55


No prog.
1060-1 
Ta gr2


48


No prog.
1086-1 
Ta gr2


34


No prog.
1105-1 
Ta gr2


31


No prog.
1145-1 
Ta gr2


39


No prog.
1352-1 
Ta gr2


26


No prog.
829-1
Ta gr2


37


No prog.
942-1
Ta gr2


37


No prog.
780-1
Ta gr2


50


Prog
1327-1 
Ta gr2
T1 gr3
8


Prog.
1062-2 
Ta gr3
T1 gr3
4



Prog.
1354-1 
Ta gr3
T1 gr3
8



Prog.
1093-1 
Ta gr3
T1 gr3
5



Prog.
925-7
Ta gr2
T1 gr3
4



Prog.
 962-10
Ta gr0
T2+ gr3
1



Prog.
970-1
Ta gr3
T2+ gr3
1



Prog.
1027-1 
Ta gr3
T2+ gr3
2



Prog.
1252-1 
T1 gr3
T2+ gr3
5



Prog.
1191-1 
T1 gr4
T2+ gr4
1











Delineation of Non-Progressing Tumors from Progressing Tumors


To delineate non-progressing tumors from progressing tumors we now profiled a total of 29 bladder tumor samples; 13 early stage bladder tumor samples without progression (median follow-up time 35 months) and 16 early stage bladder tumor samples with progression (median time to progression 7 months). See Table 1 for description of patient disease courses. We analyzed gene expression changes between the two groups of tumors by hybridizing the labeled RNA samples to customized Affymetrix GeneChips with 59,000 probe-sets to cover virtually the entire transcriptome (˜95% coverage). Low expressed and non-varying probesets were eliminated from the data set and the resulting 6,647 probe-sets that showed variation across the tumor samples were subjected to further analysis. These probe-sets represent 5,356 unique genes (Unigene clusters).


Gene Expression Similarities Between Tumor Biopsies

We analyzed gene expression similarities between the tumor biopsies using unsupervised hierarchical cluster analysis (FIG. 1). This showed a notable distinction between the non-progressing and the progressing tumors when using the 3,197 most varying probe-sets (s.d. ≧75) for clustering (4 errors; χ2 test, P=0.0001). Using other gene-sets based on different gene variation criteria demonstrated the same distinction between the tumor groups. Two of the samples that show later progression (825-3 and 112-2) were found in the non-progression branch of the cluster dendrogram and two of the non-progressing samples (815-1 and 150-6) were found in the progression branch. This distinct separation of the samples indicated a considerable biological difference between the two groups of tumors. Notably, the T1 tumors did not cluster separately from Ta tumors; however, they did form a sub-cluster in the progressing branch of the dendrogram. Based on this we decided to look for a general signature of progression disregarding pathologic staging of the tumors.


Selection of the 100 Most Significantly Up-Regulated Genes in Each Group Using T-Test Statistics

We delineated the non-progressing tumors from the progressing tumors by selecting the 100 most significantly up-regulated genes in each group using t-test statistics (FIG. 2 and Table 2). Among the genes up regulated in the non-progressing group we found the SERPINB5 and FAT tumor suppressor genes and the FGFR3 gene, which has been shown to be frequently mutated in superficial bladder tumors with low recurrence rates (van Rhijn et al. 2001). Among the genes up regulated in the progressing group we found the PLK (Yuan et al. 1997), CDC25B (Galaktionov et al. 1991), CDC20 (Weinstein et al. 1994) and MCM7 (Hiraiwa et al. 1997) genes, which are involved in regulating cell cycle and cell proliferation. Furthermore, in this group we identified the WHSC1, DD96 and GRB7 genes, which have been predicted/computed (Gene Ontology) to be involved in oncogenic transformation. Another interesting candidate in this group is the NRG1 gene, which through interaction with the HER2/HER3 receptors has been found to induce differentiation of lung epithelial cells (Liu & Kern 2002). The PPARD gene was also identified as up regulated in the tumors that show later progression. Disruption of this gene was found to decrease tumorigenicity in colon cancer cells (Park et al. 2001). Furthermore, PPARD regulates VEGF expression in bladder cancer cell lines (Fauconnet et al. 2002).









TABLE 2







The 200 best markers of progression












Eos
Unigene



Exemplar


Hu03 ID
Build 133
Description
T-test
5% perm
accession#















416640
Hs.79404
neuron-specific protein
6.03
5.62
BE262478


442220
Hs.8148
selenoprotein T
5.98
5.06
AL037800


426982
Hs.173091
ubiquitin-like 3
5.9
4.88
AA149707


416815
Hs.80120
UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-
5.52
4.67
U41514




acetylgalactosaminyltransferase 1 (GalNAc-T1)


435521
Hs.6361
mitogen-activated protein kinase kinase 1 interacting
5.24
4.51
W23814




protein 1


447343
Hs.236894
ESTs, Highly similar to S02392 alpha-2-macroglobulin
5.23
4.44
AA256641




receptor precursor [H. sapiens]


452829
Hs.63368
ESTs, Weakly similar to TRHY_HUMAN TRICHOHYALI
4.95
4.39
AI955579




[H. sapiens]


414895
Hs.116278

Homo sapiens cDNA FLJ13571 fis, clone

4.94
4.31
AW894856




PLACE1008405


426252
Hs.28917
ESTs
4.9
4.26
BE176980


444604
Hs.11441
chromosome 1 open reading frame 8
4.89
4.17
AW327695


409632
Hs.55279
serine (or cysteine) proteinase inhibitor, clade B
4.89
4.13
W74001




(ovalbumin), member 5


446556
Hs.15303
KIAA0349 protein
4.87
4.08
AB002347


426799
Hs.303154
popeye protein 3
4.86
4.03
H14843


428115
Hs.300855
KIAA0977 protein
4.86
4.00
AB023194


419847
Hs.184544
Homo sapiens, clone IMAGE: 3355383, mRNA, partial
4.82
3.97
AW390601




cds


417839
Hs.82712
fragile X mental retardation, autosomal homolog 1
4.8
3.93
AI815732


428284
Hs.183435
NM_004545: Homo sapiens NADH dehydrogenase
4.78
3.92
AA535762




(ubiquinone) 1 beta subcomplex, 1 (7 kD, MNLL)




(NDUFB1), mRNA.


422929
Hs.94011
ESTs, Weakly similar to MGB4_HUMAN MELANOMA-
4.77
3.90
AA356694




ASSOCIATED ANTIGEN B4 [H. sapiens]


414762
Hs.77257
KIAA0068 protein
4.72
3.86
AW068349


453395
Hs.377915
mannosidase, alpha, class 2A, member 1
4.71
3.84
D63998


421311
Hs.283609
hypothetical protein PRO2032
4.65
3.82
N71848


446847
Hs.82845

Homo sapiens cDNA: FLJ21930 fis, clone HEP04301,

4.65
3.82
T51454




highly similar to HSU90916 Human clone 23815




mRNA sequence


413840
Hs.356228
RNA binding motif protein, X chromosome
4.62
3.79
AI301558


418321
Hs.84087
KIAA0143 protein
4.62
3.78
D63477


430604
Hs.247309
succinate-CoA ligase, GDP-forming, beta subunit
4.61
3.74
AV650537


423185
Hs.380062
ornithine decarboxylase antizyme 1
4.61
3.74
BE299590


417615
Hs.82314
hypoxanthine phosphoribosyltransferase 1 (Lesch-
4.6
3.70
BE548641




Nyhan syndrome)


418504
Hs.85335

Homo sapiens mRNA; cDNA DKFZp564D1462 (from

4.59
3.68
BE159718




clone DKFZp564D1462)


400846

sortilin-related receptor, L(DLR class) A repeats-
4.57
3.66





containing (SORL1)


426028
Hs.172028
a disintegrin and metalloproteinase domain 10
4.53
3.65
NM_001110




(ADAM10)


425243
Hs.155291
KIAA0005 gene product
4.47
3.63
N89487


434978
Hs.4310
eukaryotic translation initiation factor 1A
4.45
3.62
AA321238


409513
Hs.54642
methionine adenosyltransferase II, beta
4.43
3.59
AW966728


433282
Hs.49007
hypothetical protein
4.43
3.56
BE539101


421628
Hs.106210
hypothetical protein FLJ10813
4.37
3.56
AL121317


452170
Hs.28285
patched related protein translocated in renal cancer
4.37
3.54
AF064801


440014
Hs.6856
ash2 (absent, small, or homeotic, Drosophila, homolog)-
4.37
3.52
AW960782




like


431857
Hs.271742
ADP-ribosyltransferase (NAD; poly (ADP-ribose)
4.36
3.52
W19144




polymerase)-like 3


417924
Hs.82932
cyclin D1 (PRAD1: parathyroid adenomatosis 1)
4.35
3.51
AU077231


421733
Hs.1420
fibroblast growth factor receptor 3 (achondroplasia,
4.34
3.50
AL119671




thanatophoric dwarfism)


440197
Hs.317714
pallid (mouse) homolog, pallidin
4.32
3.49
AW340708


434055
Hs.3726
x 003 protein
4.32
3.48
AF168712


445831
Hs.13351
LanC (bacterial lantibiotic synthetase component C)-
4.31
3.46
NM_006055




like 1


439632
Hs.334437
hypothetical protein MGC4248
4.29
3.45
AW410714


448813
Hs.22142
cytochrome b5 reductase b5R.2
4.28
3.44
AF169802


449268
Hs.23412
hypothetical protein FLJ20160
4.28
3.43
AW369278


429311
Hs.198998
conserved helix-loop-helix ubiquitous kinase
4.28
3.42
AF080157


423599
Hs.31731
peroxiredoxin 5
4.27
3.41
AI805664


422913
Hs.121599
CGI-18 protein
4.26
3.40
NM_015947


418127
Hs.83532
membrane cofactor protein (CD46, trophoblast-
4.26
3.39
BE243982




lymphocyte cross-reactive antigen)


425221
Hs.155188
TATA box binding protein (TBP)-associated factor,
4.25
3.38
AV649864




RNA polymerase II, F, 55 kD


426682
Hs.2056
UDP glycosyltransferase 1 family, polypeptide A9
4.23
3.37
AV660038


421101
Hs.101840
major histocompatibility complex, class I-like sequence
4.23
3.37
AF010446


444037
Hs.380932
CHMP1.5 protein
4.22
3.35
AV647686


443407
Hs.348514
ESTs, Moderately similar to 2109260A B cell growth
4.21
3.35
AA037683




factor [H. sapiens]


448625
Hs.178470
hypothetical protein FLJ22662
4.21
3.34
AW970786


450997
Hs.35254
hypothetical protein FLB6421
4.16
3.34
AW580830


444336
Hs.10882
HMG-box containing protein 1
4.15
3.33
AF019214


416977
Hs.406103
hypothetical protein FKSG44
4.14
3.32
AW130242


420613
Hs.406637
ESTs, Weakly similar to A47582 B-cell growth factor
4.13
3.31
AI873871




precursor [H. sapiens]


414843
Hs.77492
heterogeneous nuclear ribonucleoprotein A0
4.1
3.30
BE386038


408288
Hs.16886
gb: zI73d06.r1 Stratagene colon (937204) Homo
4.09
3.29
AA053601





sapiens cDNA clone 5′, mRNA sequence



422043
Hs.110953
retinoic acid induced 1
4.09
3.29
AL133649


432864
Hs.359682
calpastatin
4.08
3.28
D16217


410047
Hs.379753
zinc finger protein 36 (KOX 18)
4.06
3.28
AI167810


400773

NM_003105*: Homo sapiens sortilin-related receptor,
4.06
3.27





L(DLR class) A repeats-containing (SORL1), mRNA.


423960
Hs.136309
SH3-containing protein SH3GLB1
4.05
3.27
AA164516


449626
Hs.112860
zinc finger protein 258
4.04
3.27
AA774247


429953
Hs.226581
COX15 (yeast) homolog, cytochrome c oxidase assembly
4.04
3.24
NM_004376




protein


428901
Hs.146668
KIAA1253 protein
4.02
3.24
AI929568


420079
Hs.94896
PTD011 protein
3.99
3.22
NM_014051


436576
Hs.77542
ESTs
3.98
3.21
AI458213


412841
Hs.101395
hypothetical protein MGC11352
3.97
3.21
AI751157


431604
Hs.264190
vacuolar protein sorting 35 (yeast homolog)
3.96
3.21
AF175265


428318
Hs.356190
ubiquitin B
3.96
3.19
BE300110


430677
Hs.359784
desmoglein 2
3.95
3.19
Z26317


407955
Hs.9343
ESTs
3.94
3.18
BE536739


426177
Hs.167700

Homo sapiens cDNA FLJ10174 fis, clone

3.92
3.17
AA373452




HEMBA1003959


429802
Hs.5367
ESTs, Weakly similar to I38022 hypothetical protein
3.92
3.17
H09548




[H. sapiens]


423810
Hs.132955
BCL2/adenovirus E1B 19 kD-interacting protein 3-like
3.92
3.16
AL132665


421475
Hs.104640
HIV-1 inducer of short transcripts binding protein;
3.91
3.15
AF000561




lymphoma related factor


436472
Hs.46366
KIAA0948 protein
3.91
3.14
AL045404


434263
Hs.79187
ESTs
3.9
3.13
N34895


400843

NM_003105*: Homo sapiens sortilin-related receptor,
3.9
3.13





L(DLR class) A repeats-containing (SORL1), mRNA.


440357
Hs.20950
phospholysine phosphohistidine inorganic pyrophosphate
3.89
3.12
AA379353




phosphatase


437223
Hs.330716

Homo sapiens cDNA FLJ14368 fis, clone

3.88
3.12
C15105




HEMBA1001122


426125
Hs.166994
FAT tumor suppressor (Drosophila) homolog
3.86
3.11
X87241


432554
Hs.278411
NCK-associated protein 1
3.86
3.10
AI479813


422506
Hs.300741
sorcin
3.85
3.10
R20909


413786
Hs.13500
ESTs
3.83
3.09
AW613780


429561
Hs.250646
baculoviral IAP repeat-containing 6
3.83
3.08
AF265555


404977

Insulin-like growth factor 2 (somatomedin A) (IGF2)
3.83
3.08



427722
Hs.180479
hypothetical protein FLJ20116
3.82
3.08
AK000123


400844

NM_003105*: Homo sapiens sortilin-related receptor,
3.82
3.08





L(DLR class) A repeats-containing (SORL1), mRNA.


426469
Hs.363039
methylmalonate-semialdehyde dehydrogenase
3.81
3.07
BE297886


439578
Hs.350547
nuclear receptor co-repressor/HDAC3 complex sub-
3.81
3.06
AW263124




unit


426508
Hs.170171
glutamate-ammonia ligase (glutamine synthase)
3.8
3.06
W23184


448524
Hs.21356
hypothetical protein DKFZp762K2015
3.79
3.06
AB032948


448357
Hs.108923
RAB38, member RAS oncogene family
3.79
3.06
N20169


425097
Hs.154545
PDZ domain containing guanine nucleotide exchange
3.77
3.05
NM_014247




factor(GEF)1


421649
Hs.106415
peroxisome proliferative activated receptor, delta
5.76
5.50
AA721217


427747
Hs.180655
serine/threonine kinase 12
5.41
5.03
AW411425


439010
Hs.75216

Homo sapiens cDNA FLJ13713 fis, clone

4.57
4.80
AW170332




PLACE2000398, moderately similar to LAR PROTEIN




PRECURSOR (LEUKOCYTE ANTIGEN RELATED)




(EC 3.1.3.48)


438818
Hs.30738
ESTs
4.49
4.59
AW979008


438013
Hs.15670
ESTs
4.42
4.50
AI002106


452929
Hs.172816
neuregulin 1
4.37
4.40
AW954938


404826

Target Exon
4.22
4.32



429124
Hs.196914
minor histocompatibility antigen HA-1
4.2
4.26
AW505086


421505
Hs.285641
KIAA1111 protein
4.16
4.24
AW249934


428712
Hs.190452
KIAA0365 gene product
4.14
4.19
AW085131


427239
Hs.356512
ubiquitin carrier protein
4.11
4.10
BE270447


421595
Hs.301685
KIAA0620 protein
4.1
4.07
AB014520


433844
Hs.179647

Homo sapiens cDNA FLJ12195 fis, clone

4.04
4.02
AA610175




MAMMA1000865


443679
Hs.9670
hypothetical protein FLJ10948
4.01
4.00
AK001810


422959
Hs.349256
paired immunoglobulin-like receptor beta
4.01
3.98
AV647015


452012
Hs.279766
kinesin family member 4A
3.98
3.96
AA307703


435320
Hs.117864
ESTs
3.97
3.91
AA677934


456332
Hs.399939
gb: nc39d05.r1 NCI_CGAP_Pr2 Homo sapiens cDNA
3.95
3.88
AA228357




clone, mRNA sequence


427999
Hs.181369
ubiquitin fusion degradation 1-like
3.94
3.86
AI435128


427681
Hs.284232
tumor necrosis factor receptor superfamily, member
3.93
3.81
AB018263




12 (translocating chain-association membrane protein)


413929
Hs.75617
collagen, type IV, alpha 2
3.93
3.79
BE501689


420116
Hs.95231
FH1/FH2 domain-containing protein
3.9
3.77
NM_013241


433914
Hs.112160

Homo sapiens DNA helicase homolog (PIF1) mRNA,

3.88
3.75
AF108138




partial cds


420732
Hs.367762
ESTs
3.87
3.74
AA789133


452517

gb: RC-BT068-130399-068 BT068 Homo sapiens
3.84
3.70
AI904891




cDNA, mRNA sequence


437524
Hs.385719
ESTs
3.82
3.68
AI627565


435158
Hs.65588
DAZ associated protein 1
3.8
3.66
AW663317


448780
Hs.267749
Human DNA sequence from clone 366N23 on chromosome
3.8
3.65
W92071




6q27. Contains two genes similar to consecutive




parts of the C. elegans UNC-93 (protein 1,




C46F11.1) gene, a KIAA0173 and Tubulin-Tyrosine




Ligase LIKE gene, a Mitotic Feedback Control Protein




MADP2 H


445084
Hs.250848
hypothetical protein FLJ14761
3.79
3.64
H38914


423138

gb: EST385571 MAGE resequences, MAGM Homo
3.75
3.60
AW973426





sapiens cDNA, mRNA sequence



419602
Hs.91521
hypothetical protein
3.74
3.59
AW248434


442549
Hs.8375
TNF receptor-associated factor 4
3.74
3.58
AI751601


450893
Hs.25625
hypothetical protein FLJ11323
3.73
3.55
AK002185


414223
Hs.238246
hypothetical protein FLJ22479
3.73
3.55
AA954566


444312
Hs.351142
ESTs
3.72
3.53
R44007


425205
Hs.155106
receptor (calcitonin) activity modifying protein 2
3.71
3.51
NM_005854


432327
Hs.274363
neuroglobin
3.71
3.49
R36571


451970
Hs.211046
ESTs
3.67
3.48
AI825732


408049
Hs.345588
desmoplakin (DPI, DPII)
3.67
3.45
AW076098


440100
Hs.158549
ESTs, Weakly similar to T2D3_HUMAN TRANSCRIPTION
3.66
3.45
BE382685




INITIATION FACTOR TFIID 135 KDA




SUBUNIT [H. sapiens]


426468
Hs.117558
ESTs
3.65
3.43
AA379306


402384

NM_007181*: Homo sapiens mitogen-activated protein
3.64
3.43





kinase kinase kinase kinase 1 (MAP4K1),




mRNA.


458132
Hs.103267
hypothetical protein FLJ22548 similar to gene trap
3.64
3.42
AW247012




PAT 12


447400
Hs.18457
hypothetical protein FLJ20315
3.64
3.42
AK000322


443893
Hs.115472
ESTs, Weakly similar to 2004399A chromosomal
3.63
3.41
BE079602




protein [H. sapiens]


424959
Hs.153937
activated p21cdc42Hs kinase
3.62
3.40
NM_005781


409586
Hs.55044
DKFZP586H2123 protein
3.6
3.39
AL050214


445692
Hs.182099
ESTs
3.6
3.37
AI248322


433052
Hs.293003
ESTs, Weakly similar to PC4259 ferritin associated
3.6
3.36
AW971983




protein [H. sapiens]


421782
Hs.108258
actin binding protein; macrophin (microfilament and
3.59
3.35
AB029290




actin filament cross-linker protein)


414907
Hs.77597
polo (Drosophia)-like kinase
3.58
3.34
X90725


454639

gb: RC2-ST0158-091099-011-d05 ST0158 Homo
3.57
3.33
AW811633





sapiens cDNA, mRNA sequence



434547
Hs.106124
ESTs
3.56
3.32
R26240


439130
Hs.375195
ESTs
3.55
3.32
AA306090


413564

gb: 601146990F1 NIH_MGC_19 Homo sapiens cDNA
3.54
3.31
BE260120




clone 5′, mRNA sequence


443471
Hs.398102

Homo sapiens clone FLB3442 PRO0872 mRNA,

3.53
3.31
AW236939




complete cds


424415
Hs.146580
enolase 2, (gamma, neuronal)
3.52
3.30
NM_001975


405036

NM_021628*: Homo sapiens arachidonate lipoxy-
3.52
3.29





genase 3 (ALOXE3), mRNA. VERSION




NM_020229.1 GI


422068
Hs.104520

Homo sapiens cDNA FLJ13694 fis, clone

3.52
3.29
AI807519




PLACE2000115


424244
Hs.143601
hypothetical protein hCLA-iso
3.52
3.28
AV647184


451867
Hs.27192
hypothetical protein dJ1057B20.2
3.51
3.26
W74157


429187
Hs.163872
ESTs, Weakly similar to S65657 alpha-1C-adrenergic
3.49
3.26
AA447648




receptor splice form 2 [H. sapiens]


415200
Hs.78202
SWI/SNF related, matrix associated, actin dependent
3.48
3.25
AL040328




regulator of chromatin, subfamily a, member 4


405667

Target Exon
3.48
3.25



421075
Hs.101474
KIAA0807 protein
3.47
3.23
AB018350


424909
Hs.153752
cell division cycle 25B
3.46
3.22
S78187


451164
Hs.60659
ESTs, Weakly similar to T46471 hypothetical protein
3.46
3.21
AA015912




DKFZp434L0130.1 [H. sapiens]


438644
Hs.129037
ESTs
3.46
3.20
AI126162


432258
Hs.293039
ESTs
3.45
3.19
AW973078


411817
Hs.72241
mitogen-activated protein kinase kinase 2
3.45
3.19
BE302900


414918
Hs.72222
hypothetical protein FLJ13459
3.45
3.18
AI219207


437256
Hs.97871
Homo sapiens, clone IMAGE: 3845253, mRNA, partial
3.43
3.17
AL137404




cds


404208

C6001282: gi|4504223|ref|NP_000172.1|glucuronidase,
3.42
3.16





beta [Homo sapiens] gi|114963|sp|P082


421989
Hs.110457
Wolf-Hirschhorn syndrome candidate 1
3.4
3.15
AJ007042


438942
Hs.6451
PRO0659 protein
3.39
3.14
AW875398


412649
Hs.74369
integrin, alpha 7
3.38
3.14
NM_002206


414840
Hs.23823
hairy/enhancer-of-split related with YRPW motif-like
3.37
3.13
R27319


434831
Hs.273397
KIAA0710 gene product
3.35
3.12
AA248060


431842
Hs.271473
epithelial protein up-regulated in carcinoma, membrane
3.34
3.11
NM_005764




associated protein 17


402328

Target Exon
3.34
3.10



405371

NM_005569*: Homo sapiens LIM domain kinase 2
3.33
3.10





(LIMK2), transcript variant 2a, mRNA.


441650
Hs.132545
ESTs
3.32
3.09
AI261960


418629
Hs.86859
growth factor receptor-bound protein 7
3.3
3.09
BE247550


406002

Target Exon
3.3
3.08



420307
Hs.66219
ESTs
3.29
3.08
AW502869


425093
Hs.154525
KIAA1076 protein
3.28
3.07
AB028999


427351
Hs.123253
hypothetical protein FLJ22009
3.28
3.07
AW402593


417900
Hs.82906
CDC20 (cell division cycle 20, S. cerevisiae, homolog)
3.28
3.06
BE250127


457228
Hs.195471
Human cosmid CRI-JC2015 at D10S289 in 10sp13
3.27
3.05
U15177


421026
Hs.101067
GCN5 (general control of amino-acid synthesis, yeast,
3.27
3.04
AL047332




homolog)-like 2


430746
Hs.406256
ESTs
3.27
3.03
AW977370


409556
Hs.54941
phosphorylase kinase, alpha 2 (liver)
3.27
3.03
D38616


451225
Hs.57655
ESTs
3.26
3.03
AI433694


404913

NM_024408*: Homo sapiens Notch (Drosophila)
3.25
3.02





homolog 2 (NOTCH2), mRNA. VERSION




NM_024410.1 GI


404875

NM_022819*: Homo sapiens phospholipase A2, group
3.23
3.02





IIF (PLA2G2F), mRNA. VERSION NM_020245.2 GI


404606

Target Exon
3.23
3.01



414732
Hs.77152
minichromosome maintenance deficient (S. cerevisiae) 7
3.22
3.01
AW410976


425380
Hs.32148
AD-015 protein
3.22
3.00
AA356389


421186
Hs.270563
ESTs, Moderately similar to T12512 hypothetical
3.21
2.98
AI798039




protein DKFZp434G232.1 [H. sapiens]


445462
Hs.288649
hypothetical protein MGC3077
3.2
2.97
AA378776









Permutation analysis of 100 most significantly up-regulated genes in each group By permuting the sample labels 500 times we estimated the significance of the differentially expressed genes. The permutation analysis revealed that it was highly unlikely to find as good markers by chance, as similar godd markers were only found in 5% of the permutated data sets, see Table 2.


Molecular Predictor of Progression

A molecular predictor of progression using a combination of genes may have higher prediction accuracy than when using single marker genes. Therefore, to identify the gene-set that gives the best prediction results using the lowest number of genes we built a predictor using the “leave one out” cross-validation approach, as previously described (Golub et al. 1999). Selecting the 100 best genes in each cross-validation loop gave the lowest number of pre-diction errors (5 errors, 83% correct classification) in our training set consisting of the 29 tumors (see FIG. 3). As in our previous study we used a maximum likelihood classification approach. We selected a gene-expression signature consisting of those 45 genes that were present in 75% of the cross-validation loops, and these represent our optimal gene-set for progression prediction (FIG. 4a and Table 3).


Many of these 45 genes were also found among the 200 best markers of progression, however, the cross-validation approach also identified other interesting markers of progression like BIRC5 (Survivin), an apoptosis inhibitor that is up regulated in the tumors that show later progression. BIRC5 has been reported to be expressed in most common cancers (Ambrosini et al. 1997). To validate the significance of the 45-gene expression signature we used a test set consisting of 19 early stage bladder tumors (9 tumors with no progression and 10 tumors with later progression). Total RNA from these samples were amplified, labeled and hybridized to customized 60mer-oligonucleotide microarray glass slides and the relative expressions of the 45 classifier genes were measured following appropriate normalization and background adjustments of the microarray data. The independent tumor samples were classified as non-progressing or progressing according to the degree of correlation to the average no progression profile from the training samples (FIG. 3b). When applying no cutoff limits to the predictions the predictor identified 74% of the samples correctly. However, as done recently in a breast cancer study (van't Veer et al. 2002), we applied correlation cutoff limits of 0.1 and −0.1 in order to disregard samples with really low correlation values and in this way we obtained 92% correct predictions of samples with correlation values above 0.1 or below −0.1. Although the test-set is limited in size the performance is notable and could be of clinical use.









TABLE 3







The 45 optimal genes for disease progression prediction.














Eos
Unigene




Exemplar



Hu03 ID
Build 133
Description
T-Test
5% perm
Gene Name
Accession
CV

















439010
Hs.75216
protein tyrosine phosphatase, receptor
4.57
4.39
PTPRF
AW170332
29




type, F


429124
Hs.196914
minor histocompatibility antigen HA-1
4.20
4.09
HA-1
AW505086
29


421649
Hs.106415
peroxisome proliferative activated
5.76
5.64
PPARD
AA721217
29




receptor, delta


433914
Hs.112160
DNA helicase homolog (PIF1)
3.88
3.61
PIF1
AF108138
29


429187
Hs.163872
ESTs, Weakly similar to hypothetical
3.49
3.17

AA447648
28




protein FLJ20489


422765
Hs.1578
baculoviral IAP repeat-containing 5
2.68
2.56
BIRC5
AW409701
28




(survivin)


433844
Hs.179647
ESTs
4.04
3.80

AA610175
26


450893
Hs.25625
Hypothetical protein FLJ11323
3.73
3.46
FLJ11323
AK002185
25


452866
Hs.268016
ESTs
3.10
3.02

R26969
24


424909
Hs.153752
cell division cycle 25B
3.46
3.16
CDC25B
S78187
24


452929
Hs.172816
neuregulin 1
4.37
4.23
NRG1
AW954938
23


420116
Hs.95231
formin homology 2 domain containing 1
3.90
3.63
FHOD1
NM_013241
22


453963
Hs.28959
cDNA FLJ36513 fis, clone
3.44
2.88

AA040311
29




TRACH2001523


429561
Hs.250646
baculoviral IAP repeat-containing 6
3.83
3.03
BIRC6
AF265555
29




(apollon)


418127
Hs.83532
membrane cofactor protein (CD46,
4.26
3.37
MCP
BE243982
29




trophoblast-lymphocyte cross-reactive




antigen)


422119
Hs.111862
KIAA0590 gene product
2.33
1.95
KIAA0590
AI277829
29


435521
Hs.6361
mitogen-activated protein kinase
5.24
4.53
MAP2K1IP1
W23814
29




kinase 1 interacting protein 1


409632
Hs.55279
serine (or cysteine) proteinase inhibitor,
4.89
4.11
SERPINB5
W74001
29




clade B (ovalbumin), member 5


452829
Hs.63368
ESTs
4.95
4.31

AI955579
29


416640
Hs.79404
DNA segment on chromosome 4
6.03
5.51
D4S234E
BE262478
29




(unique) 234 expressed sequence


425097
Hs.154545
PDZ domain containing guanine nucleotide
3.77
3.18
PDZ-GEF1
NM_014247
28




exchange factor(GEF)1


445926
Hs.334826
splicing factor 3b, subunit 1, 155 kDa
2.40
2.03
SF3B1
AF054284
28


437325
Hs.5548
F-box and leucine-rich repeat protein 5
2.48
2.09
FBXL5
AF142481
28


448813
Hs.22142
cytochrome b5 reductase b5R.2
4.28
3.41
LOC51700
AF169802
28


426799
Hs.303154
ESTs
4.86
4.04

H14843
28


446847
Hs.82845
ESTs
4.65
3.79

T51454
28


428016
Hs.181461
ariadne homolog, ubiquitin-conjugating
3.77
3.15
ARIH1
AJ243190
27




enzyme E2 binding protein, 1 (Drosophila)


418321
Hs.84087
KIAA0143 protein
4.62
3.76
KIAA0143
D63477
27


422984
Hs.351597
ESTs
3.50
2.93

W28614
26


408688
Hs.152925
KIAA1268 protein
3.52
2.95
KIAA1268
AI634522
26


440357
Hs.20950
phospholysine phosphohistidine inorganic
3.89
3.07
LHPP
AA379353
26




pyrophosphate phosphatase


420269
Hs.96264
alpha thalassemia/mental retardation
3.39
2.85
ATRX
U72937
26




syndrome X-linked (RAD54 (S. cerevisiae)




homolog)


423185
?
ornithine decarboxylase antizyme 1
4.61
3.71
OAZ1
BE299590
26


443407
Hs.348514
clone IMAGE: 4052238, mRNA, partial cds
4.21
3.32

AA037683
25


457329
Hs.359682
calpastatin
3.59
2.99
CAST
AI634860
25


452714
Hs.30340
KIAA1165: likely ortholog of mouse
3.62
3.01
KIAA1165
AW770994
25




Nedd4 WW domain-binding protein 5A


444773
Hs.11923
hypothetical protein DJ167A19.1
3.71
3.11
DJ167A19.1
BE156256
24


418504
Hs.85335
ESTs
4.59
3.67

BE159718
24


444604
Hs.11441
Chromosome 1 open reading frame 8
4.89
4.17
C1orf8
AW327695
23


410691
Hs.65450
reticulon 4


RTN4
AW239226
23


430604
Hs.247309
succinate-CoA ligase, GDP-forming,
4.61
3.72
SUCLG2
AV650537
23




beta subunit


421311
Hs.283609
muscleblind-like protein MBLL39
4.65
3.82
MBLL39
N71848
23


439632
Hs.334437
hypothetical protein MGC4248
4.29
3.42
MGC4248
AW410714
22


417924
Hs.82932
cyclin D1 (PRAD1: parathyroid adenomatosis 1)
4.35
3.49
CCND1
AU077231
22


453395
Hs.377915
mannosidase, alpha, class 2A, member 1
4.71
3.84
MAN2A1
D63998
22









Permutation Analysis of 45 Genes

Again permutation analysis revealed that for all of the 45 genes similar good markers were only found in 5% of the 500 permuted datasets (see Table 3).


Expression Profiling of Metachrone Higher Stage Tumors

Expression profiling of the metachrone higher stage tumors could provide important information on the degree of expression similarities between the primary and the secondary tumors. Tissues from secondary tumors were available from 14 of the patients with disease progression and these were also hybridized to the customized Affymetrix GeneChips. Hierarchical cluster analysis of all tumor samples based on the 3,213 most varying probesets showed that tumors originating from the same patient in 9 of the cases clustered tightly together indicating a high degree of intra individual similarity in expression profiles (FIG. 5). Notable, one tight clustering pair of tumors was a Ta and a T2+ tumor (patient 941). It was remarkable that Ta and T1 tumors and T1 or T2+ tumors from a single individual were more similar than e.g. Ta tumors from two individuals. There was no correlation between presence and absence of the tight clustering of samples from the same patient and time interval to tumor progression. The tight clustering of the 9 tumor pairs probably reflects the monoclonal nature of many bladder tumors (Sidransky et al. 1997). A set of genomic abnormalities like chromosomal gains and losses characterize bladder tumors of different stages from single individuals (Primdahl et al. 2002), and such physical abnormalities could be one of the causes of the strong similarity of metachronous tumors. The fact that 5 of the tumor pairs clustered apart may be explained by an oligoclonal origin of these tumors.


Customized GeneChip Design, Normalization and Expression Measures

We used a customized Affymetrix GeneChip (Eos Hu03) designed by Eos Biotech Inc., as described (Eaves et al. 2002). Approximately 45,000 mRNA/EST clusters and 6,200 predicted exons are represented by the 59,000 probesets on Eos Hu03 array. Data were normalized using protocols and software developed at Eos Biotechnology, Inc. (WO0079465). An “average intensity” (AI) for each probeset was calculated by taking the trimean of probe intensities following background subtraction and normalization to a gamma distribution (Turkey 1977).


cRNA Preparation, Array Hybridization and Scanning


Preparation of cRNA from total RNA and subsequent hybridization and scanning of the customized GeneChip microarrays (Eos Hu03) were performed as described previously (Dyrskjot et al. 2003).


Custom Oligonucleotide Microarray Procedures

Three 60mer oligonucleotides were designed for each of the 45 genes using Array Designer 2.0. All steps in the customized oligonucleotide microarray analysis were performed essentially as described (Kruhoffer et al.) Each of the probes was spotted in duplicates and all hybridisations were carried out twice. The samples were labelled with Cy3 and a common reference pool was labelled with Cy5. The reference pool was made by pooling of cRNA generated from investigated samples and from universal human RNA. Following scanning of the glass slides the fluorescent intensities were quantified and background adjusted using SPOT 2.0 (Jain et al. 2002). Data were subsequently normalized using a LOWESS normalisation procedure implemented in the SMA package to R. To select the best oligonucleotide probe for each of the 45 genes, 13 of the samples from the training set were re-analysed on the custom oligonucleotide microarray platform and the obtained expression ratios were compared to the expression levels from the Affymetrix GeneChips. The oligonucleotide probes with the highest correlation to the Affymetrix GeneChip probes were selected.


Expression Data Analysis

Before analysing the expression data from the Eos Hu03 GeneChips control probes were removed and only probes with AI levels above 100 in at least 8 experiments and with max/min equal to or above 1.6 were selected. This filtering generated a gene-set consisting of 6,647 probes for further analysis. Average linkage hierarchical cluster analysis of the tumour samples was carried out using a modified Pearson correlation as similarity metric (Eisen et al. 1998). Genes and arrays were median centered and normalised to the magnitude of 1 before clustering. We used the GeneCluster 2.0 software for the supervised selection of markers and for performing permutation tests. The 45 genes for predicting progression were selected by t-test statistics and cross-validation performance as previously described (Dyrskjot et al. 2003) and independent samples were classified according to the correlation to the average no progression signature profile of the 45 genes.


Example 2
Identifying Distinct Classes of Bladder Carcinoma Using Microarrays
Patient Disease Course Information—Class Discovery

We selected tumours from the entire spectrum of bladder carcinoma for expression profiling in order to discover the molecular classes of the disease. The tumours analysed are listed in Table 4 below together with the available patient disease course information.









TABLE 4







Disease course information of all patients involved-class discovery.



















Reviewed

Carcinoma in


Group
Patient
Previous tumours
Tumour examined on array
Pattern
histology
Subsequent tumours
situ*





A
 709-1

Ta gr 2 (200297)
Papillary
Ta gr3

no



 968-1

Ta gr 2 (011098)
Papillary
+
Ta gr 2 (150101)
no



 934-1

Ta gr 2 (220798)
Papillary
+

no



 928-1

Ta gr 2 (240698)
Papillary
+

no



 930-1

Ta gr 2 (300698)
Papillary
+

no


B
 989-1

Ta gr 3 (281098)
Papillary
+

no



1264-1

Ta gr 3 (130600)
Papillary
+
Ta gr 2 (231000)
no








Ta gr 2 (220101)








Ta gr 2 (300401)



 876-5
Ta gr 2 (230398)
Ta gr 3 (170400)
Papillary
+

no




Ta gr 2 (271098)




Ta gr 2 (090699)




Ta gr 2 (011199)



 669-7
Ta gr 2 (101296)
Ta gr 3 (230899)
Papillary
Ta gr2
Ta gr 2 (120100)
no




Ta gr 2 (150897)



Ta gr 2 (250500)




Ta gr 1 (161297)



Ta gr 2 (250900)




Ta gr 3 (270498)



Ta gr 2 (050201)




Ta gr 2 (220299)



 716-2
Ta gr 2 (070397)
Ta gr 3 (230497)
Papillary
+
Ta gr 2 (040697)
no








Ta gr 1 (170698)


C
1070-1

Ta gr 3 (150399)
Papillary
+
Ta gr 3 (291099)
Subsequent









visit



 956-2

Ta gr 3 (061299)
Papillary
+
Ta gr 3 (061200)
Sampling visit



1062-2

Ta gr 3 (120799)
Papillary
+
T1 gr 3 (161199)
Sampling visit



1166-1

Ta gr 3 (271099)
Papillary
+

Sampling visit



1330-1

Ta gr 3 (311000)
Papillary
+

Sampling visit


D
112-10
Ta gr 2 (070794)
Ta gr 3 (060198)
Papillary
+
Ta gr 3 (110698)
Previous visit




Ta gr 3 (011294)



T1 gr 3 (191098)




T1 gr 3 (150695)



Ta gr 3 (240299)




Ta gr 3 (121095)



T1 gr 3 (050799)




T1 gr 3 (040396)



T1 gr 3 (081199)




Ta gr 2 (200896)



T1 gr 3 (180400)




Ta gr 2 (111296)




Ta gr 2 (230497)




Ta gr 2 (030997)



 320-7
T1 gr 3 (011194)
Ta gr 3 (290997)
Papillary
+
Ta gr 3 (290198)
Sampling visit




T1 gr 3 (150896)



Ta gr 3 (290698)




Ta gr 3 (100897)



 747-7
Ta gr 2 (010597)
Ta gr 3 (161298)
Papillary
+
Ta gr 2 (050599)
Sampling visit




Ta gr 2 (220597)



Ta gr 2 (280999)




Ta gr 2 (230997)



Ta gr 2 (141299)




Ta gr 2 (260198)




T1 gr 3 (270498)




Ta gr 2 (170898)



 967-3
T1 gr 3 (280998)
Ta gr 3 (140699)
Papillary
+
T1 gr 3 (080999)
Sampling visit




T1 gr 3 (250199)


E
 625-1

T1 gr 3 (200996)
Papillary
+

No



 847-1

T1 gr 3 (210198)
Papillary
+

No



1257-1

T1 gr 3 (240500)
Solid
+

Sampling visit



 919-1

T1 gr 3 (220698)
Papillary
+

No



 880-1

T1 gr 3 (300398)
Papillary
+
Ta gr 2 (091198)
No








Ta gr 1 (090399)








Ta gr 2 (050900)








Ta gr 2 (190301)



 812-1

T1 gr 3 (061098)
Papillary
+

No



1269-1

T1 gr 3 (230600)
Papillary


No



1083-2
Ta gr 2 (280499)
T1 gr 3 (120599)
Papillary


No



1238-1

T1 gr 3 (020500)
Papillary
+
T2 gr 3 (211100)
No








Ta gr 2 (211100)



1065-1

T1 gr 3 (160399)
Papillary


Subsequent









visit



1134-1

T1 gr 3 (181099)
Papillary
T2 gr3
T1 gr 3 (280200)
Sampling visit








T1 gr 3 (020500)








T1 gr 3 (131100)


F
1164-1

T2+ gr 4 (101299)
Solid
gr 3

No



1032-1

T2+ gr ? (050199)
Mixed


Not measured



1117-1

T2+ gr 3 (010999)
Solid
+

Sampling visit



1178-1

T2+ gr 3 (200100)
Solid
+

Not measured



1078-1

T2+ gr 3 (120499)
Solid
+

Not measured



 875-1

T2+ gr 3 (180398)
Solid
+

No



1044-1

T2+ gr 3 (010299)
Solid
+
T2+ gr 3 (060999)
Not measured



1133-1

T2+ gr 3 (081099)
Solid
+

Not measured



1068-1

T2+ gr 3 (220399)
Solid
+

No



 937-1

T2+ gr 3 (280798)
Solid


Not measured





Group A: Ta gr2 tumours - no recurrence within 2 years.


Group B: Ta gr3 tumours - no prior T1 tumour and no carcinoma in situ in random biopsies.


Group C: Ta gr3 tumours - no prior T1 tumour but carcinoma in situ in random biopsies.


Group D: Ta gr3 tumours - a prior T1 tumour and carcinoma in situ in random biopsies.


Group E: T1 gr3 tumours - no prior T2+ tumour. Group F: T2+ tumours gr3/4 - only primary tumours.


*Carcinoma in situ detected in selected site biopsies at previous, sampling or subsequent visits.






Two-Way Hierarchical Cluster Analysis of Tumor Samples

A two-way hierarchical cluster analysis of the tumour samples based on the 1767 gene-set (see class discovery using hierarchical clustering) remarkably separated all 40 tumours according to conventional pathological stages and grades with only few exceptions (FIG. 6a). We identified two main branches containing the superficial Ta tumours, and the invasive T1 and T2+ tumours. In the superficial branch two sub-clusters of tumours could be identified, one holding 8 tumours that had frequent recurrences and one holding 3 out of the five Ta grade 2 tumours with no recurrences. In the invasive branch, it was notable that four Ta grade 3 tumours clustered tightly with the muscle invasive T2+ tumours. These four Ta tumours, from patients with no previous tumour history, showed concomitant CIS in the surrounding mucosa, indicating that this sub-fraction of Ta tumours has some of the more aggressive features found in muscle invasive tumours. The stage T1 cluster could be separated into three sub-clusters with no clear clinical difference. The one stage T1 grade 3 tumour that clustered with the stage T2+ muscle invasive tumours was the only T1 tumour that showed a solid growth pattern, all others showing papillary growth. Nine out of ten T2+ tumours were found in one single cluster. The remarkable distinct separation of the tumour groups according to stage, with practically no overlap between groups, was also demonstrated by multidimensional scaling analysis (FIG. 6c).


In an attempt to reduce the number of genes needed for class prediction we identified those genes that were scored by the Cancer Genome Anatomy Project (at NCI) as belonging to cancer-related groups such as tumour suppressors, oncogenes, cell cycle, etc. These genes were then selected from the initial 1767 gene-set, and those 88 which showed largest variation (SD of the gene vector >=4), were used for hierarchical clustering of the tumour samples. The obtained clusters was almost identical to the 1767 gene-set cluster dendrogram (FIG. 6b), indicating that the tumour clustering does not simply reflect larger amounts of stromal components in the invasive tumour biopsies.


The clustering of the 1767 genes revealed several characteristic profiles in which there was a distinct difference between the tumour groups (FIG. 6d; black lines identifying clusters a to j).


Cluster a, shows a high expression level in all the Ta grade 3 tumours (FIG. 7a) and, as a novel finding, contains genes encoding 8 transcription factors as well as other nuclear genes related to transcriptional activity. Cluster c contains genes that are up-regulated in both Ta grade 3 with high recurrence rate and CIS, in T2+ and some T1 tumours. This cluster shows a remarkable tight co-regulation of genes related to cell cycle control and mitosis (FIG. 7c). Genes encoding cyclins, PCNA as well as a number of centromere related proteins are present in this cluster. They indicate increased cellular proliferation and may form new targets for small molecule therapy (Seymour 1999). Cluster f shows a tight cluster of genes related to keratinisation (FIG. 7f). Two tumours (875-1 and 1178-1) had a very high expression of these genes and a re-evaluation of the pathology slides revealed that these were the only two samples to show squamous metaplasia. Thus, activation of this cluster of genes promotes the squamous metaplasia not infrequently seen by light microscopy in invasive bladder tumours. The genes in this cluster is listed in Table 5.









TABLE 5







Genes for classifying samples with squamous metaplasia









Chip acc. #
UniGene Build 162
description





D83657_at
Hs.19413
NM_005621; S100 calcium-binding protein A12


HG3945-HT4215_at


J00124_at


L05187_at


L05188_f_at
Hs.505327


L10343_at
Hs.112341
NM_002638; skin-derived protease inhibitor 3 preproprotein


L42583_f_at
Hs.367762
NM_005554; keratin 6A


L42601_f_at
Hs.367762
NM_005554; keratin 6A


L42611_f_at
Hs.446417
NM_173086; keratin 6 isoform K6e


M19888_at
Hs.1076
NM_003125; small proline-rich protein 1B (cornifin)


M20030_f_at
Hs.505352


M21005_at


M21302_at
Hs.505327


M21539_at
Hs.2421
NM_006518; small proline-rich protein 2C


M86757_s_at
Hs.112408
NM_002963; S100 calcium-binding protein A7


S72493_s_at
Hs.432448
NM_005557; keratin 16


U70981_at
Hs.336046
NM_000640; interleukin 13 receptor, alpha 2 precursor


V01516_f_at
Hs.367762
NM_005554; keratin 6A


X53065_f_at


X57766_at
Hs.143751
NM_005940; matrix metalloproteinase 11 preproprotein


Z19574_rna1_at









Cluster g contains genes that are up-regulated in T2+ tumours and in the Ta grade 3 tumours with CIS that cluster in the invasive branch (FIG. 7g). This cluster contains genes related to angiogenesis and connective tissue such as laminin, myosin, caldesmon, collagen, dystrophin, fibronectin, and endoglin. The increased transcription of these genes may indicate a profound remodelling of the stroma that could reflect signalling from the tumour cells, from infiltrating lymphocytes, or both. Some of these may also form new drug targets (Fox et al. 2001). It is remarkable that these genes are those that most clearly separate the Ta grade 3 tumours surrounded by CIS from all other Ta grade 3 tumours. The presence of adjacent CIS is usually diagnosed by taking a set of eight biopsies from different places in the bladder mucosa. However, the present data clearly indicate that analysis of stroma remodelling genes in the Ta tumours could eliminate this invasive procedure.


The clusters b, d, e, h, i, and j contain genes related to nuclear proteins, cell adhesion, growth factors, stromal proteins, immune system, and proteases, respectively (see FIG. 8). A summary of the stage related gene expression is shown in Table 6.









TABLE 6







Table 6. Summary of stage related gene expression


Functional gene clustersa















Nuclear

Matrix re-
Extracellular
Immune


Tumour stage
Transcription
processes
Proliferation
modelling
matrix
system





Ta gr2




↓↓



Ta gr3
↑↑↑
↑↑
↑↑

↓↓



T1 gr3
b

↑↑b


b


T2 gr3


↑↑↑
↑↑↑




Ta gr3 + CIS
↑↑↑
↑↑
↑↑↑
↑↑↑








aFor a detailed description of gene clusters see FIG. 8.




bAn increase in gene expression was only found in about half of the samples analysed.







Class Prediction of Bladder Tumours

An objective class prediction of bladder tumours based on a limited gene-set is clinically useful. We therefore built a classifier using tumours correctly separated in the three main groups as identified in the cluster dendrogram (FIG. 6a). We used a maximum likelihood classification method with a “leave one out” cross-validation scheme (Shipp et al. 2002; van't Veer et al. 2002) in which one test tumour was removed from the set, and a set of predictive genes was selected from the remaining tumour samples for classifying the test tumour. This process was repeated for all tumours. Predictive genes that showed the largest possible separation of the three groups were selected for classification, and each tumour was classified according to how close it was to the mean of the three groups (FIG. 8a).


Classification of Samples

From the hierarchical cluster analysis of the samples (class discovery) we identified three major “molecular classes” of bladder carcinoma highly associated with the pathologic staging of the samples. Based on this finding we decided to build a molecular classifier that assigns tumours to these three “molecular classes”. To build the classifier, we only used the tumours in which there was a correlation between the “molecular class” and the associated pathologic stage. Consequently, a T1 tumour clustering in the “molecular class” of T2 tumours was not used to build the classifier.


The genes used in the classifier were those genes with the highest values of the ratio (B/W) of the variation between the groups to the variation within the groups. High values of the ratio (B/W) signify genes with good group separation performance. We calculated the sum over the genes of the squared distance from the sample value to the group mean and classified the sample as belonging to the group where the distance to the group mean was smallest. If the relative difference between the distance to the closest and the second closest group compared to the distance to the closest group were below 5%, the classification failed and the sample was classified as belonging to both groups. The relative difference is referred to as the classifier strength.


Classifier Performance

The classifier performance was tested using from 1-160 genes in cross-validation loops. FIG. 9 shows that the closest correlation to histopathology is obtained in the cross-validation model using from 69-97 genes. Based on this we chose the model using 80 genes for cross-validation as our final classifier model.


Classifier Model Using 71 Genes

We selected those genes for our final classifier model that were used in at least 75% (25 times) of the cross-validation loops. These 71 genes are listed in table 7.









TABLE 7







Feature: Accession number on HuGene fl array. Number: Number of times used in


the 80 genes cross validation loops. Test (B/W): see below.












Unigene


Test


Feature
Build 162
Description
Number
(B/W)














AF000231_at
Hs.75618
NM_004663; Ras-related protein Rab-11A
33
26.77


D13666_s_at
Hs.136348
NM_006475; osteoblast specific factor 2 (fasciclin I-like)
33
27.71


D49372_s_at
Hs.54460
NM_002986; small inducible cytokine A11 precursor
31
25.78


D83920_at
Hs.440898
NM_002003; ficolin 1 precursor
33
31.18


D86479_at
Hs.439463
NM_001129; adipocyte enhancer binding protein 1 precursor
33
28.29


D89077_at
Hs.75367
NM_006748; Src-like-adaptor
33
30.03


D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2
33
51.50


HG4069-HT4339_s_at


27
25.06


HG67-HT67_f_at


33
27.81


HG907-HT907_at


33
25.76


J02871_s_at
Hs.436317
NM_000779; cytochrome P450, family 4, subfamily B,
33
32.61




polypeptide 1


J03278_at
Hs.307783
NM_002609; platelet-derived growth factor receptor beta
33
28.02




precursor


J04058_at
Hs.169919
NM_000126; electron transfer flavoprotein, alpha poly-
33
29.46




peptide


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase
33
38.21


J05070_at
Hs.151738
NM_004994; matrix metalloproteinase 9 preproprotein
33
35.34


J05448_at
Hs.79402
NM_002694; DNA directed RNA polymerase II polypeptide
32
26.51




C NM_032940; DNA directed RNA polymerase II




polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or cysteine) proteinase inhibitor,
33
28.66




clade A (alpha-1 antiproteinase, antitrypsin), member 1


L13720_at
Hs.437710
NM_000820; growth arrest-specific 6
33
29.69


M12125_at
Hs.300772
NM_003289; tropomyosin 2 (beta)
28
24.89


M15395_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor
33
29.40


M16591_s_at
Hs.89555
NM_002110; hemopoietic cell kinase isoform p61HCK
33
32.34


M20530_at


33
30.28


M23178_s_at
Hs.73817
NM_002983; chemokine (C-C motif) ligand 3
33
35.36


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2
33
41.88


M33195_at
Hs.433300
NM_004106; Fc fragment of IgE, high affinity I, receptor
33
30.40




for, gamma polypeptide precursor


M55998_s_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
33
26.83


M57731_s_at
Hs.75765
NM_002089; chemokine (C—X—C motif) ligand 2
33
31.84


M68840_at
Hs.183109
NM_000240; monoamine oxidase A
33
32.39


M69203_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor
33
36.21


M72885_rna1_s_at


33
27.94


M83822_at
Hs.209846
NM_006726; LPS-responsive vesicle trafficking, beach
33
26.44




and anchor containing


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)
33
49.85


U01833_at
Hs.81469
NM_002484; nucleotide binding protein 1 (MinD homolog,
33
30.62




E. coli)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1
33
39.10


U09937_rna1_s_at


33
30.88


U10550_at
Hs.79022
NM_005261; GTP-binding mitogen-induced T-cell protein
28
25.26




NM_181702; GTP-binding mitogen-induced T-cell protein


U20158_at
Hs.2488
NM_005565; lymphocyte cytosolic protein 2
33
32.41


U41315_rna1_s_at


33
43.56


U47414_at
Hs.13291
NM_004354; cyclin G2
33
44.42


U49352_at
Hs.414754
NM_001359; 2,4-dienoyl CoA reductase 1 precursor
33
37.04


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase
33
42.89




E1, beta polypeptide precursor NM_183050; branched




chain keto acid dehydrogenase E1, beta polypeptide




precursor


U52101_at
Hs.9999
NM_001425; epithelial membrane protein 3
33
29.86


U64520_at
Hs.66708
NM_004781; vesicle-associated membrane protein 3
33
30.17




(cellubrevin)


U65093_at
Hs.82071
NM_006079; Cbp/p300-interacting transactivator, with
33
32.07




Glu/Asp-rich carboxy-terminal domain, 2


U68019_at
Hs.288261
NM_005902; MAD, mothers against decapentaplegic
31
26.70




homolog 3


U68385_at
Hs.380923

33
31.56


U74324_at
Hs.90875
NM_002871; RAB-interacting factor
33
30.26


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2
33
50.37




NM_032235;


U90549_at
Hs.236774
NM_006353; high mobility group nucleosomal binding
33
32.16




domain 4


X04085_rna1_at


28
25.13


X07743_at
Hs.77436
NM_002664; pleckstrin
33
28.13


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor
33
35.79


X14046_at
Hs.153053
NM_001774; CD37 antigen
30
24.70


X15880_at
Hs.415997
NM_001848; collagen, type VI, alpha 1 precursor
33
31.51


X15882_at
Hs.420269
NM_001849; alpha 2 type VI collagen isoform 2C2 precursor
33
32.32




NM_058174; alpha 2 type VI collagen isoform




2C2a precursor NM_058175; alpha 2 type VI collagen




isoform 2C2a precursor


X51408_at
Hs.380138
NM_001822; chimerin (chimaerin) 1
33
30.51


X53800_s_at
Hs.89690
NM_002090; chemokine (C—X—C motif) ligand 3
33
33.63


X54489_rna1_at


33
33.57


X57579_s_at


33
41.43


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor
33
43.21


X67491_f_at
Hs.355697
NM_005271; glutamate dehydrogenase 1
33
30.97


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a
33
46.53




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7
33
53.16


X78520_at
Hs.372528
NM_001829; chloride channel 3
33
47.38


Y00787_s_at
Hs.624
NM_000584; interleukin 8 precursor
32
27.54


Z12173_at
Hs.334534
NM_002076; glucosamine (N-acetyl)-6-sulfatase precursor
30
25.44


Z19554_s_at
Hs.435800
NM_003380; vimentin
27
24.59


Z26491_s_at
Hs.240013
NM_000754; catechol-O-methyltransferase isoform MB-
32
26.92




COMT NM_007310; catechol-O-methyltransferase isoform




S-COMT


Z29331_at
Hs.372758
NM_003344; ubiquitin-conjugating enzyme E2H isoform 1
33
33.49




NM_182697; ubiquitin-conjugating enzyme E2H isoform 2


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2
33
44.45




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2




isoform 4 NM_176869; inorganic pyrophosphatase 2




isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
33
55.18









Test for Significance of Classifier

To test the class separation performance of the 71 selected genes we compared the B/W ratios with the similar ratios of all the genes calculated from permutations of the arrays. For each permutation we construct three pseudogroups, pseudo-Ta, pseudo-T1, and pseudo-T2, so that the proportion of samples from the three original groups is approximately the same in the three pseudogroups. We then calculate the ratio of the variation between the pseudogroups to the variation within the pseudogroups for all the genes. For 500 permutations we only two times had one gene for which the B/W value was higher than the lowest value for the original B/W values of the 71 selected genes (the two values being 25.28 and 25.93).


The classifier performance was tested using from 1-160 genes in cross-validation loops, and a model using an 80 gene cross-validation scheme showed the best correlation to pathologic staging (p<10−9). The 71 genes that were used in at least 75% of the cross validation loops were selected to constitute our final classifier model. See the expression profiles of the 71 genes in FIG. 10. The genes are clustered to obtain a better overview of similar expression patterns. From this it is obvious that the T1 stage is characterised by having expression patterns in common with either Ta or T2 tumours. There are no single genes that can be used as a T1 marker.


Permutation Analysis

To test the class separation performance of the 71 selected genes we compared their performance to those of a permutated set of pseudo-Ta, T1 and T2 tumours. In 500 permutations we only detected two genes with a performance equal to the poorest performing classifying genes.


Classification Using 80 Predictive Genes and Other Gene-Sets

The classification using 80 predictive genes in cross-validation loops identified the Ta group with no surrounding CIS and no previous tumor or no previous tumor of a higher stage (Table 8). Interestingly, the Ta tumours surrounded by CIS that were classified as T2 or T1 clearly demonstrate the potential of the classification method for identifying surrounding CIS in a non-invasive way, thereby supplementing clinical and pathologic information.









TABLE 8







Table 8 Clinical data on disease courses and results of molecular classification

















Carci-





Previous
Tumour
Subsequent
noma
Reviewed
Molecular classifier
















Tumoursa
Patient
tumours
analysed
tumours
in situb
histologyc
320
80
20










Ta grade II tumours - no progression

















709-1

Ta gr2

No
Ta gr3
Ta
Ta
Ta



968-1

Ta gr2
1 Ta
No

Ta/T1
Ta
Ta



934-1

Ta gr2

No

T1
Ta
Ta



928-1

Ta gr2

No

Ta
Ta
T1



930-1

Ta gr2

No

Ta
Ta
Ta







Ta grade III tumours - no prior T1 tumour or CIS

















989-1

Ta gr3

No

Ta
Ta
Ta



1264-1 

Ta gr3
3 Ta
No

Ta
Ta
Ta



876-5
4 Ta
Ta gr3

No

Ta
Ta
Ta



669-7
5 Ta
Ta gr3
4 Ta
No
Ta gr2
Ta
Ta
Ta



716-2
1 Ta
Ta gr3
2 Ta
No

Ta
Ta
Ta







Ta grade III tumours - no prior T1 tumour but CIS in selected site biopsies

















1070-1 

Ta gr3
1 Ta
Subsequent

Ta
Ta
Ta







visit



956-2

Ta gr3
1 Ta
Sampling

T2
T2
T2/T1







visit



1062-2 

Ta gr3
1 T1
Sampling

T2/Ta
T1/Ta
Ta







visit



1166-1 

Ta gr3

Sampling

Ta/T1
Ta
Ta







visit



1330-1 

Ta gr3

Sampling

T2
T2
Ta







visit







Ta grade III tumours - a prior T1 tumour and CIS in selected site biopsies

















747-7
5 Ta, 1 T1
Ta gr3
3 Ta
Sampling

Ta
Ta
Ta







visit



112-10
7 Ta, 2 T1
Ta gr3
2 Ta, 4 T1
Previous

Ta
Ta
Ta







visit



320-7
1 Ta, 2 T1
Ta gr3
2 Ta
Sampling

T2
T2
Ta







visit



967-3
2 T1
Ta gr3
1 T1
Sampling

Ta
Ta
Ta







visit







T1 grade III tumours - no prior muscle invasive tumour

















625-1

T1 gr3

No

T1
T1
T1



847-1

T1 gr3

No

T1
T1
T1



1257-1 

T1 gr3

Sampling

T1
T1
T1







visit



919-1

T1 gr3

No

T1
T1
T1



880-1

T1 gr3
4 Ta
No

T1
T1
T1



812-1

T1 gr3

No

T1
T1
T1



1269-1 

T1 gr3

No
No review
T1
T1
T1



1083-2 
1 Ta
T1 gr3

No
No review
T1
T1
T1



1238-1 

T1 gr3
1 Ta, 1 T2+
No

T1
T1
T1



1065-1 

T1 gr3

Subsequent
No review
T1
T1
T1







visit



1134-1 

T1 gr3
3 T1
Sampling
T2 gr3
T1
T1
T1







visit







T2+ grade III/IV tumours - only primary tumours

















1164-1 

T2+ gr4

No
T2+ gr3
T2/T1
T1
T1



1032-1 

T2+ gr?

ND
No review
T2
T2
T2



1117-1 

T2+ gr3

ND

T2
T2
T1



1178-1 

T2+ gr3

ND

T2
T2
T2



1078-1 

T2+ gr3

ND

T2
T2
T2



875-1

T2+ gr3

No

T2
T2
T2



1044-1 

T2+ gr3
1 T2+
ND

T2
T2
T2



1133-1 

T2+ gr3

ND

T2
T2
T2



1068-1 

T2+ gr3

No

T2
T2
T2



937-1

T2+ gr3

ND
No review
T1
T1
T1






aExamples of tumour histology.




bCarcinoma in situ detected in selected site biopsies at the time of sampling tumour tissue for the arrays or at previous or subsequent visits.




cAll tumours were reviewed by a single uro-pathologist and any change compared to the routine classification is listed.




dMolecular classification based on 320, 80, and 20 genes cross-validation loops.







Classification Using Other Gene-Sets

Classification was also carried out using other gene-sets (10, 20, 32, 40, 80, 160, and 320 genes). These gene-sets demonstrated the same classification tendency as the 71 genes. See Tables 9-15 for gene-sets.









TABLE 9







320 genes for classifier









Chip acc. #
UniGene Build 162
description





AB000220_at
Hs.171921
NM_006379; semaphorin 3C


AB000220_at
Hs.171921
NM_006379; semaphorin 3C


AC002073_cds1_at


AF000231_at
Hs.75618
NM_004663; Ras-




related protein Rab-11A


D10922_s_at
Hs.99855
NM_001462; formyl




peptide receptor-like 1


D10925_at
Hs.301921
NM_001295;




chemokine (C-C motif)




receptor 1


D11086_at
Hs.84
NM_000206; interleukin




2 receptor, gamma




chain, precursor


D11151_at
Hs.211202
NM_001957; endothelin




receptor type A


D13435_at
Hs.426142
NM_002643; phosphatidylinositol




glycan, class F isoform 1




NM_173074; phosphatidylinositol




glycan, class F isoform 2


D13666_s_at
Hs.136348
NM_006475; osteoblast




specific factor 2 (fasciclin I-like)


D14520_at
Hs.84728
NM_001730; Kruppel-like factor 5


D21878_at
Hs.169998
NM_004334; bone marrow stromal cell




antigen 1 precursor


D26443_at
Hs.371369
NM_004172; solute




carrier family 1 (glial




high affinity glutamate transporter),




member 3


D28589_at
Hs.17719


D42046_at
Hs.194665


D45370_at
Hs.74120
NM_006829; adipose




specific 2


D49372_s_at
Hs.54460
NM_002986; small




inducible cytokine A11




precursor


D50495_at
Hs.224397
NM_003195; transcription




elongation factor A




(SII), 2


D63135_at
Hs.27935
NM_032646; tweety




homolog 2


D64053_at
Hs.198288
NM_002849; protein




tyrosine phosphatase,




receptor type, R isoform




1 precursor




NM_130846; protein




tyrosine phosphatase,




receptor type, R isoform 2


D83920_at
Hs.440898
NM_002003; ficolin 1




precursor


D85131_s_at
Hs.433881
NM_002383; MYC-




associated zinc finger




protein


D86062_s_at
Hs.413482
NM_004649; chromosome




21 open reading




frame 33


D86479_at
Hs.439463
NM_001129; adipocyte




enhancer binding protein




1 precursor


D86957_at
Hs.307944


D86959_at
Hs.105751
NM_014720; Ste20-




related serine/threonine




kinase


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D87443_at
Hs.409862
NM_014758; sorting




nexin 19


D87682_at
Hs.134792


D89077_at
Hs.75367
NM_006748; Src-like-




adaptor


D89377_at
Hs.89404
NM_002449; msh




homeo box homolog 2


D90279_s_at
Hs.433695
NM_000093; alpha 1




type V collagen prepro-




protein


HG1996-HT2044_at


HG2090-HT2152_s_at


HG2463-HT2559_at


HG2994-HT4850_s_at


HG3044-HT3742_s_at


HG3187-HT3366_s_at


HG3342-HT3519_s_at


HG371-HT26388_s_at


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome




P450, family 4,




subfamily B, polypeptide 1


J03040_at
Hs.111779
NM_003118; secreted




protein, acidic, cysteine-




rich (osteonectin)


J03060_at


J03068_at


J03241_s_at
Hs.2025
NM_003239; transforming




growth factor, beta 3


J03278_at
Hs.307783
NM_002609; platelet-




derived growth factor




receptor beta precursor


J03909_at


J03925_at
Hs.172631
NM_000632; integrin




alpha M precursor


J04056_at
Hs.88778
NM_001757; carbonyl




reductase 1


J04058_at
Hs.169919
NM_000126; electron




transfer flavoprotein,




alpha polypeptide


J04093_s_at
Hs.278896
NM_019075; UDP




glycosyltransferase 1




family, polypeptide A10


J04130_s_at
Hs.75703
NM_002984;




chemokine (C-C motif)




ligand 4 precursor


J04152_rna1_s_at


J04162_at
Hs.372679
NM_000569; Fc fragment




of IgG, low affinity




IIIa, receptor for (CD16)


J04456_at
Hs.407909
NM_002305; beta-




galactosidase binding




lectin precursor


J05032_at
Hs.32393
NM_001349; aspartyl-




tRNA synthetase


J05036_s_at
Hs.1355
NM_001910; cathepsin




E isoform a preproprotein




NM_148964;




cathepsin E isoform b




preproprotein


J05070_at
Hs.151738
NM_004994; matrix




metalloproteinase 9




preproprotein


J05448_at
Hs.79402
NM_002694; DNA




directed RNA polymerase




II polypeptide C




NM_032940; DNA




directed RNA polymerase




II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or




cysteine) proteinase




inhibitor, clade A (alpha-




1 antiproteinase,




antitrypsin), member 1


K03430_at


L06797_s_at
Hs.421986
NM_003467;




chemokine (C—X—C




motif) receptor 4


L10343_at
Hs.112341
NM_002638; skin-




derived protease inhibitor




3 preproprotein


L11708_at
Hs.155109
NM_002153; hydroxysteroid




(17-beta) dehydrogenase 2


L13391_at
Hs.78944
NM_002923; regulator




of G-protein signalling




2, 24 kDa


L13698_at
Hs.65029
NM_002048; growth




arrest-specific 1


L13720_at
Hs.437710
NM_000820; growth




arrest-specific 6


L13923_at
Hs.750
NM_000138; fibrillin 1


AB000220_at
Hs.171921
NM_006379; semaphorin




3C


AC002073_cds1_at


AF000231_at
Hs.75618
NM_004663; Ras-




related protein Rab-11A


D10922_s_at
Hs.99855
NM_001462; formyl




peptide receptor-like 1


D10925_at
Hs.301921
NM_001295;




chemokine (C-C motif)




receptor 1


D11086_at
Hs.84
NM_000206; interleukin




2 receptor, gamma




chain, precursor


D11151_at
Hs.211202
NM_001957; endothelin




receptor type A


D13435_at
Hs.426142
NM_002643; phosphatidylinositol




glycan,




class F isoform 1




NM_173074; phosphatidylinositol




glycan,




class F isoform 2


D13666_s_at
Hs.136348
NM_006475; osteoblast




specific factor 2 (fasciclin




I-like)


D14520_at
Hs.84728
NM_001730; Kruppel-




like factor 5


D21878_at
Hs.169998
NM_004334; bone




marrow stromal cell




antigen 1 precursor


D26443_at
Hs.371369
NM_004172; solute




carrier family 1 (glial




high affinity glutamate




transporter), member 3


D28589_at
Hs.17719


D42046_at
Hs.194665


D45370_at
Hs.74120
NM_006829; adipose




specific 2


D49372_s_at
Hs.54460
NM_002986; small




inducible cytokine A11




precursor


D50495_at
Hs.224397
NM_003195; transcription




elongation factor A




(SII), 2


D63135_at
Hs.27935
NM_032646; tweety




homolog 2


D64053_at
Hs.198288
NM_002849; protein




tyrosine phosphatase,




receptor type, R isoform




1 precursor




NM_130846; protein




tyrosine phosphatase,




receptor type, R isoform 2


D83920_at
Hs.440898
NM_002003; ficolin 1




precursor


D85131_s_at
Hs.433881
NM_002383; MYC-




associated zinc finger




protein


D86062_s_at
Hs.413482
NM_004649; chromosome




21 open reading




frame 33


D86479_at
Hs.439463
NM_001129; adipocyte




enhancer binding protein




1 precursor


D86957_at
Hs.307944


D86959_at
Hs.105751
NM_014720; Ste20-




related serine/threonine




kinase


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D87443_at
Hs.409862
NM_014758; sorting




nexin 19


D87682_at
Hs.134792


D89077_at
Hs.75367
NM_006748; Src-like-




adaptor


D89377_at
Hs.89404
NM_002449; msh




homeo box homolog 2


D90279_s_at
Hs.433695
NM_000093; alpha 1




type V collagen prepro-




protein


HG1996-HT2044_at


HG2090-HT2152_s_at


HG2463-HT2559_at


HG2994-HT4850_s_at


HG3044-HT3742_s_at


HG3187-HT3366_s_at


HG3342-HT3519_s_at


HG371-HT26388_s_at


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome




P450, family 4,




subfamily B, polypeptide 1


J03040_at
Hs.111779
NM_003118; secreted




protein, acidic, cysteine-




rich (osteonectin)


J03060_at


J03068_at


J03241_s_at
Hs.2025
NM_003239; transforming




growth factor, beta 3


J03278_at
Hs.307783
NM_002609; platelet-




derived growth factor




receptor beta precursor


J03909_at


J03925_at
Hs.172631
NM_000632; integrin




alpha M precursor


J04056_at
Hs.88778
NM_001757; carbonyl




reductase 1


J04058_at
Hs.169919
NM_000126; electron




transfer flavoprotein,




alpha polypeptide


J04093_s_at
Hs.278896
NM_019075; UDP




glycosyltransferase 1




family, polypeptide A10


J04130_s_at
Hs.75703
NM_002984;




chemokine (C-C motif)




ligand 4 precursor


J04152_rna1_s_at


J04162_at
Hs.372679
NM_000569; Fc fragment




of IgG, low affinity




IIIa, receptor for (CD16)


J04456_at
Hs.407909
NM_002305; beta-




galactosidase binding




lectin precursor


J05032_at
Hs.32393
NM_001349; aspartyl-




tRNA synthetase


J05036_s_at
Hs.1355
NM_001910; cathepsin




E isoform a preproprotein




NM_148964;




cathepsin E isoform b




preproprotein


J05070_at
Hs.151738
NM_004994; matrix




metalloproteinase 9




preproprotein


J05448_at
Hs.79402
NM_002694; DNA




directed RNA polymerase




II polypeptide C




NM_032940; DNA




directed RNA polymerase




II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or




cysteine) proteinase




inhibitor, clade A (alpha-




1 antiproteinase,




antitrypsin), member 1


K03430_at


L06797_s_at
Hs.421986
NM_003467;




chemokine (C—X—C




motif) receptor 4


L10343_at
Hs.112341
NM_002638; skin-




derived protease inhibitor




3 preproprotein


L11708_at
Hs.155109
NM_002153; hydroxysteroid




(17-beta) dehydrogenase 2


L13391_at
Hs.78944
NM_002923; regulator




of G-protein signalling




2, 24 kDa


L13698_at
Hs.65029
NM_002048; growth




arrest-specific 1


L13720_at
Hs.437710
NM_000820; growth




arrest-specific 6


L13923_at
Hs.750
NM_000138; fibrillin 1


AB000220_at
Hs.171921
NM_006379; semaphorin




3C


AC002073_cds1_at


AF000231_at
Hs.75618
NM_004663; Ras-




related protein Rab-11A


D10922_s_at
Hs.99855
NM_001462; formyl




peptide receptor-like 1


D10925_at
Hs.301921
NM_001295;




chemokine (C-C motif)




receptor 1


D11086_at
Hs.84
NM_000206; interleukin




2 receptor, gamma




chain, precursor


D11151_at
Hs.211202
NM_001957; endothelin




receptor type A


D13435_at
Hs.426142
NM_002643; phosphatidylinositol




glycan,




class F isoform 1




NM_173074; phosphatidylinositol




glycan,




class F isoform 2


D13666_s_at
Hs.136348
NM_006475; osteoblast




specific factor 2 (fasciclin




I-like)


D14520_at
Hs.84728
NM_001730; Kruppel-




like factor 5


D21878_at
Hs.169998
NM_004334; bone




marrow stromal cell




antigen 1 precursor


D26443_at
Hs.371369
NM_004172; solute




carrier family 1 (glial




high affinity glutamate




transporter), member 3


D28589_at
Hs.17719


D42046_at
Hs.194665


D45370_at
Hs.74120
NM_006829; adipose




specific 2


D49372_s_at
Hs.54460
NM_002986; small




inducible cytokine A11




precursor


D50495_at
Hs.224397
NM_003195; transcription




elongation factor A




(SII), 2


D63135_at
Hs.27935
NM_032646; tweety




homolog 2


D64053_at
Hs.198288
NM_002849; protein




tyrosine phosphatase,




receptor type, R isoform




1 precursor




NM_130846; protein




tyrosine phosphatase,




receptor type, R isoform 2


D83920_at
Hs.440898
NM_002003; ficolin 1




precursor


D85131_s_at
Hs.433881
NM_002383; MYC-




associated zinc finger




protein


D86062_s_at
Hs.413482
NM_004649; chromosome




21 open reading




frame 33


D86479_at
Hs.439463
NM_001129; adipocyte




enhancer binding protein




1 precursor


D86957_at
Hs.307944


D86959_at
Hs.105751
NM_014720; Ste20-




related serine/threonine




kinase


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D87443_at
Hs.409862
NM_014758; sorting




nexin 19


D87682_at
Hs.134792


D89077_at
Hs.75367
NM_006748; Src-like-




adaptor


D89377_at
Hs.89404
NM_002449; msh




homeo box homolog 2


D90279_s_at
Hs.433695
NM_000093; alpha 1




type V collagen prepro-




protein


HG1996-HT2044_at


HG2090-HT2152_s_at


HG2463-HT2559_at


HG2994-HT4850_s_at


HG3044-HT3742_s_at


HG3187-HT3366_s_at


HG3342-HT3519_s_at


HG371-HT26388_s_at


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome




P450, family 4,




subfamily B, polypeptide 1


J03040_at
Hs.111779
NM_003118; secreted




protein, acidic, cysteine-




rich (osteonectin)


J03060_at


J03068_at


J03241_s_at
Hs.2025
NM_003239; transforming




growth factor, beta 3


J03278_at
Hs.307783
NM_002609; platelet-




derived growth factor




receptor beta precursor


J03909_at


J03925_at
Hs.172631
NM_000632; integrin




alpha M precursor


J04056_at
Hs.88778
NM_001757; carbonyl




reductase 1


J04058_at
Hs.169919
NM_000126; electron




transfer flavoprotein,




alpha polypeptide


J04093_s_at
Hs.278896
NM_019075; UDP




glycosyltransferase 1




family, polypeptide A10


J04130_s_at
Hs.75703
NM_002984;




chemokine (C-C motif)




ligand 4 precursor


J04152_rna1_s_at


J04162_at
Hs.372679
NM_000569; Fc fragment




of IgG, low affinity




IIIa, receptor for (CD16)


J04456_at
Hs.407909
NM_002305; beta-




galactosidase binding




lectin precursor


J05032_at
Hs.32393
NM_001349; aspartyl-




tRNA synthetase


J05036_s_at
Hs.1355
NM_001910; cathepsin




E isoform a preproprotein




NM_148964;




cathepsin E isoform b




preproprotein


J05070_at
Hs.151738
NM_004994; matrix




metalloproteinase 9




preproprotein


J05448_at
Hs.79402
NM_002694; DNA




directed RNA polymerase




II polypeptide C




NM_032940; DNA




directed RNA polymerase




II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or




cysteine) proteinase




inhibitor, clade A (alpha-




1 antiproteinase,




antitrypsin), member 1


K03430_at


L06797_s_at
Hs.421986
NM_003467;




chemokine (C—X—C




motif) receptor 4


L10343_at
Hs.112341
NM_002638; skin-




derived protease inhibitor




3 preproprotein


L11708_at
Hs.155109
NM_002153; hydroxysteroid




(17-beta) dehydrogenase 2


L13391_at
Hs.78944
NM_002923; regulator




of G-protein signalling




2, 24 kDa


L13698_at
Hs.65029
NM_002048; growth




arrest-specific 1


L13720_at
Hs.437710
NM_000820; growth




arrest-specific 6


L13923_at
Hs.750
NM_000138; fibrillin 1


AB000220_at
Hs.171921
NM_006379; semaphorin




3C


AC002073_cds1_at


AF000231_at
Hs.75618
NM_004663; Ras-




related protein Rab-11A


D10922_s_at
Hs.99855
NM_001462; formyl




peptide receptor-like 1


D10925_at
Hs.301921
NM_001295;




chemokine (C-C motif)




receptor 1


D11086_at
Hs.84
NM_000206; interleukin




2 receptor, gamma




chain, precursor


D11151_at
Hs.211202
NM_001957; endothelin




receptor type A


D13435_at
Hs.426142
NM_002643; phosphatidylinositol




glycan,




class F isoform 1




NM_173074; phosphatidylinositol




glycan,




class F isoform 2


D13666_s_at
Hs.136348
NM_006475; osteoblast




specific factor 2 (fasciclin




I-like)


D14520_at
Hs.84728
NM_001730; Kruppel-




like factor 5


D21878_at
Hs.169998
NM_004334; bone




marrow stromal cell




antigen 1 precursor


D26443_at
Hs.371369
NM_004172; solute




carrier family 1 (glial




high affinity glutamate




transporter), member 3


D28589_at
Hs.17719


D42046_at
Hs.194665


D45370_at
Hs.74120
NM_006829; adipose




specific 2


D49372_s_at
Hs.54460
NM_002986; small




inducible cytokine A11




precursor


D50495_at
Hs.224397
NM_003195; transcription




elongation factor A




(SII), 2


D63135_at
Hs.27935
NM_032646; tweety




homolog 2


D64053_at
Hs.198288
NM_002849; protein




tyrosine phosphatase,




receptor type, R isoform




1 precursor




NM_130846; protein




tyrosine phosphatase,




receptor type, R isoform 2


D83920_at
Hs.440898
NM_002003; ficolin 1




precursor


D85131_s_at
Hs.433881
NM_002383; MYC-




associated zinc finger




protein


D86062_s_at
Hs.413482
NM_004649; chromosome




21 open reading




frame 33


D86479_at
Hs.439463
NM_001129; adipocyte




enhancer binding protein




1 precursor


D86957_at
Hs.307944


D86959_at
Hs.105751
NM_014720; Ste20-




related serine/threonine




kinase


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D87443_at
Hs.409862
NM_014758; sorting




nexin 19


D87682_at
Hs.134792


D89077_at
Hs.75367
NM_006748; Src-like-




adaptor


D89377_at
Hs.89404
NM_002449; msh




homeo box homolog 2


D90279_s_at
Hs.433695
NM_000093; alpha 1




type V collagen prepro-




protein


HG1996-HT2044_at


HG2090-HT2152_s_at


HG2463-HT2559_at


HG2994-HT4850_s_at


HG3044-HT3742_s_at


HG3187-HT3366_s_at


HG3342-HT3519_s_at


HG371-HT26388_s_at


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome




P450, family 4,




subfamily B, polypeptide 1


J03040_at
Hs.111779
NM_003118; secreted




protein, acidic, cysteine-




rich (osteonectin)


J03060_at


J03068_at


J03241_s_at
Hs.2025
NM_003239; transforming




growth factor, beta 3


J03278_at
Hs.307783
NM_002609; platelet-




derived growth factor




receptor beta precursor


J03909_at


J03925_at
Hs.172631
NM_000632; integrin




alpha M precursor


J04056_at
Hs.88778
NM_001757; carbonyl




reductase 1


J04058_at
Hs.169919
NM_000126; electron




transfer flavoprotein,




alpha polypeptide


J04093_s_at
Hs.278896
NM_019075; UDP




glycosyltransferase 1




family, polypeptide A10


J04130_s_at
Hs.75703
NM_002984;




chemokine (C-C motif)




ligand 4 precursor


J04152_rna1_s_at


J04162_at
Hs.372679
NM_000569; Fc fragment




of IgG, low affinity




IIIa, receptor for (CD16)


J04456_at
Hs.407909
NM_002305; beta-




galactosidase binding




lectin precursor


J05032_at
Hs.32393
NM_001349; aspartyl-




tRNA synthetase


J05036_s_at
Hs.1355
NM_001910; cathepsin




E isoform a preproprotein




NM_148964;




cathepsin E isoform b




preproprotein


J05070_at
Hs.151738
NM_004994; matrix




metalloproteinase 9




preproprotein


J05448_at
Hs.79402
NM_002694; DNA




directed RNA polymerase




II polypeptide C




NM_032940; DNA




directed RNA polymerase




II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or




cysteine) proteinase




inhibitor, clade A (alpha-




1 antiproteinase,




antitrypsin), member 1


K03430_at


L06797_s_at
Hs.421986
NM_003467;




chemokine (C—X—C




motif) receptor 4


L10343_at
Hs.112341
NM_002638; skin-




derived protease inhibitor




3 preproprotein


L11708_at
Hs.155109
NM_002153; hydroxysteroid




(17-beta) dehydrogenase 2


L13391_at
Hs.78944
NM_002923; regulator




of G-protein signalling




2, 24 kDa


L13698_at
Hs.65029
NM_002048; growth




arrest-specific 1


L13720_at
Hs.437710
NM_000820; growth




arrest-specific 6


L13923_at
Hs.750
NM_000138; fibrillin 1


AB000220_at
Hs.171921
NM_006379; semaphorin




3C


AC002073_cds1_at


AF000231_at
Hs.75618
NM_004663; Ras-




related protein Rab-11A


D10922_s_at
Hs.99855
NM_001462; formyl




peptide receptor-like 1


D10925_at
Hs.301921
NM_001295;




chemokine (C-C motif)




receptor 1


D11086_at
Hs.84
NM_000206; interleukin




2 receptor, gamma




chain, precursor


D11151_at
Hs.211202
NM_001957; endothelin




receptor type A


D13435_at
Hs.426142
NM_002643; phosphatidylinositol




glycan,




class F isoform 1




NM_173074; phosphatidylinositol




glycan,




class F isoform 2


D13666_s_at
Hs.136348
NM_006475; osteoblast




specific factor 2 (fasciclin




I-like)


D14520_at
Hs.84728
NM_001730; Kruppel-




like factor 5


D21878_at
Hs.169998
NM_004334; bone




marrow stromal cell




antigen 1 precursor


D26443_at
Hs.371369
NM_004172; solute




carrier family 1 (glial




high affinity glutamate




transporter), member 3


D28589_at
Hs.17719


D42046_at
Hs.194665


D45370_at
Hs.74120
NM_006829; adipose




specific 2


D49372_s_at
Hs.54460
NM_002986; small




inducible cytokine A11




precursor


D50495_at
Hs.224397
NM_003195; transcription




elongation factor A




(SII), 2


D63135_at
Hs.27935
NM_032646; tweety




homolog 2


D64053_at
Hs.198288
NM_002849; protein




tyrosine phosphatase,




receptor type, R isoform




1 precursor




NM_130846; protein




tyrosine phosphatase,




receptor type, R isoform 2


D83920_at
Hs.440898
NM_002003; ficolin 1




precursor


D85131_s_at
Hs.433881
NM_002383; MYC-




associated zinc finger




protein


D86062_s_at
Hs.413482
NM_004649; chromosome




21 open reading




frame 33


D86479_at
Hs.439463
NM_001129; adipocyte




enhancer binding protein




1 precursor


D86957_at
Hs.307944


D86959_at
Hs.105751
NM_014720; Ste20-




related serine/threonine




kinase


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D87443_at
Hs.409862
NM_014758; sorting




nexin 19


D87682_at
Hs.134792


D89077_at
Hs.75367
NM_006748; Src-like-




adaptor


D89377_at
Hs.89404
NM_002449; msh




homeo box homolog 2


D90279_s_at
Hs.433695
NM_000093; alpha 1




type V collagen prepro-




protein


HG1996-HT2044_at


HG2090-HT2152_s_at


HG2463-HT2559_at


HG2994-HT4850_s_at
















TABLE 10







160 Genes for classifier









Chip acc. #
UniGene Build 162
Description





AF000231_at
Hs.75618
NM_004663; Ras-related protein Rab-11A


D13666_s_at
Hs.136348
NM_006475; osteoblast specific factor 2 (fasciclin I-like)


D21878_at
Hs.169998
NM_004334; bone marrow stromal cell antigen 1 precursor


D45370_at
Hs.74120
NM_006829; adipose specific 2


D49372_s_at
Hs.54460
NM_002986; small inducible cytokine A11 precursor


D83920_at
Hs.440898
NM_002003; ficolin 1 precursor


D85131_s_at
Hs.433881
NM_002383; MYC-associated zinc finger protein


D86062_s_at
Hs.413482
NM_004649; chromosome 21 open reading frame 33


D86479_at
Hs.439463
NM_001129; adipocyte enhancer binding protein 1 precursor


D86957_at
Hs.307944


D86976_at
Hs.196914


D87433_at
Hs.301989
NM_015136; stabilin 1


D89077_at
Hs.75367
NM_006748; Src-like-adaptor


D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2


HG3044-HT3742_s_at


HG371-HT26388_s_at


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome P450, family 4, subfamily B, poly-




peptide 1


J03040_at
Hs.111779
NM_003118; secreted protein, acidic, cysteine-rich (osteonectin)


J03068_at


J03241_s_at
Hs.2025
NM_003239; transforming growth factor, beta 3


J03278_at
Hs.307783
NM_002609; platelet-derived growth factor receptor beta




precursor


J03909_at


J04058_at
Hs.169919
NM_000126; electron transfer flavoprotein, alpha polypeptide


J04130_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor


J04162_at
Hs.372679
NM_000569; Fc fragment of IgG, low affinity IIIa, receptor for




(CD16)


J04456_at
Hs.407909
NM_002305; beta-galactosidase binding lectin precursor


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase


J05070_at
Hs.151738
NM_004994; matrix metalloproteinase 9 preproprotein


J05448_at
Hs.79402
NM_002694; DNA directed RNA polymerase II polypeptide C




NM_032940; DNA directed RNA polymerase II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or cysteine) proteinase inhibitor, clade A




(alpha-1 antiproteinase, antitrypsin), member 1


K03430_at


L13698_at
Hs.65029
NM_002048; growth arrest-specific 1


L13720_at
Hs.437710
NM_000820; growth arrest-specific 6


L13923_at
Hs.750
NM_000138; fibrillin 1


L15409_at
Hs.421597
NM_000551; elogin binding protein


L17325_at
Hs.195825
NM_006867; RNA-binding protein with multiple splicing


L19872_at
Hs.170087
NM_001621; aryl hydrocarbon receptor


L27476_at
Hs.75608
NM_004817; tight junction protein 2 (zona occludens 2)


L33799_at
Hs.202097
NM_002593; procollagen C-endopeptidase enhancer


L40388_at
Hs.30212
NM_004236; thyroid receptor interacting protein 15


L40904_at
Hs.387667
NM_005037; peroxisome proliferative activated receptor




gamma isoform 1 NM_015869; peroxisome proliferative activated




receptor gamma isoform 2 NM_138711; peroxisome




proliferative activated receptor gamma isoform 1 NM_138712;




peroxisome proliferative activated receptor gamma isoform 1


L41919_rna1_at


M11433_at
Hs.101850
NM_002899; retinol binding protein 1, cellular


M11718_at
Hs.283393
NM_000393; alpha 2 type V collagen preproprotein


M12125_at
Hs.300772
NM_003289; tropomyosin 2 (beta)


M14218_at
Hs.442047
NM_000048; argininosuccinate lyase


M15395_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


M16591_s_at
Hs.89555
NM_002110; hemopoietic cell kinase isoform p61HCK


M17219_at
Hs.203862
NM_002069; guanine nucleotide binding protein (G protein),




alpha inhibiting activity polypeptide 1


M20530_at


M23178_s_at
Hs.73817
NM_002983; chemokine (C-C motif) ligand 3


M28130_rna1_s_at


M29550_at
Hs.187543
NM_021132; protein phosphatase 3 (formerly 2B), catalytic




subunit, beta isoform (calcineurin A beta)


M31165_at
Hs.407546
NM_007115; tumor necrosis factor, alpha-induced protein 6




precursor


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2


M33195_at
Hs.433300
NM_004106; Fc fragment of IgE, high affinity I, receptor for,




gamma polypeptide precursor


M37033_at
Hs.443057
NM_000560; CD53 antigen


M37766_at
Hs.901
NM_001778; CD48 antigen (B-cell membrane protein)


M55998_s_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein


M57731_s_at
Hs.75765
NM_002089; chemokine (C—X—C motif) ligand 2


M62840_at
Hs.82542
NM_001637; acyloxyacyl hydrolase precursor


M63262_at


M68840_at
Hs.183109
NM_000240; monoamine oxidase A


M69203_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor


M72885_rna1_s_at


M77349_at
Hs.421496
NM_000358; transforming growth factor, beta-induced, 68 kDa


M82882_at
Hs.124030
NM_172373; E74-like factor 1 (ets domain transcription factor)


M83822_at
Hs.209846
NM_006726; LPS-responsive vesicle trafficking, beach and




anchor containing


M92934_at
Hs.410037
NM_001901; connective tissue growth factor


M95178_at
Hs.119000
NM_001102; actinin, alpha 1


S69115_at
Hs.10306
NM_005601; natural killer cell group 7 sequence


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


S78187_at
Hs.153752
NM_004358; cell division cycle 25B isoform 1 NM_021872;




cell division cycle 25B isoform 2 NM_021873; cell division




cycle 25B isoform 3 NM_021874; cell division cycle 25B isoform 4


U01833_at
Hs.81469
NM_002484; nucleotide binding protein 1 (MinD homolog, E. coli)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1


U09278_at
Hs.436852
NM_004460; fibroblast activation protein, alpha subunit


U09937_rna1_s_at


U10550_at
Hs.79022
NM_005261; GTP-binding mitogen-induced T-cell protein




NM_181702; GTP-binding mitogen-induced T-cell protein


U12424_s_at
Hs.108646
NM_000408; glycerol-3-phosphate dehydrogenase 2 (mitochondrial)


U16306_at
Hs.434488
NM_004385; chondroitin sulfate proteoglycan 2 (versican)


U20158_at
Hs.2488
NM_005565; lymphocyte cytosolic protein 2


U20536_s_at
Hs.3280
NM_001226; caspase 6 isoform alpha preproprotein




NM_032992; caspase 6 isoform beta


U24266_at
Hs.77448
NM_003748; aldehyde dehydrogenase 4A1 precursor




NM_170726; aldehyde dehydrogenase 4A1 precursor


U28249_at
Hs.301350
NM_005971; FXYD domain containing ion transport regulator




3 isoform 1 precursor NM_021910; FXYD domain containing




ion transport regulator 3 isoform 2 precursor


U28488_s_at
Hs.155935
NM_004054; complement component 3a receptor 1


U29680_at
Hs.227817
NM_004049; BCL2-related protein A1


U37143_at
Hs.152096
NM_000775; cytochrome P450, family 2, subfamily J, polypeptide 2


U38864_at
Hs.108139
NM_012256; zinc finger protein 212


U39840_at
Hs.163484
NM_004496; forkhead box A1


U41315_rna1_s_at


U44111_at
Hs.42151
NM_006895; histamine N-methyltransferase


U47414_at
Hs.13291
NM_004354; cyclin G2


U49352_at
Hs.414754
NM_001359; 2,4-dienoyl CoA reductase 1 precursor


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase E1,




beta polypeptide precursor NM_183050; branched chain keto




acid dehydrogenase E1, beta polypeptide precursor


U52101_at
Hs.9999
NM_001425; epithelial membrane protein 3


U59914_at
Hs.153863
NM_005585; MAD, mothers against decapentaplegic homolog 6


U60205_at
Hs.393239
NM_006745; sterol-C4-methyl oxidase-like


U61981_at
Hs.42674
NM_002439; mutS homolog 3


U64520_at
Hs.66708
NM_004781; vesicle-associated membrane protein 3 (cellubrevin)


U65093_at
Hs.82071
NM_006079; Cbp/p300-interacting transactivator, with




Glu/Asp-rich carboxy-terminal domain, 2


U66619_at
Hs.444445
NM_003078; SWI/SNF-related matrix-associated actin-




dependent regulator of chromatin d3


U68019_at
Hs.288261
NM_005902; MAD, mothers against decapentaplegic homolog 3


U68385_at
Hs.380923


U68485_at
Hs.193163
NM_004305; bridging integrator 1 isoform 8 NM_139343;




bridging integrator 1 isoform 1 NM_139344; bridging integrator




1 isoform 2 NM_139345; bridging integrator 1 isoform 3




NM_139346; bridging integrator 1 isoform 4 NM_139347;




bridging integrator 1 isoform 5 NM_139348; bridging integrator




1 isoform 6 NM_139349; bridging integrator 1 isoform 7




NM_139350; bridging integrator 1 isoform 9 NM_139351;




bridging integrator 1 isoform 10


U74324_at
Hs.90875
NM_002871; RAB-interacting factor


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


U83303_cds2_at
Hs.164021
NM_002993; chemokine (C—X—C motif) ligand 6 (granulocyte




chemotactic protein 2)


U88871_at
Hs.79993
NM_000288; peroxisomal biogenesis factor 7


U90549_at
Hs.236774
NM_006353; high mobility group nucleosomal binding domain 4


U90716_at
Hs.79187
NM_001338; coxsackie virus and adenovirus receptor


V00594_at
Hs.118786
NM_005953; metallothionein 2A


V00594_s_at
Hs.118786
NM_005953; metallothionein 2A


X02761_s_at
Hs.418138
NM_002026; fibronectin 1 isoform 1 preproprotein




NM_054034; fibronectin 1 isoform 2 preproprotein


X04011_at
Hs.88974
NM_000397; cytochrome b-245, beta polypeptide (chronic




granulomatous disease)


X04085_rna1_at


X07438_s_at


X07743_at
Hs.77436
NM_002664; pleckstrin


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor


X14046_at
Hs.153053
NM_001774; CD37 antigen


X14813_at
Hs.166160
NM_001607; acetyl-Coenzyme A acyltransferase 1


X15880_at
Hs.415997
NM_001848; collagen, type VI, alpha 1 precursor


X15882_at
Hs.420269
NM_001849; alpha 2 type VI collagen isoform 2C2 precursor




NM_058174; alpha 2 type VI collagen isoform 2C2a precursor




NM_058175; alpha 2 type VI collagen isoform 2C2a precursor


X51408_at
Hs.380138
NM_001822; chimerin (chimaerin) 1


X53800_s_at
Hs.89690
NM_002090; chemokine (C—X—C motif) ligand 3


X54489_rna1_at


X57351_s_at
Hs.174195
NM_006435; interferon induced transmembrane protein 2 (1-




8D)


X57579_s_at


X58072_at
Hs.169946
NM_002051; GATA binding protein 3 NM_032742;


X62048_at
Hs.249441
NM_003390; wee1 tyrosine kinase


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


X65614_at
Hs.2962
NM_005980; S100 calcium binding protein P


X66945_at
Hs.748
NM_000604; fibroblast growth factor receptor 1 isoform 1




precursor NM_015850; fibroblast growth factor receptor 1




isoform 2 precursor NM_023105; fibroblast growth factor




receptor 1 isoform 3 precursor NM_023106; fibroblast growth




factor receptor 1 isoform 4 precursor NM_023107; fibroblast




growth factor receptor 1 isoform 5 precursor NM_023108;




fibroblast growth factor receptor 1 isoform 6 precursor




NM_023109; fibroblast growth factor receptor 1 isoform 7




precursor NM_023110; fibroblast growth factor receptor 1




isoform 8 precursor NM_023111; fibroblast growth factor




receptor 1 isoform 9 precursor


X67491_f_at
Hs.355697
NM_005271; glutamate dehydrogenase 1


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


X78549_at
Hs.51133
NM_005975; PTK6 protein tyrosine kinase 6


X78565_at
Hs.98998
NM_002160; tenascin C (hexabrachion)


X78669_at
Hs.79088
NM_002902; reticulocalbin 2, EF-hand calcium binding domain


X83618_at
Hs.59889
NM_005518; 3-hydroxy-3-methylglutaryl-Coenzyme A synthase




2 (mitochondrial)


X84908_at
Hs.78060
NM_000293; phosphorylase kinase, beta


X90908_at
Hs.147391
NM_001445; gastrotropin


X91504_at
Hs.389277
NM_003224; ADP-ribosylation factor related protein 1


X95632_s_at
Hs.387906
NM_005759; abl-interactor 2


X97267_rna1_s_at


Y00705_at
Hs.407856
NM_003122; serine protease inhibitor, Kazal type 1


Y00787_s_at
Hs.624
NM_000584; interleukin 8 precursor


Y00815_at
Hs.75216
NM_002840; protein tyrosine phosphatase, receptor type, F




isoform 1 precursor NM_130440; protein tyrosine phosphatase,




receptor type, F isoform 2 precursor


Y08374_rna1_at


Z12173_at
Hs.334534
NM_002076; glucosamine (N-acetyl)-6-sulfatase precursor


Z19554_s_at
Hs.435800
NM_003380; vimentin


Z26491_s_at
Hs.240013
NM_000754; catechol-O-methyltransferase isoform MB-COMT




NM_007310; catechol-O-methyltransferase isoform S-COMT


Z29331_at
Hs.372758
NM_003344; ubiquitin-conjugating enzyme E2H isoform 1




NM_182697; ubiquitin-conjugating enzyme E2H isoform 2


Z35491_at
Hs.377484
NM_004323; BCL2-associated athanogene isoform 1L


Z48199_at
Hs.82109
NM_002997; syndecan 1


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
















TABLE 11







80 genes for classifier









Chip acc. #
UniGene Build 162
Description





AF000231_at
Hs.75618
NM_004663; Ras-related protein Rab-11A


D13666_s_at
Hs.136348
NM_006475; osteoblast specific factor 2 (fasciclin I-like)


D49372_s_at
Hs.54460
NM_002986; small inducible cytokine A11 precursor


D83920_at
Hs.440898
NM_002003; ficolin 1 precursor


D86479_at
Hs.439463
NM_001129; adipocyte enhancer binding protein 1 precursor


D87433_at
Hs.301989
NM_015136; stabilin 1


D89077_at
Hs.75367
NM_006748; Src-like-adaptor


D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2


HG4069-HT4339_s_at


HG67-HT67_f_at


HG907-HT907_at


J02871_s_at
Hs.436317
NM_000779; cytochrome P450, family 4, subfamily B, poly-




peptide 1


J03278_at
Hs.307783
NM_002609; platelet-derived growth factor receptor beta




precursor


J04058_at
Hs.169919
NM_000126; electron transfer flavoprotein, alpha polypeptide


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase


J05070_at
Hs.151738
NM_004994; matrix metalloproteinase 9 preproprotein


J05448_at
Hs.79402
NM_002694; DNA directed RNA polymerase II polypeptide C




NM_032940; DNA directed RNA polymerase II polypeptide C


K01396_at
Hs.297681
NM_000295; serine (or cysteine) proteinase inhibitor, clade A




(alpha-1 antiproteinase, antitrypsin), member 1


L13720_at
Hs.437710
NM_000820; growth arrest-specific 6


L40904_at
Hs.387667
NM_005037; peroxisome proliferative activated receptor




gamma isoform 1 NM_015869; peroxisome proliferative activated




receptor gamma isoform 2 NM_138711; peroxisome




proliferative activated receptor gamma isoform 1 NM_138712;




peroxisome proliferative activated receptor gamma isoform 1


M12125_at
Hs.300772
NM_003289; tropomyosin 2 (beta)


M15395_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


M16591_s_at
Hs.89555
NM_002110; hemopoietic cell kinase isoform p61HCK


M20530_at


M23178_s_at
Hs.73817
NM_002983; chemokine (C-C motif) ligand 3


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2


M33195_at
Hs.433300
NM_004106; Fc fragment of IgE, high affinity I, receptor for,




gamma polypeptide precursor


M55998_s_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein


M57731_s_at
Hs.75765
NM_002089; chemokine (C—X—C motif) ligand 2


M63262_at


M68840_at
Hs.183109
NM_000240; monoamine oxidase A


M69203_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor


M72885_rna1_s_at


M83822_at
Hs.209846
NM_006726; LPS-responsive vesicle trafficking, beach and




anchor containing


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


U01833_at
Hs.81469
NM_002484; nucleotide binding protein 1 (MinD homolog, E. coli)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1


U09937_rna1_s_at


U10550_at
Hs.79022
NM_005261; GTP-binding mitogen-induced T-cell protein




NM_181702; GTP-binding mitogen-induced T-cell protein


U20158_at
Hs.2488
NM_005565; lymphocyte cytosolic protein 2


U28488_s_at
Hs.155935
NM_004054; complement component 3a receptor 1


U29680_at
Hs.227817
NM_004049; BCL2-related protein A1


U41315_rna1_s_at


U47414_at
Hs.13291
NM_004354; cyclin G2


U49352_at
Hs.414754
NM_001359; 2,4-dienoyl CoA reductase 1 precursor


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase E1,




beta polypeptide precursor NM_183050; branched chain keto




acid dehydrogenase E1, beta polypeptide precursor


U52101_at
Hs.9999
NM_001425; epithelial membrane protein 3


U59914_at
Hs.153863
NM_005585; MAD, mothers against decapentaplegic homolog 6


U64520_at
Hs.66708
NM_004781; vesicle-associated membrane protein 3 (cellubrevin)


U65093_at
Hs.82071
NM_006079; Cbp/p300-interacting transactivator, with




Glu/Asp-rich carboxy-terminal domain, 2


U68019_at
Hs.288261
NM_005902; MAD, mothers against decapentaplegic homolog 3


U68385_at
Hs.380923


U74324_at
Hs.90875
NM_002871; RAB-interacting factor


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


U90549_at
Hs.236774
NM_006353; high mobility group nucleosomal binding domain 4


X04085_rna1_at


X07438_s_at


X07743_at
Hs.77436
NM_002664; pleckstrin


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor


X14046_at
Hs.153053
NM_001774; CD37 antigen


X15880_at
Hs.415997
NM_001848; collagen, type VI, alpha 1 precursor


X15882_at
Hs.420269
NM_001849; alpha 2 type VI collagen isoform 2C2 precursor




NM_058174; alpha 2 type VI collagen isoform 2C2a precursor




NM_058175; alpha 2 type VI collagen isoform 2C2a precursor


X51408_at
Hs.380138
NM_001822; chimerin (chimaerin) 1


X53800_s_at
Hs.89690
NM_002090; chemokine (C—X—C motif) ligand 3


X54489_rna1_at


X57579_s_at


X62048_at
Hs.249441
NM_003390; wee1 tyrosine kinase


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


X67491_f_at
Hs.355697
NM_005271; glutamate dehydrogenase 1


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


X97267_rna1_s_at


Y00787_s_at
Hs.624
NM_000584; interleukin 8 precursor


Z12173_at
Hs.334534
NM_002076; glucosamine (N-acetyl)-6-sulfatase precursor


Z19554_s_at
Hs.435800
NM_003380; vimentin


Z26491_s_at
Hs.240013
NM_000754; catechol-O-methyltransferase isoform MB-COMT




NM_007310; catechol-O-methyltransferase isoform S-COMT


Z29331_at
Hs.372758
NM_003344; ubiquitin-conjugating enzyme E2H isoform 1




NM_182697; ubiquitin-conjugating enzyme E2H isoform 2


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
















TABLE 12







40 genes for classifier









Chip acc. #
UniGene Build 162
description





D83920_at
Hs.440898
NM_002003; ficolin 1 precursor


D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2


J02871_s_at
Hs.436317
NM_000779; cytochrome P450, family 4, subfamily B, poly-




peptide 1


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase


J05070_at
Hs.151738
NM_004994; matrix metalloproteinase 9 preproprotein


M16591_s_at
Hs.89555
NM_002110; hemopoietic cell kinase isoform p61HCK


M23178_s_at
Hs.73817
NM_002983; chemokine (C-C motif) ligand 3


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2


M33195_at
Hs.433300
NM_004106; Fc fragment of IgE, high affinity I, receptor for,




gamma polypeptide precursor


M57731_s_at
Hs.75765
NM_002089; chemokine (C—X—C motif) ligand 2


M68840_at
Hs.183109
NM_000240; monoamine oxidase A


M69203_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


U01833_at
Hs.81469
NM_002484; nucleotide binding protein 1 (MinD homolog, E. coli)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1


U09937_rna1_s_at


U20158_at
Hs.2488
NM_005565; lymphocyte cytosolic protein 2


U41315_rna1_s_at


U47414_at
Hs.13291
NM_004354; cyclin G2


U49352_at
Hs.414754
NM_001359; 2,4-dienoyl CoA reductase 1 precursor


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase E1,




beta polypeptide precursor NM_183050; branched chain keto




acid dehydrogenase E1, beta polypeptide precursor


U65093_at
Hs.82071
NM_006079; Cbp/p300-interacting transactivator, with




Glu/Asp-rich carboxy-terminal domain, 2


U68385_at
Hs.380923


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


U90549_at
Hs.236774
NM_006353; high mobility group nucleosomal binding domain 4


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor


X15880_at
Hs.415997
NM_001848; collagen, type VI, alpha 1 precursor


X15882_at
Hs.420269
NM_001849; alpha 2 type VI collagen isoform 2C2 precursor




NM_058174; alpha 2 type VI collagen isoform 2C2a precursor




NM_058175; alpha 2 type VI collagen isoform 2C2a precursor


X51408_at
Hs.380138
NM_001822; chimerin (chimaerin) 1


X53800_s_at
Hs.89690
NM_002090; chemokine (C—X—C motif) ligand 3


X54489_rna1_at


X57579_s_at


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


X67491_f_at
Hs.355697
NM_005271; glutamate dehydrogenase 1


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


Z29331_at
Hs.372758
NM_003344; ubiquitin-conjugating enzyme E2H isoform 1




NM_182697; ubiquitin-conjugating enzyme E2H isoform 2


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
















TABLE 13







20 genes for classifier









Chip acc. #
UniGene Build 162
description





D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase


M23178_s_at
Hs.73817
NM_002983; chemokine (C-C motif) ligand 3


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2


M69203_s_at
Hs.75703
NM_002984; chemokine (C-C motif) ligand 4 precursor


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1


U41315_rna1_s_at


U47414_at
Hs.13291
NM_004354; cyclin G2


U49352_at
Hs.414754
NM_001359; 2,4-dienoyl CoA reductase 1 precursor


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase E1,




beta polypeptide precursor NM_183050; branched chain keto




acid dehydrogenase E1, beta polypeptide precursor


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor


X57579_s_at


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
















TABLE 14







10 genes for classifier









Chip acc. #
UniGene Build 162
description





D89377_at
Hs.89404
NM_002449; msh homeo box homolog 2


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


U41315_rna1_s_at


U47414_at
Hs.13291
NM_004354; cyclin G2


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein
















TABLE 15







32 genes for classifier









Chip acc. #
UniGene Build 162
description





D83920_at
Hs.440898
NM_002003; ficolin 1 precursor


HG67-HT67_f_at


HG907-HT907_at


J05032_at
Hs.32393
NM_001349; aspartyl-tRNA synthetase


K01396_at
Hs.297681
NM_000295; serine (or cysteine) proteinase inhibitor, clade A




(alpha-1 antiproteinase, antitrypsin), member 1


M16591_s_at
Hs.89555
NM_002110; hemopoietic cell kinase isoform p61HCK


M32011_at
Hs.949
NM_000433; neutrophil cytosolic factor 2


M33195_at
Hs.433300
NM_004106; Fc fragment of IgE, high affinity I, receptor for,




gamma polypeptide precursor


M37033_at
Hs.443057
NM_000560; CD53 antigen


M57731_s_at
Hs.75765
NM_002089; chemokine (C—X—C motif) ligand 2


M63262_at


S77393_at
Hs.145754
NM_016531; Kruppel-like factor 3 (basic)


U01833_at
Hs.81469
NM_002484; nucleotide binding protein 1 (MinD homolog, E. coli)


U07231_at
Hs.309763
NM_002092; G-rich RNA sequence binding factor 1


U41315_rna1_s_at


U47414_at
Hs.13291
NM_004354; cyclin G2


U50708_at
Hs.1265
NM_000056; branched chain keto acid dehydrogenase E1,




beta polypeptide precursor NM_183050; branched chain keto




acid dehydrogenase E1, beta polypeptide precursor


U52101_at
Hs.9999
NM_001425; epithelial membrane protein 3


U74324_at
Hs.90875
NM_002871; RAB-interacting factor


U77970_at
Hs.321164
NM_002518; neuronal PAS domain protein 2 NM_032235;


U90549_at
Hs.236774
NM_006353; high mobility group nucleosomal binding domain 4


X13334_at
Hs.75627
NM_000591; CD14 antigen precursor


X54489_rna1_at


X57579_s_at


X64072_s_at
Hs.375957
NM_000211; integrin beta chain, beta 2 precursor


X68194_at
Hs.80919
NM_006754; synaptophysin-like protein isoform a




NM_182715; synaptophysin-like protein isoform b


X73882_at
Hs.254605
NM_003980; microtubule-associated protein 7


X78520_at
Hs.372528
NM_001829; chloride channel 3


X95632_s_at
Hs.387906
NM_005759; abl-interactor 2


Z29331_at
Hs.372758
NM_003344; ubiquitin-conjugating enzyme E2H isoform 1




NM_182697; ubiquitin-conjugating enzyme E2H isoform 2


Z48605_at
Hs.421825
NM_006903; inorganic pyrophosphatase 2 isoform 2




NM_176865; NM_176866; inorganic pyrophosphatase 2




isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform 4




NM_176869; inorganic pyrophosphatase 2 isoform 1


Z74615_at
Hs.172928
NM_000088; alpha 1 type I collagen preproprotein









Recurrence Predictor

We furthermore tested an outcome predictor able to identify the likely presence or absence of recurrence in patients with superficial Ta tumours (see Table 16).


Table 16. Patient Disease Course Information—Recurrence Vs. No Recurrence


From the hierarchical cluster analysis of the tumour samples we found that the tumours with a high recurrence frequency were separated from the tumours with low recurrence frequency. To study this further we profiled two groups of Ta tumours-15 tumours with low recurrence frequency and 16 tumours with high recurrence frequency. To avoid influence from other tumour characteristics we only used tumours that showed the same growth pattern and tumours that showed no sign of concomitant carcinoma in situ. Furthermore, the tumours were all primary tumours. The tumours used for identifying genes differentially expressed in recurrent and non-recurrent tumours are listed in Table 16 below.









TABLE 16







Disease course information of all patients involved.












Group
Patient
Tumour (date)
Pattern
Carcinoma in situ
Time to recurrence





A
 968-1
Ta gr2
Papillary
no
27 month  


A
 928-1
Ta gr2
Papillary
no
38 month. 


A
 934-1
Ta gr2 (220798)
Papillary
no



A
 709-1
Ta gr2 (210798)
Papillary
no



A
 930-1
Ta gr2 (300698)
Papillary
no



A
 524-1
Ta gr2 (201095)
Papillary
no



A
 455-1
Ta gr2 (060695)
Papillary
no



A
 370-1
Ta gr2 (100195)
Papillary
no



A
 810-1
Ta gr2 (031097)
Papillary
no



A
1146-1
Ta gr2 (231199)
Papillary
no



A
1161-1
Ta gr2 (101299)
Mixed
no



A
1006-1
Ta gr2 (231198)
Papillary
no



A
 942-1
Ta gr2
Papillary
no
24 month. 


A
1060-1
Ta gr2
Papillary
no
36 month. 


A
1255-1
Ta gr2
Papillary
no
24 month. 


B
 441-1
Ta gr2
Papillary
no
6 month.


B
 780-1
Ta gr2
Papillary
no
2 month.


B
 815-2
Ta gr2
Papillary
no
6 month.


B
 829-1
Ta gr2
Papillary
no
4 month.


B
 861-1
Ta gr2
Papillary
no
4 month.


B
 925-1
Ta gr2
Papillary
no
5 month.


B
1008-1
Ta gr2
Papillary
no
5 month.


B
1086-1
Ta gr2
Papillary
no
6 month.


B
1105-1
Ta gr2
Papillary
no
8 month.


B
1145-1
Ta gr2
Papillary
no
4 month.


B
1327-1
Ta gr2
Papillary
no
5 month.


B
1352-1
Ta gr2
Papillary
no
6 month.


B
1379-1
Ta gr2
Papillary
no
5 month.


B
 533-1
Ta gr2
Papillary
no
4 month.


B
 679-1
Ta gr2
Papillary
no
4 month.


B
 692-1
Ta gr2
Papillary
no
5 month.





Group A: Primary tumours from patients with no recurrence of the disease for 2 years.


Group B: Primary tumours from patients with recurrence of the disease within 8 months.






Supervised Learning Prediction of Recurrence

In this part of the work we identified genes differentially expressed between non-recurring and recurring tumours. Cross-validation and prediction was performed as previously described, except that genes are selected based on the value of the Wilcoxon statistic for difference between the two groups.


Prediction Performance

The prediction performance was tested using from 1-200 genes in the cross-validation loops. FIG. 11 shows that the lowest error rate (8 errors) is obtained in e.g. the cross-validation model using from 39 genes. Based on this we selected this cross-validation model as our final predictor. The results of the predictions from the 39 gene cross-validation loops are listed in Table 17. The predictor misclassified four of the samples in each group and in one of the predictions the difference in the distances between the two group means is below the 5% difference limit as described above.


The probability of misclassifying 8 or less arrays by a random classification is 0.0053.









TABLE 17







Recurrence prediction results of 39 gene cross-validation loops.

















Prediction


Group
Patient
Tumour (date)
Prediction
Error
strength















A
968-1
Ta gr2
0

0.19


A
928-1
Ta gr2
0

0.49


A
934-1
Ta gr2 (220798)
0

1.73


A
709-1
Ta gr2 (210798)
0

0.45


A
930-1
Ta gr2 (300698)
0

0.82


A
524-1
Ta gr2 (201095)
0

0.14


A
455-1
Ta gr2 (060695)
1
*
0.68


A
370-1
Ta gr2 (100195)
0

0.32


A
810-1
Ta gr2 (031097)
0

0.45


A
1146-1 
Ta gr2 (231199)
0

0.98


A
1161-1 
Ta gr2 (101299)
0

0.03


A
1006-1 
Ta gr2 (231198)
1
*
1.57


A
942-1
Ta gr2
0

0.31


A
1060-1 
Ta gr2
1
*
0.81


A
1255-1 
Ta gr2
1
*
0.71


B
441-1
Ta gr2
1

1.03


B
780-1
Ta gr2
1

0.37


B
815-2
Ta gr2
1

0.35


B
829-1
Ta gr2
1

0.75


B
861-1
Ta gr2
0
*
2.55


B
925-1
Ta gr2
1

0.78


B
1008-1 
Ta gr2
0
*
0.12


B
1086-1 
Ta gr2
0
*
0.51


B
1105-1 
Ta gr2
1

0.37


B
1145-1 
Ta gr2
1

0.44


B
1327-1 
Ta gr2
1

1.96


B
1352-1 
Ta gr2
0
*
0.97


B
1379-1 
Ta gr2
1

0.67


B
533-1
Ta gr2
1

0.31


B
679-1
Ta gr2
1

0.82


B
692-1
Ta gr2
1

0.45





Group A: Primary tumours from patients with no recurrence of the disease for 2 years.


Group B: Primary tumours from patients with recurrence of the disease within 8 months.


Prediction, 0 = no recurrence, 1 = recurrence.






The optimal number of genes in cross-validation loops was found to be 39 (75% of the samples were correct classified, p<0.006) and from this we selected those 26 genes that were used in at least 75% of the cross-validation loops to constitute our final recurrence predictor. Consequently, this set of genes is to be used for predicting recurrence in independent samples. We tested the strength of the predictive genes by permutation analysis, see Table 18. We selected the genes used in at least 29 of the 31 cross-validation loops to constitute our final recurrence prediction model. The expression pattern of those 26 genes is shown in FIG. 12.









TABLE 18







The 26 genes that we find optimal for recurrence prediction.












Unigene





Feature
build 168
Description
Number
Test (W-N)





AF006041_at
Hs.336916
NM_001350; death-associated protein 6
31
0.054 (161-7)


D21337_at
Hs.408
NM_001847; type IV alpha 6 collagen
31
0.058 (160-6)




isoform A precursor NM_033641; type IV




alpha 6 collagen isoform B precursor


D49387_at
Hs.294584
NM_012212; NADP-dependent leukotriene
31
0.118 (313-8)




B4 12-hydroxydehydrogenase


D64154_at
Hs.90107
NM_007002; adhesion regulating molecule
31
0.078 (165-9)




1 precursor NM_175573; adhesion regulating




molecule 1 precursor


D83780_at
Hs.437991
NM_014846; KIAA0196 gene product
31
0.094 (159-4)


D87258_at
Hs.75111
NM_002775; protease, serine, 11
30
0.112 (168-11)


D87437_at
Hs.43660
NM_014837; chromosome 1 open reading
31
0.058 (160-6)




frame 16


HG1879-HT1919_at


31
0.122 (314-7)


HG3076-HT3238_s_at


31
0.080 (309-17)


HG511-HT511_at


31
0.348 (319-2)


L34155_at
Hs.83450
NM_000227; laminin alpha 3 subunit precursor
31
0.122 (314-7)


L38928_at
Hs.118131
NM_006441; 5,10-methenyltetrahydrofolate
29
0.348 (319-2)




synthetase (5-formyltetrahydrofolate cyclo-




ligase)


L49169_at
Hs.75678
NM_006732; FBJ murine osteosarcoma
31
0.108 (155-2)




viral oncogene homolog B


M16938_s_at
Hs.820
NM_004503; homeo box C6 isoform 1
29
 0.09 (170-16)




NM_153693; homeo box C6 isoform 2


M63175_at
Hs.295137
NM_001144; autocrine motility factor receptor
29
0.098 (308-18)




isoform a NM_138958; autocrine motility




factor receptor isoform b


M64572_at
Hs.405666
NM_002829; protein tyrosine phosphatase,
31
0.064 (305-31)




non-receptor type 3


M98528_at
Hs.79404
NM_014392; DNA segment on chromosome
31
0.122 (314-7)




4 (unique) 234 expressed sequence


U21858_at
Hs.60679
NM_003187; TBP-associated factor 9
31
0.122 (314-7)




NM_016283; adrenal gland protein AD-004


U45973_at
Hs.178347
NM_016532; skeletal muscle and kidney
31
0.094 (310-14)




enriched inositol phosphatase isoform 1




NM_130766; skeletal muscle and kidney




enriched inositol phosphatase isoform 2


U58516_at
Hs.3745
NM_005928; milk fat globule-EGF factor 8
29
0.100 (175-28)




protein


U62015_at
Hs.8867
NM_001554; cysteine-rich, angiogenic
31
0.106 (169-13)




inducer, 61


U66702_at
Hs.74624
NM_002847; protein tyrosine phosphatase,
31
0.146 (149-1)




receptor type, N polypeptide 2 isoform 1




precursor NM_130842; protein tyrosine




phosphatase, receptor type, N polypeptide




2 isoform 2 precursor NM_130843; protein




tyrosine phosphatase, receptor type, N




polypeptide 2 isoform 3 precursor


U70439_s_at
Hs.84264
NM_006401; acidic (leucine-rich) nuclear
30
 0.08 (309-17)




phosphoprotein 32 family, member B


U94855_at
Hs.381255
NM_003754; eukaryotic translation initiation
30
0.092 (311-12)




factor 3, subunit 5 epsilon, 47 kDa


X63469_at
Hs.77100
NM_002095; general transcription factor
31
0.092 (311-12)




IIE, polypeptide 2, beta 34 kDa


Z23064_at
Hs.380118
NM_002139; RNA binding motif protein, X
30
0.066 (307-24)




chromosome









Number: Number of times the gene has been used in a cross-validation loop. Test: The numbers in parenthesis are the value W of the Wilcoxon test statistic for no difference between the two groups together with the number N of genes for which the Wilcoxon test statistic is bigger than or equal to the value W. The test value is obtained from 500 permutations of the arrays. In each permutation we form new pseudogroups where both of the pseudogroups have the same proportion of arrays from the two original groups. For each permutation we count the number of genes for which the Wilcoxon test statistic based on the pseudogroups is bigger than or equal to W, and the test value is the proportion of the permutations for which this number is bigger than or equal to N. Thus the test value measures the significance of the observed value W. Consequently, for most of our selected genes we only find as least as good predictive genes in about 10% of the formed pseudogroups.


We present data on expression patterns that classify the benign and muscle-invasive bladder carcinomas. Furthermore, we can identify subgroups of bladder cancer such as Ta tumours with surrounding CIS, Ta tumours with a high probability of progression as well as recurrence, and T2 tumours with squamous metaplasia. As a novel finding, the matrix remodelling gene cluster was specifically expressed in the tumours having the worst prognosis, namely the T2 tumours and tumours surrounded by CIS. For some of these genes new small molecule inhibitors already exist (Kerr et al. 2002), and thus they form drug targets. At present it is not possible clinically to identify patients who will experience recurrence and not recurrence, but it would be a great benefit to both the patients and the health system by reducing the number of unnecessary control examinations in bladder tumour patients. To determine the optimal gene-set for separating non-recurrent and recurrent tumours, we again applied a cross-validation scheme using from 1-200 genes. We determined the optimal number of genes in cross-validation loops to be 39 (75% of the samples were correct classified, p<0.01, FIG. 11) and from this we selected those 26 genes (FIG. 12) that were used in at least 75% of the cross-validation loops to constitute our final recurrence predictor. Consequently, this set of genes is to be used for predicting recurrence in independent samples. We tested the strength of the predictive genes by performing 500 permutations of the arrays. This revealed that for most of our predictive genes we would only in a small number of the new pseudo-groups obtain at least as good predictors as in the real groups.


Biological Material

66 bladder tumour biopsies were sampled from patients following removal of the necessary amount of tissue for routine pathology examination. The tumours were frozen immediately after surgery and stored at −80° C. in a guanidinium thiocyanate solution. All tumours were graded according to Bergkvist et al. 1965 and re-evaluated by a single pathologist. As normal urothelial reference samples we used a pool of biopsies (from 37 patients) as well as three single bladder biopsies from patients with prostatic hyperplasia or urinary incontinence. Informed consent was obtained in all cases and protocols were approved by the local scientific ethical committee.


RNA Purification and cRNA Preparation


Total RNA was isolated from crude tumour biopsies using a Polytron homogenisator and the RNAzol B RNA isolation method (WAK-Chemie Medical GmbH). 10 μg total RNA was used as starting material for the cDNA preparation. The first and second strand cDNA synthesis was performed using the SuperScript Choice System (Life Technologies) according to the manufacturers instructions except using an oligo-dT primer containing a T7 RNA polymerase promoter site. Labelled cRNA was prepared using the BioArray High Yield RNA Transcript Labelling Kit (Enzo). Biotin labelled CTP and UTP (Enzo) were used in the reaction together with unlabeled NTP's. Following the IVT reaction, the unincorporated nucleotides were removed using RNeasy columns (Qiagen).


Array Hybridisation and Scanning

15 μg of cRNA was fragmented at 94° C. for 35 min in a fragmentation buffer containing 40 mM Tris-acetate pH 8.1, 100 mM KOAc, 30 mM MgOAc. Prior to hybridisation, the fragmented cRNA in a 6×SSPE-T hybridisation buffer (1 M NaCl, 10 mM Tris pH 7.6, 0.005% Triton), was heated to 95° C. for 5 min and subsequently to 45° C. for 5 min before loading onto the Affymetrix probe array cartridge (HuGeneFL). The probe array was then incubated for 16 h at 45° C. at constant rotation (60 rpm). The washing and staining procedure was performed in the Affymetrix Fluidics Station. The probe array was exposed to 10 washes in 6×SSPE-T at 25° C. followed by 4 washes in 0.5×SSPE-T at 50° C. The biotinylated cRNA was stained with a streptavidin-phycoerythrin conjugate, final concentration 2 μg/μl (Molecular Probes, Eugene, Oreg.) in 6×SSPE-T for 30 min at 25° C. followed by 10 washes in 6×SSPE-T at 25° C. The probe arrays were scanned at 560 nm using a confocal laser-scanning microscope (Hewlett Packard GeneArray Scanner G2500A). The readings from the quantitative scanning were analysed by the Affymetrix Gene Expression Analysis Software. An antibody amplification step followed using normal goat IgG as blocking reagent, final concentration 0.1 mg/ml (Sigma) and biotinylated anti-streptavidin antibody (goat), final concentration 3 μg/ml (Vector Laboratories). This was followed by a staining step with a streptavidin-phycoerythrin conjugate, final concentration 2 μg/μl (Molecular Probes, Eugene, Oreg.) in 6×SSPE-T for 30 min at 25° C. and 10 washes in 6×SSPE-T at 25° C. The arrays were then subjected to a second scan under similar conditions as described above.


Class Discovery Using Hierarchical Clustering

All microarray results were scaled to a global intensity of 150 units using the Affymetrix GeneChip software. Other ways of array normalisation exist (Li and Hung 2001), however, using the dCHIP approach did not change the expression profiles of the obtained classifier genes in this study (results not shown). For hierarchical cluster analysis and molecular classification procedures we used expression level ratios between tumours and the normal urothelium reference pool calculated using the comparison analysis implemented in the Affymetrix GeneChip software. In order to avoid expression ratios based on saturated gene-probes, we used the antibody amplified expression-data for genes with a mean Average Difference value across all samples below 1000 and the non-amplified expression-data for genes with values equal to or above 1000 in mean Average Difference value across all samples. Consequently, gene expression levels across all samples were either from the amplified or the non-amplified expression-data. We applied different filtering criteria to the expression data in order to avoid including non-varying and very low expressed genes in the data analysis. Firstly, we selected only genes that showed significant changes in expression levels compared to the normal reference pool in at least three samples. Secondly, only genes with at least three “Present” calls across all samples were selected. Thirdly, we eliminated genes varying less than 2 standard deviations across all samples. The final gene-set contained 1767 genes following filtering. Two-way hierarchical agglomerative cluster analysis was performed using the Cluster software25. We used average linkage clustering with a modified Pearson correlation as similarity metric. Genes and arrays were median centred and normalised to the magnitude of 1 prior to cluster analysis. The TreeView software was used for visualisation of the cluster analysis results (Eisen et al. 1998). Multidimensional scaling was performed on median centred and normalised data using an implementation in the SPSS statistical software package.


Tumour Stage Classifier

We based the classifier on the log-transformed expression level ratios. For these trans-formed values we used a normal distribution with the mean dependent on the gene and the group (Ta, T1, and T2, respectively) and the variance dependent on the gene only. For each gene we calculated the variation within the groups (W) and the three variations between two groups (B(Ta/T1), B(Ta/T2), B(T1/T2)) and used the three ratios B/W to select genes. We selected those genes having a high value of B(Ta/T1)/W, those genes having a high value of B(Ta/T2)/W, and those genes with a high value of B(T1/T2)/W. To classify a sample, we calculated the sum over the genes of the squared distance from the sample value to the group mean, standardised by the variance. Thus, we got a distance to each of the three groups and the sample was classified as belonging to the group in which the distance was smallest. When calculating these distances the group means and the variances were estimated from all the samples in the training set excluding the sample being classified.


Recurrence Prediction Using a Supervised Learning Method

Average Difference values were generated using the Affymetrix GeneChip software and all values below 20 were set to 20 to avoid very low and negative numbers. We only included genes that had a “Present” call in at least 7 samples and genes that showed intensity variation (Max−Min>100, Max/Min>2). The values were log transformed and rescaled. We used a supervised learning method essentially as described (Shipp et al. 2002). Genes were selected using t-test statistics and cross-validation and sample classification was performed as described above.


Immunohistochemistry

Tumour tissue microarrays were prepared essentially as described (Kononen et al. 1998), with four representative 0.6 mm paraffin cores from each study case. Immunohistochemical staining was performed using standard highly sensitive techniques after appropriate heat-induced antigen retrieval. Primary polyclonal goat antibodies against Smad 6 (S-20) and cyclin G2 (N-19) were from Santa Cruz Biotechnology. Antibodies to p53 (monoclonal DO-7) and Her-2 (polyclonal anti-c-erbB-2) were from Dako A/S. Ki-67 monoclonal antibody (MIBI) was from Novocastra Laboratories Ltd. Staining intensity was scored at four levels, Negative, Weak, Moderate and Strong by an experienced pathologist who considered both colour intensity and number of stained cells, and who was unaware of array results.


Example 3

A molecular classifier detects carcinoma in situ expression signatures in tumors and normal urothelium of the bladder.


Clinical Samples

Bladder tumour samples were obtained directly from surgery following removal of tissue for routine pathological examination. The samples were immediately submerged in a guadinium thiocyanate solution for RNA preservation and stored at −80° C. Informed consent was obtained in all cases, and the protocols were approved by the scientific ethical committee of Aarhus County. Samples in the No-CIS group were selected based on the following criteria: a) Ta tumours with no CIS in selected site biopsies in all visits; b) no previous muscle invasive tumour. Samples in the CIS group were selected based on the criteria: a) Ta or T1 tumours with CIS in selected site biopsies in any visit (preferable Ta tumours with CIS in the sampling visit); b) no previous muscle invasive tumours. Normal biopsies were obtained from individuals with prostatic hyperplasia or urinary incontinence. CIS and “normal” biopsies were obtained from cystectomy specimens directly following removal of the bladder. A grid was placed in the bladder for orientation and biopsies were taken from 8 positions covering the bladder surface. At each position, three biopsies were taken—two for pathologic examination and one in between these for RNA extraction for microarray expression profiling. The samples for RNA extraction were immediately transferred to the guadinium thiocyanate solution and stored at −80° C. until use. Samples used for RNA extraction were assumed to have CIS if CIS was detected in both adjacent biopsies. The “normal” samples were assumed to be normal if both adjacent biopsies were normal.


cRNA Preparation, Array Hybridisation and Scanning


Purification of total RNA, preparation of cRNA from cDNA and hybridisation and scanning were performed as previously described (Dyrskjot et al. 2003). The labelled samples were hybridised to Affymetrix U133A GeneChips.


Expression Data Analysis

Following scanning all data were normalised using the RMA normalisation approach in the Bioconductor Affy package to R. Variation filters were applied to the data to eliminate non-varying and presumably non-expressed genes. For gene-set 1 this was done by only including genes with a minimum expression above 200 in at least 5 samples and genes with max/min expression intensities above or equal to 3. The filtering for gene-set 2 including only genes with a minimum expression of 200 in at least 3 samples and genes with max/min expression intensities above or equal to 3. Average linkage hierarchical cluster analysis was carried out using the Cluster software with a modified Pearson correlation as similarity metric (Eisen et al. 1998). We used the TreeView software for visualisation of the cluster analysis results (Eisen et al. 1998). Genes were log-transformed, median centred and normalised to the magnitude of 1 before clustering. We used GeneCluster 2.0 (http://www-genome.wi.mit.edu/cancer/software/genecluster2/qc2.html) for the supervised selection of markers and for permutation testing. The algorithms used in the software are based on (Golub et al. 1999, Tamayo et al. 1999). Classifiers for CIS detection were built using the same methods as described previously (Dyrskjot et al. 2003).


Gene Expression Profiling

We used high-density oligonucleotide microarrays for gene expression profiling of approximately 22,000 genes in 28 superficial bladder tumour biopsies (13 tumours with surrounding CIS and 15 without surrounding CIS) and in 13 invasive carcinomas. See table 19 for patient disease course descriptions. Furthermore, expression profiles were obtained from 9 normal biopsies and from 10 biopsies from cystectomy specimens (5 histologically normal biopsies and 5 biopsies with CIS).









TABLE 19







Clinical data on patient disease courses and results of molecular CIS classification













Sample

Previous
Tumour
Subsequent




groupa
Patientb
tumours
analysed
tumours
CISc
CIS classifierd





1
1060-1

Ta gr2
2 Ta
No
No CIS


1
1146-1

Ta gr2

No
No CIS


1
1216-1

Ta gr2

No
No CIS


1
1303-1

Ta gr2

No
No CIS


1
 524-1

Ta gr2

No
No CIS


1
 692-1

Ta gr2
2 Ta
No
No CIS


1
1264-1

Ta gr3
20 Ta
No
No CIS


1
1350-1

Ta gr3
1 Ta
No
No CIS


1
1354-1

Ta gr3
11 T1
No
No CIS


1
 775-1

Ta gr3
1 Ta
No
No CIS


1
1066-1

Ta gr3
1 Ta
No
No CIS


1
1276-1

Ta gr3
2 T1
No
No CIS


1
1070-1

Ta gr3
1 Ta
No
No CIS


1
 989-1

Ta gr3

No
No CIS


1
1482-1

Ta gr3
20 Ta
No
CIS


2
1345-2
1 T1
Ta gr3

Sampling visit
CIS


2
1062-2

Ta gr3
1 T1
Sampling visit
CIS


2
 956-2

Ta gr3
1 Ta
Sampling visit
CIS


2
 320-7
1 Ta, 2 T1
Ta gr3
2 Ta
Sampling visit
CIS


2
1330-1

Ta gr3

Sampling visit
CIS


2
 602-8
5 Ta
Ta gr3
3 Ta
Sampling visit
CIS


2
 763-1

Ta gr2
14 Ta
Sampling visit
CIS


2
1024-1

T1 gr3
2 Ta, 1 T1
Sampling visit
CIS


2
1182-1

Ta gr3
7 Ta
Subsequent visit
CIS


2
1093-1

Ta gr3
4 Ta, 1 T1
Subsequent visit
CIS


2
 979-1

Ta gr3

Sampling visit
CIS


2
1337-1

T1 gr3

Sampling visit
CIS


2
1625-1

Ta gr2

Sampling visit
CIS


3
1015-1

T3b gr4

No



3
1337-1

T4a gr3

Sampling visit



3
1041-1

T4b gr3

No



3
1044-1

T4b gr3

ND



3
1055-1
1 Ta gr2
T3a gr3

No



3
1109-1

T2 gr3
1 T2-4
No



3
1124-1

T4a gr3
2 T2-4
No



3
1154-1

T3a gr3
1 Ta, 1 T2-4
No



3
1167-1
1 T2-4
T3b gr4
2 T2-4
ND



3
1178-1

T4b gr3

ND



3
1215-1

T4b gr3

ND



3
1271-1

T3b gr4

No



3
1321-1
1 T1
T3b gr?

ND







aThe tumour groups involved were TCC without CIS (1), TCC with CIS (2) and invasive TCC (3).




bThe numbers indicate the patient number followed by the clinic visit number.




cCIS in selected site biopsies in previous, present or subsequent visits to the clinic. ND: not determined.




dMolecular classification of the samples using 25 genes in cross-validation loops.







Hierarchical Cluster Analysis

Following appropriate normalisation and expression intensity calculations we selected those genes that showed high variation across the 41 TCC samples for further analysis. The filtering produced a gene-set consisting of 5,491 genes (gene-set 1) and two-way hierarchical cluster analysis was performed based on this gene-set. The sample clustering showed a separation of the three groups of samples with only few exceptions (FIG. 14a). Superficial TCC with surrounding CIS clustered in the one main branch of the dendrogram, while the superficial TCC without CIS and the invasive TCC clustered in two separate sub-branches in the other main branch of the dendrogram. The only exceptions were that the invasive TCC samples 1044-1 and 1124-1 clustered in the CIS group and two TCC with CIS clustered in the invasive group (samples 1330-1 and 956-2). The only TCC without CIS that clustered in the CIS group was sample 1482-1. The distinct clustering of the tumour groups indicated a large difference in gene expression patterns.


Hierarchical clustering of the genes (FIG. 14c) identified large clusters of genes characteristic for the each tumour phenotype. Cluster 1 showed a cluster of genes down-regulated in cystectomy biopsies, TCC with adjacent CIS and in some invasive carcinomas (FIG. 14c). There is no obvious functional relationship between the genes in this cluster. Cluster 2 showed a tight cluster of genes related to immunology and cluster 3 contained mostly genes expressed in muscle and connective tissue. Expression of genes in this cluster was observed in the normal and cystectomy samples, in a fraction of the TCC with CIS and in the invasive tumours. Cluster 4 contained genes up-regulated in the cystectomy biopsies, TCC with adjacent CIS and in invasive carcinomas (FIG. 14c). This cluster includes genes involved in cell cycle regulation, cell proliferation and apoptosis. However, for most of the genes in this cluster there is not apparent functional relationship either. Comparisons of chromosomal location of the genes in the clusters revealed no correlation between the observed gene clusters and chromosomal position of the identified genes. A positive correlation could have indicated chromosomal loss or gain or chromosomal inactivation by e.g. methylation of common promoter regions.


To analyse the impact of surrounding CIS lesions further we used the 28 superficial tumours only, and created a new gene set consisting of 5,252 varying genes (gene-set 2). Hierarchical cluster analysis of the tumour samples (FIG. 13b) based on the new gene-set separated the samples according to the presence of CIS in the surrounding urothelium with only 1 exception (P<0.000001, χ2-test). Sample 1482-1 clustered in the TCC with CIS group, however, no CIS has been detected in selected site biopsies during routine examinations of this patient. Tumour samples 1182-1 and 1093-1 did not have CIS in selected site biopsies in the same visit as the profiled tumour but showed this in later visits. However, the profile of these two superficial tumour samples already showed the adjacent CIS profile.


Marker Selection

To delineate the tumours with surrounding CIS from the tumours without CIS we used t-test statistics to select the 50 most up-regulated genes in each group (FIG. 15a). Permutation of the sample labels 500 times revealed that the 50 genes up-regulated in the CIS-group are highly significant differentially expressed and unlikely to find by chance, as all markers were significant on a 5% confidence level. Consequently, in 500 random datasets it was only possible to select as good genes in less than 5% of the datasets. The 50 genes up-regulated in the no-CIS group showed a poorer performance in the permutation tests, as these were not significant on a 5% confidence level. See Table 20 for details. The relative expression of these 100 genes is 9 normal and 10 biopsies from cystectomies with CIS are shown in FIG. 15b. The no-CIS profile was found in all of the normal samples. However, all histologically normal samples adjacent to the CIS lesions as well as the CIS biopsies showed the CIS profile.









TABLE 20







The best 100 markers














Feature


Perm
Perm
Perm




(U133 array)
Class
T-test
1%
5%
10%
UniGene Build 162
RefSeq; description

















221204_s_at
no_CIS
3.74
5.12
4.61
4.33
Hs.326444
NM_018058; cartilage









acidic protein 1


205927_s_at
no_CIS
3.67
4.53
3.98
3.73
Hs.1355
NM_001910; cathepsin E









isoform a preproprotein









NM_148964; cathepsin E









isoform b preproprotein


210143_at
no_CIS
3.35
4.03
3.73
3.45
Hs.188401
NM_007193; annexin A10


204540_at
no_CIS
3.15
3.87
3.51
3.32
Hs.433839
NM_001958; eukaryotic









translation elongation









factor 1 alpha 2


214599_at
no_CIS
3.02
3.75
3.37
3.14
Hs.157091
NM_005547; involucrin


203649_s_at
no_CIS
2.84
3.63
3.20
3.00
Hs.76422
NM_000300; phospholipase









A2, group IIA (platelets,









synovial fluid)


203980_at
no_CIS
2.74
3.47
3.12
2.89
Hs.391561
NM_001442; fatty acid









binding protein 4, adipocyte


209270_at
no_CIS
2.39
3.38
3.10
2.85
Hs.436983
NM_000228; laminin









subunit beta 3 precursor


206658_at
no_CIS
2.35
3.37
3.05
2.78
Hs.284211
NM_030570; uroplakin 3B









isoform a NM_182683;









uroplakin 3B isoform c









NM_182684; uroplakin 3B









isoform b


220779_at
no_CIS
2.35
3.33
2.97
2.73
Hs.149195
NM_016233; peptidylarginine









deiminase type









III


216971_s_at
no_CIS
2.28
3.29
2.91
2.71
Hs.79706
NM_000445; plectin 1,









intermediate filament









binding protein 500 kDa


206191_at
no_CIS
2.25
3.24
2.86
2.68
Hs.47042
NM_001248; ectonucleoside









triphosphate diphosphohydrolase 3


218484_at
no_CIS
2.18
3.20
2.81
2.62
Hs.221447
NM_020142;









NADH: ubiquinone oxidoreductase









MLRQ









subunit homolog


221854_at
no_CIS
2.1
3.19
2.80
2.60
Hs.313068
NM_000299; plakophilin 1


203792_x_at
no_CIS
2.02
3.16
2.74
2.55
Hs.371617
NM_007144; ring finger









protein 110


207862_at
no_CIS
2.01
3.16
2.72
2.52
Hs.379613
NM_006760; uroplakin 2


218960_at
no_CIS
1.93
3.14
2.65
2.47
Hs.414005
NM_019894; transmembrane









protease, serine 4









isoform 1 NM_183247;









transmembrane protease,









serine 4 isoform 2


203009_at
no_CIS
1.93
3.12
2.62
2.45
Hs.155048
NM_005581; Lutheran









blood group (Auberger b









antigen included)


204508_s_at
no_CIS
1.88
3.10
2.60
2.42
Hs.279916
NM_017689; hypothetical









protein FLJ20151


211692_s_at
no_CIS
1.87
3.06
2.58
2.39
Hs.87246
NM_014417; BCL2 binding









component 3


206465_at
no_CIS
1.86
3.04
2.54
2.38
Hs.277543
NM_015162; lipidosin


206122_at
no_CIS
1.85
2.92
2.52
2.36
Hs.95582
NM_006942; SRY-box 15


206393_at
no_CIS
1.83
2.89
2.49
2.33
Hs.83760
NM_003282; troponin I,









skeletal, fast


214639_s_at
no_CIS
1.79
2.87
2.49
2.30
Hs.67397
NM_005522; homeobox









A1 protein isoform a









NM_153620; homeobox









A1 protein isoform b


214630_at
no_CIS
1.79
2.84
2.44
2.28
Hs.184927
NM_000497; cytochrome









P450, subfamily XIB









(steroid 11-beta-









hydroxylase), polypeptide









1 precursor


204465_s_at
no_CIS
1.77
2.81
2.42
2.27
Hs.76888
NM_004692;









NM_032727; internexin









neuronal intermediate









filament protein, alpha


204990_s_at
no_CIS
1.76
2.79
2.41
2.24
Hs.85266
NM_000213; integrin,









beta 4


205453_at
no_CIS
1.75
2.77
2.39
2.22
Hs.290432
NM_002145; homeo box









B2


215812_s_at
no_CIS
1.74
2.77
2.37
2.20
Hs.499113
NM_018058; cartilage









acidic protein 1


217040_x_at
no_CIS
1.74
2.75
2.36
2.18
Hs.95582
NM_001910; cathepsin E









isoform a preproprotein









NM_148964; cathepsin E









isoform b preproprotein


203759_at
no_CIS
1.73
2.75
2.34
2.17
Hs.75268
NM_007193; annexin A10


211002_s_at
no_CIS
1.73
2.74
2.33
2.17
Hs.82237
NM_001958; eukaryotic









translation elongation









factor 1 alpha 2


216641_s_at
no_CIS
1.73
2.73
2.31
2.15
Hs.18141
NM_005547; involucrin


221660_at
no_CIS
1.71
2.67
2.30
2.13
Hs.247831
NM_000300; phospholipase









A2, group IIA (platelets,









synovial fluid)


220026_at
no_CIS
1.71
2.66
2.28
2.13
Hs.227059
NM_001442; fatty acid









binding protein 4, adipocyte


209591_s_at
no_CIS
1.69
2.63
2.28
2.11
Hs.170195
NM_000228; laminin









subunit beta 3 precursor


219922_s_at
no_CIS
1.68
2.61
2.26
2.08
Hs.289019
NM_030570; uroplakin 3B









isoform a NM_182683;









uroplakin 3B isoform c









NM_182684; uroplakin 3B









isoform b


201641_at
no_CIS
1.67
2.61
2.26
2.07
Hs.118110
NM_016233; peptidylarginine









deiminase type









III


204952_at
no_CIS
1.66
2.59
2.24
2.07
Hs.377028
NM_000445; plectin 1,









intermediate filament









binding protein 500 kDa


204487_s_at
no_CIS
1.65
2.59
2.23
2.06
Hs.367809
NM_001248; ectonucleoside









triphosphate diphosphohydrolase 3


210761_s_at
no_CIS
1.64
2.59
2.23
2.05
Hs.86859
NM_020142;









NADH: ubiquinone oxidoreductase









MLRQ









subunit homolog


217626_at
no_CIS
1.63
2.58
2.21
2.04
Hs.201967
NM_000299; plakophilin 1


204380_s_at
no_CIS
1.62
2.58
2.19
2.03
Hs.1420
NM_007144; ring finger









protein 110


205455_at
no_CIS
1.61
2.58
2.17
2.02
Hs.2942
NM_006760; uroplakin 2


205073_at
no_CIS
1.61
2.58
2.17
2.01
Hs.152096
NM_019894; transmembrane









protease, serine 4









isoform 1 NM_183247;









transmembrane protease,









serine 4 isoform 2


203287_at
no_CIS
1.61
2.58
2.16
2.00
Hs.18141
NM_005581; Lutheran









blood group (Auberger b









antigen included)


210735_s_at
no_CIS
1.58
2.55
2.15
1.99
Hs.5338
NM_017689; hypothetical









protein FLJ20151


203842_s_at
no_CIS
1.57
2.54
2.15
1.97
Hs.172740
NM_014417; BCL2 binding









component 3


206561_s_at
no_CIS
1.57
2.53
2.14
1.96
Hs.116724
NM_015162; lipidosin


214752_x_at
no_CIS
1.56
2.52
2.13
1.95
Hs.195464
NM_006942; SRY-box 15


217028_at
CIS
4.87
5.17
4.67
4.40
Hs.421986
NM_003282; troponin I,









skeletal, fast


213975_s_at
CIS
4.65
4.43
4.01
3.76
Hs.234734
NM_005522; homeobox









A1 protein isoform a









NM_153620; homeobox









A1 protein isoform b


201859_at
CIS
4.59
4.15
3.70
3.45
Hs.1908
NM_000497; cytochrome









P450, subfamily XIB









(steroid 11-beta-









hydroxylase), polypeptide









1 precursor


219410_at
CIS
4.49
3.98
3.49
3.29
Hs.104800
NM_004692;









NM_032727; internexin









neuronal intermediate









filament protein, alpha


207173_x_at
CIS
4.37
3.88
3.33
3.11
Hs.443435
NM_000213; integrin,









beta 4


214651_s_at
CIS
4.14
3.83
3.22
2.99
Hs.127428
NM_002145; homeo box









B2


201858_s_at
CIS
4.06
3.78
3.09
2.91
Hs.1908
NM_018058; cartilage









acidic protein 1


211430_s_at
CIS
4.03
3.63
3.05
2.83
Hs.413826
NM_001910; cathepsin E









isoform a preproprotein









NM_148964; cathepsin E









isoform b preproprotein


213891_s_at
CIS
3.86
3.63
3.02
2.77
Hs.359289
NM_007193; annexin A10


221872_at
CIS
3.82
3.52
2.89
2.73
Hs.82547
NM_001958; eukaryotic









translation elongation









factor 1 alpha 2


212386_at
CIS
3.77
3.50
2.87
2.69
Hs.359289
NM_005547; involucrin


211161_s_at
CIS
3.76
3.42
2.84
2.65

NM_000300; phospholipase









A2, group IIA (platelets,









synovial fluid)


214669_x_at
CIS
3.55
3.36
2.80
2.62
Hs.377975
NM_001442; fatty acid









binding protein 4, adipocyte


217388_s_at
CIS
3.44
3.31
2.79
2.58
Hs.444471
NM_000228; laminin









subunit beta 3 precursor


203477_at
CIS
3.36
3.28
2.75
2.56
Hs.409034
NM_030570; uroplakin 3B









isoform a NM_182683;









uroplakin 3B isoform c









NM_182684; uroplakin 3B









isoform b


204688_at
CIS
3.35
3.26
2.74
2.52
Hs.409798
NM_016233; peptidylarginine









deiminase type









III


218718_at
CIS
3.35
3.22
2.70
2.48
Hs.43080
NM_000445; plectin 1,









intermediate filament









binding protein 500 kDa


215176_x_at
CIS
3.32
3.14
2.67
2.45
Hs.503443
NM_001248; ectonucleoside









triphosphate diphosphohydrolase 3


201842_s_at
CIS
3.31
3.11
2.65
2.44
Hs.76224
NM_020142;









NADH: ubiquinone oxidoreductase









MLRQ









subunit homolog


212667_at
CIS
3.3
3.11
2.63
2.42
Hs.111779
NM_000299; plakophilin 1


209340_at
CIS
3.27
3.10
2.61
2.39
Hs.21293
NM_007144; ring finger









protein 110


215379_x_at
CIS
3.26
3.10
2.59
2.39
Hs.449601
NM_006760; uroplakin 2


200762_at
CIS
3.25
3.05
2.56
2.34
Hs.173381
NM_019894; transmembrane









protease, serine 4









isoform 1 NM_183247;









transmembrane protease,









serine 4 isoform 2


211896_s_at
CIS
3.21
3.05
2.53
2.32
Hs.156316
NM_005581; Lutheran









blood group (Auberger b









antigen included)


204141_at
CIS
3.19
3.05
2.53
2.28
Hs.300701
NM_017689; hypothetical









protein FLJ20151


201744_s_at
CIS
3.18
3.03
2.50
2.27
Hs.406475
NM_014417; BCL2 binding









component 3


209138_x_at
CIS
3.17
3.03
2.47
2.24
Hs.505407
NM_015162; lipidosin


214677_x_at
CIS
3.14
3.02
2.47
2.23
Hs.449601
NM_006942; SRY-box 15


212077_at
CIS
3.11
2.99
2.46
2.21
Hs.443811
NM_003282; troponin I,









skeletal, fast


206392_s_at
CIS
3.11
2.98
2.43
2.20
Hs.82547
NM_005522; homeobox









A1 protein isoform a









NM_153620; homeobox









A1 protein isoform b


212998_x_at
CIS
3.09
2.94
2.40
2.19
Hs.375115
NM_000497; cytochrome









P450, subfamily XIB









(steroid 11-beta-









hydroxylase), polypeptide









1 precursor


201616_s_at
CIS
3.08
2.93
2.38
2.18
Hs.443811
NM_004692;









NM_032727; internexin









neuronal intermediate









filament protein, alpha


205382_s_at
CIS
3.07
2.88
2.37
2.15
Hs.155597
NM_000213; integrin,









beta 4


212671_s_at
CIS
3.07
2.85
2.35
2.14
Hs.387679
NM_002145; homeo box









B2


215121_x_at
CIS
3.06
2.84
2.34
2.13
Hs.356861
NM_018058; cartilage









acidic protein 1


200600_at
CIS
3.05
2.83
2.33
2.11
Hs.170328
NM_001910; cathepsin E









isoform a preproprotein









NM_148964; cathepsin E









isoform b preproprotein


202746_at
CIS
3.03
2.80
2.32
2.10
Hs.17109
NM_007193; annexin A10


202917_s_at
CIS
3
2.79
2.31
2.08
Hs.416073
NM_001958; eukaryotic









translation elongation









factor 1 alpha 2


201560_at
CIS
3
2.79
2.30
2.08
Hs.25035
NM_005547; involucrin


218918_at
CIS
2.99
2.77
2.29
2.06
Hs.8910
NM_000300; phospholipase









A2, group IIA (platelets,









synovial fluid)


218656_s_at
CIS
2.99
2.76
2.27
2.06
Hs.93765
NM_001442; fatty acid









binding protein 4, adipocyte


201088_at
CIS
2.99
2.76
2.26
2.04
Hs.159557
NM_000228; laminin









subunit beta 3 precursor


201291_s_at
CIS
2.97
2.75
2.25
2.04
Hs.156346
NM_030570; uroplakin 3B









isoform a NM_182683;









uroplakin 3B isoform c









NM_182684; uroplakin 3B









isoform b


215076_s_at
CIS
2.95
2.72
2.24
2.03
Hs.443625
NM_016233; peptidylarginine









deiminase type









III


212195_at
CIS
2.94
2.71
2.22
2.02
Hs.71968
NM_000445; plectin 1,









intermediate filament









binding protein 500 kDa


209732_at
CIS
2.94
2.68
2.22
2.00
Hs.85201
NM_001248; ectonucleoside









triphosphate diphosphohydrolase 3


212192_at
CIS
2.94
2.67
2.22
1.99
Hs.109438
NM_020142;









NADH: ubiquinone oxidoreductase









MLRQ









subunit homolog


221671_x_at
CIS
2.92
2.67
2.20
1.98
Hs.377975
NM_000299; plakophilin 1


211671_s_at
CIS
2.91
2.66
2.20
1.98
Hs.126608
NM_007144; ring finger









protein 110


214352_s_at
CIS
2.88
2.66
2.19
1.97
Hs.412107
NM_006760; uroplakin 2





Feature: Probe-set on U133A GeneChip


Class: The group in which the marker is up-regulated


T-test: The t-test value


Perm 1%: The 1% permutation level


Perm 5%: The 5% permutation level


Perm 10%: The 10% permutation level






Construction of a Molecular CIS Classifier

A classifier able to diagnose CIS from gene expressions in TCC or in bladder biopsies may increase the detection rate of CIS. Our first approach was to be able to classify superficial TCC with or without CIS in the surrounding mucosa. This could have the diverse effect that the number of random biopsies to be taken could be reduced.


We build a CIS-classifier as previously described (Dyrskjot et al. 2003) using cross-validation for determining the optimal number of genes for classifying CIS with fewest errors. The best classifier performance (1 error) was obtained in cross-validation loops using 25 genes (see FIG. 16); 16 of these were included in 70% of the cross-validation loops and these were selected to represent our final classifier for CIS diagnosis (FIG. 17a and table 21). Permutation analysis shoved that 13 of these were significant at a 1% confidence level—the remaining three genes were above a 10% confidence level.









TABLE 21







The 16 gene molecular classifier of CIS














Feature


Perm
Perm
Perm
UniGene Build



(U133a array)
Class
t-test
1%
5%
10%
162
RefSeq; description

















213633_at
no_CIS
1.51
2.46
2.04
1.85
Hs.97858
NM_018957; SH3-









domain binding protein 1


212784_at
no_CIS
1.36
2.27
1.86
1.70
Hs.388236
NM_015125; capicua









homolog


209241_x_at
no_CIS
1.13
1.78
1.48
1.33
Hs.112028
NM_015716; mis-









shapen/NIK-related









kinase isoform 1









NM_153827; mis-









shapen/NIK-related









kinase isoform 3









NM_170663; mis-









shapen/NIK-related









kinase isoform 2


217941_s_at
CIS
2.3
1.96
1.66
1.47
Hs.8117
NM_018695; erbb2









interacting protein


201877_s_at
CIS
2.27
1.90
1.62
1.45
Hs.249955
NM_002719; gamma









isoform of regulatory









subunit B56, protein









phosphatase 2A isoform









a NM_178586; gamma









isoform of regulatory









subunit B56, protein









phosphatase 2A isoform









b NM_178587; gamma









isoform of regulatory









subunit B56, protein









phosphatase 2A isoform









c NM_178588; gamma









isoform of regulatory









subunit B56, protein









phosphatase 2A isoform d


209630_s_at
CIS
1.97
1.54
1.31
1.15
Hs.444354
NM_012164; F-box and









WD-40 domain protein 2


202777_at
CIS
1.93
1.51
1.29
1.12
Hs.104315
NM_007373; soc-2









suppressor of clear









homolog


200958_s_at
CIS
1.92
1.49
1.28
1.11
Hs.164067
NM_005625; syndecan









binding protein









(syntenin)


209579_s_at
CIS
1.79
1.36
1.16
1.01
Hs.35947
NM_003925; methyl-









CpG binding domain









protein 4


209004_s_at
CIS
1.63
1.21
1.00
0.89
Hs.5548
NM_012161; F-box and









leucine-rich repeat









protein 5 isoform 1









NM_033535; F-box and









leucine-rich repeat









protein 5 isoform 2


218150_at
CIS
1.6
1.18
0.98
0.86
Hs.342849
NM_012097; ADP-









ribosylation factor-like 5









isoform 1 NM_177985;









ADP-ribosylation factor-









like 5 isoform 2


202076_at
CIS
1.53
1.12
0.92
0.82
Hs.289107
NM_001166; baculoviral









IAP repeat-containing









protein 2


204640_s_at
CIS
1.45
1.03
0.83
0.75
Hs.129951
NM_003563; speckle-









type POZ protein


201887_at
CIS
1.32
0.92
0.74
0.66
Hs.285115
NM_001560; interleukin









13 receptor, alpha 1









precursor


212802_s_at
CIS
1.31
0.91
0.72
0.65
Hs.287266


212899_at
CIS
1.29
0.89
0.71
0.64
Hs.129836
NM_015076; cyclin-









dependent kinase









(CDC2-like) 11





Feature: Probe-set on U133A GeneChip


Class: The group in which the marker is up-regulated


T-test: The t-test value


Perm 1%: The 1% permutation level


Perm 5%: The 5% permutation level


Perm 10%: The 10% permutation level






Exploration of Strength of CIS Classifier

To further explore the strength of classifying CIS we also built a classifier by randomly selecting half of the samples for training and used the other half for testing. Cross validation was used again in the training of this classifier for optimisation of the gene-set for classifying independent samples. Cross-validation with 15 genes showed a good performance (see FIG. 18) and 7 of these genes were included in 70% of the class-validation loops. These 7 genes classified the samples in the test set with one error only—sample 1482-1 (χ2-test, P<0.002). Only two of the genes were also included in the 16-gene classifier, which is understandable considering the number of tests performed and the limitations in sample size. This classification performance is notable considering the small number of samples used for training the classifier.


Grouping of Normal and Cystectomies with CIS


We used hierarchical cluster analysis to group the 9 normal and 10 biopsies from cystectomies with CIS based on the normalised expression profiles of the 16 classifier genes (FIG. 17b). This clustering separated the samples from cystectomies with CIS lesions from the normal samples with only few exceptions as 8 of the 10 biopsies from cystectomies were found in the one main branch of the dendrogram and 8 of the 9 normal biopsies were found on the other main branch (χ2-test, P<0.002).


Tables
Table B
REFERENCES



  • Pisani, P., Parkin, D. M., Bray, F. & Ferlay, J. Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83, 18-29 (1999).

  • Wolf, H. et al. Bladder tumors. Treated natural history. Prog Clin Biol Res 221, 223-55 (1986).

  • Althausen, A. F., Prout, G. R., Jr., and Daly, J. J. Non-invasive papillary carcinoma of the bladder associated with carcinoma in situ. J Urol, 116:575-580, 1976.

  • Spruck, C. H., 3rd, Ohneseit, P. F., Gonzalez-Zulueta, M., Esrig, D., Miyao, N., Tsai, Y. C., Lerner, S. P., Schmutte, C., Yang, A. S., Cote, R., and et al. Two molecular pathways to transitional cell carcinoma of the bladder. Cancer Res, 54: 784-788, 1994.

  • Rosin, M. P., Cairns, P., Epstein, J. I., Schoenberg, M. P., and Sidransky, D. Partial allelotype of carcinoma in situ of the human bladder. Cancer Res, 55: 5213-5216, 1995.

  • Anderstrom, C., Johansson, S., and Nilsson, S. The significance of lamina propria invasion on the prognosis of patients with bladder tumors. J Urol, 124:23-26, 1980.

  • Cummings, K. B. Carcinoma of the bladder: predictors. Cancer, 45:1849-1855, 1980.

  • Cheng, L., Cheville, J. C., Neumann, R. M., Leibovich, B. C., Egan, K. S., Spotts, B. E., and Bostwick, D. G. Survival of patients with carcinoma in situ of the urinary bladder. Cancer, 85:2469-2474, 1999.

  • Kriegmair, M., Baumgartner, R., Lumper, W., Waidelich, R., and Hofstetter, A. Early clinical experience with 5-aminolevulinic acid for the photodynamic therapy of superficial bladder cancer. Br J Urol, 77: 667-671, 1996.

  • van't Veer, L. J. et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530-6 (2002).

  • Dyrskjot, L. et al. Identifying distinct classes of bladder carcinoma using microarrays. Nat Genet 33, 90-6 (2003).

  • van Rhijn, B. W. et al. The fibroblast growth factor receptor 3 (FGFR3) mutation is a strong indicator of superficial bladder cancer with low recurrence rate. Cancer Res 61, 1265-8 (2001).

  • Yuan, J. et al. Polo-like kinase, a novel marker for cellular proliferation. Am J Pathol 150, 1165-72 (1997).

  • Galaktionov, K. & Beach, D. Specific activation of cdc25 tyrosine phosphatases by B-type cyclins: evidence for multiple roles of mitotic cyclins. Cell 67, 1181-94 (1991).

  • Weinstein, J., Jacobsen, F. W., Hsu-Chen, J., Wu, T. & Baum, L. G. A novel mammalian protein, p55CDC, present in dividing cells is associated with protein kinase activity and has homology to the Saccharomyces cerevisiae cell division cycle proteins Cdc20 and Cdc4. Mol Cell Biol 14, 3350-63 (1994).

  • Hiraiwa, A. et al. Immunolocalization of hCDC47 protein in normal and neoplastic human tissues and its relation to growth. Int J Cancer 74, 180-4 (1997).

  • Liu, J. & Kern, J. A. Neuregulin-1 activates the JAK-STAT pathway and regulates lung epithelial cell proliferation. Am J Respir Cell Mol Biol 27, 306-13 (2002).

  • Park, B. H., Vogelstein, B. & Kinzler, K. W. Genetic disruption of PPARdelta decreases the tumorigenicity of human colon cancer cells. Proc Natl Acad Sci USA 98, 2598-603 (2001).

  • Fauconnet, S. et al. Differential regulation of vascular endothelial growth factor expression by peroxisome proliferator-activated receptors in bladder cancer cells. J Biol Chem 277, 23534-43 (2002).

  • Golub, T. R. et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science 286, 531-7 (1999).

  • Ambrosini, G., Adida, C. & Altieri, D. C. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3, 917-21 (1997).

  • Sidransky, D. et al. Clonal origin bladder cancer. N Engl J Med 326, 737-40 (1992).

  • Primdahl, H. et al. Allelic imbalances in human bladder cancer: genome-wide detection with high-density single-nucleotide polymorphism arrays. J Natl Cancer Inst 94, 216-23 (2002).

  • Eaves, I. A. et al. Combining mouse congenic strains and microarray gene expression analyses to study a complex trait: the NOD model of type 1 diabetes. Genome Res 12, 232-43 (2002).

  • Ghandour, G. & Glynne, R. Method and apparatus for analysis of data from biomolecular arrays. International patent #WO0079465 (2000).

  • Turkey, J. Exploratory Data Analysis, (Addison-Wesley, Reading, Mass., 1977).

  • Kruhoffer, M., Magnusson, N. E., Aaboe, M., Dyrskjot, L. & Orntoft, T. F. Microarrays for gene expression profiling: Fabrication of Oligonucleotide microarrays, Isolation of RNA, Fluorescent labelling of cRNA, Hybridisation, and Scanning. in Cell Biology—A laboratory handbook (ed. Celis, J. E.) (To appear in the 4th edition of this book).

  • Jain, A. N. et al. Fully automatic quantification of microarray image data. Genome Res 12, 325-32 (2002).

  • Eisen, M. B., Spellman, P. T., Brown, P. O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA 95, 14863-8 (1998).

  • Seymour, L. Novel anti-cancer agents in development: exciting prospects and new challenges. Cancer Treat. Rev. 25, 301-312 (1999).

  • Fox, S. B., Gasparini, G., & Harris, A. L. Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to trial design and anticancer drugs. Lancet Oncol. 2, 278-289 (2001).

  • Shipp, M. A. et al. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat. Med. 8, 68-74 (2002).

  • Kerr, J. S., Slee, A. M., & Mousa, S. A. Small molecule alpha(v) integrin antagonists: novel anticancer agents. Expert. Opin. Investig. Drugs 9, 1271-1279 (2000).

  • Bergkvist, A., Ljungqvist, A., & Moberger, G. Classification of bladder tumours based on the cellular pattern. Preliminary report of a clinical-pathological study of 300 cases with a minimum follow-up of eight years. Acta Chir. Scand. 130, 371-378 (1965).

  • Li, C. & Hung, W. W. Model-based analysis of oligonucleotide arrays: model validation, design issues and standard error application. Genome Biol. 2, RESEARCH0032 (2001).

  • Kononen, J. et al. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat. Med. 4, 844-847 (1998).


Claims
  • 1. A method of predicting the prognosis of a biological condition in animal tissue, comprising collecting a sample comprising cells from the tissue and/or expression products from the cells,determining an expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562,correlating the expression level to at least one standard expression level to predict the prognosis of the biological condition in the animal tissue.
  • 2. The method of claim 1, wherein the animal tissue is selected from body organs.
  • 3. The method of claim 2, wherein the animal tissue is selected from epithelial tissue in body organs.
  • 4. The method of claim 3, wherein the animal tissue is selected from epithelial tissue in the urinary bladder.
  • 5. The method of claim 4, wherein the stage of the biological condition is selected from bladder cancer stages Ta, Carcinoma in situ (CIS), T1, T2, T3 and T4.
  • 6. The method of claim 5, comprising determining at least the expression of a Ta stage gene from a Ta stage gene group, at least one T1 stage gene from a T1 stage gene group, at least a T2 stage gene from a T2 stage gene group, at least a T3 stage gene from a T3 stage gene group, at least a T4 stage gene group from a T4 stage gene group, wherein at least one gene from each gene group is expressed in a significantly different amount in that stage than in one of the other stages.
  • 7. The method of claim 4, wherein the stage is bladder cancer stage Ta.
  • 8. The method of claim 4, wherein the animal tissue is mucosa.
  • 9. The method of claim 1, wherein the biological condition is an adenocarcinoma, a carcinoma, a teratoma, a sarcoma, and/or a lymphoma and/or carcinoma-in-situ, and/or dysplasia-in-situ.
  • 10. The method of claim 1, wherein the sample is a biopsy of the tissue or of metastasis originating from said tissue.
  • 11. (canceled)
  • 12. The method of claim 1, wherein the sample comprises substantially only cells from said tissue.
  • 13. The method according to claim 9, wherein the sample comprises substantially only cells from mucosa or tumors derived from said mucosa cells.
  • 14. The method of claim 1, wherein the gene from the group of genes is selected individually from the consisting of gene No. 1 to gene No. 188 (stages).
  • 15. The method of claim 1, wherein the gene from the group of genes is selected individually from the group consisting of gene No. 189 to gene No. 214 (recurrence).
  • 16. The method of claim 1, wherein the gene from the group of genes is selected individually from the group consisting of gene No. 215 to gene No. 232 (SCC).
  • 17. The method of claim 1, wherein the gene from the group of genes is selected individually from the group consisting of gene No. 233 to gene No. 446 (progression).
  • 18. The method of claim 1, wherein the gene from the group of genes is selected individually from the group consisting of gene No. 447 to gene No. 562 (CIS).
  • 19. The method of claim 1, wherein the expression level of at least two genes from the group of genes are determined.
  • 20. The method of claim 1, wherein the expression level of at least three genes from the group of genes are determined.
  • 21-23. (canceled)
  • 24. The method of claim 1, wherein the difference in expression level of a gene from the gene group to the at least one standard expression level is at least two-fold.
  • 25. The method of claim 1, wherein the difference in expression level of a gene from the gene group to the at least one standard expression is at least three-fold.
  • 26. The method of claim 1, wherein the difference in expression level of a gene from the gene group to the at least one standard expression is at least four-fold.
  • 27. The method of claim 1, wherein the expression level is determined by determining the mRNA of the cells.
  • 28. The method of claim 1, wherein the expression level is a) determined by determining expression products in the cells, or b) is determined by determining expression products in a body fluid.
  • 29. (canceled)
  • 30. The method of claim 1, wherein the stage of the biological condition has been determined prior to the prediction of the prognosis.
  • 31. The method claim 30, wherein the stage of the biological condition has been determined by histological examination of the tissue or by genotyping of the tissue.
  • 32. (canceled)
  • 33. The method of claim 31, wherein the stage of the biological condition has been determined by determining the expression of at least a first stage gene from a first stage gene group and/or at least a second stage gene from a second stage gene group, wherein at least one of said genes is expressed in said first stage of the condition in a higher amount than in said second stage, and the other gene is a expressed in said first stage of the condition in a lower amount than in said second stage of the condition,correlating the expression level of the assessed genes to a standard level of expression determining the stage of the condition.
  • 34. The method of claim 1, wherein the expression level of at least two genes is determined, by determining a first expression level of at least one gene from a first gene group, wherein the gene from the first gene group is selected from the group consisting of gene No. 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 369, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, and 444 (progressorgener), anddetermining a second expression level of at least one gene from a second gene group, wherein the second gene group is selected from the group consisting of genes No. 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, and 446 (non-progressorgener), andcorrelating the first expression level to a standard expression level for progressors, and/or the second expression level to a standard expression level for non-progressors to predict the prognosis of the biological condition in the animal tissue.
  • 35. A method of determining the stage of a biological condition in animal tissue, comprising collecting a sample comprising cells from the tissue, determining an expression level of at least one gene selected from the group of genes consisting of gene No 1 to gene No. 562correlating the expression level of the assessed genes to at least one standard level of expression determining the stage of the condition.
  • 36. The method of claim 35, wherein the expression level of at least two genes is determined by determining the expression of at least a first stage gene from a first stage gene group and at least a second stage gene from a second stage gene group, wherein at least one of said genes is expressed in said first stage of the condition in a higher amount than in said second stage, and the other gene is a expressed in said first stage of the condition in a lower amount than in said second stage of the condition, andcorrelating the expression level of the assessed genes to a standard level of expression determining the stage of the condition
  • 37. The method of claim 35, wherein the stage is selected from bladder cancer stages Ta, carcinoma in situ (CIS), T1, T2, T3 and T4.
  • 38. The method of claim 37, comprising determining at least the expression of a Ta stage gene from a Ta stage gene group, at least one T1 stage gene from a T1 stage gene group, at least a T2 stage gene from a T2 stage gene group, at least a T3 stage gene from a T3 stage gene group, or at least a T4 stage gene from a T4 stage gene group, wherein at least one gene from each gene group is expressed in a significantly different amount in that stage than in one of the other stages.
  • 39. The method of claim 38, wherein a Ta stage gene is selected individually from the group of Table B1.
  • 40. The method of claim 38, wherein a T1 stage gene is selected individually from the group of Table B2.
  • 41. The method of claim 38, wherein a T2 stage gene is selected individually from the group of Table B3.
  • 42. (canceled)
  • 43. A method of determining an expression pattern of a bladder cell sample, comprising: collecting sample comprising bladder cells and/or expression products from bladder cells,determining the expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562, and obtaining an expression pattern of the bladder cell sample.
  • 44. The method of 43, wherein the expression level of at least two genes are determined.
  • 45. The method of 43, wherein the expression level of at least three genes are determined.
  • 46-48. (canceled)
  • 49. The method of claim 43, wherein the genes exclude genes which are expressed in the submucosal, muscle, or connective tissue, whereby a pattern of expression is formed for the sample which is independent of the proportion of submucosal, muscle, or connective tissue cells in the sample.
  • 50. The method of claim 49, comprising determining the expression level of one or more genes in the sample comprising predominantly submucosal, muscle, and connective tissue cells, obtaining a second pattern, subtracting said second pattern from the expression pattern of the bladder cell sample, forming a third pattern of expression, said third pattern of expression reflecting expression of the bladder mucosa or bladder cancer cells independent of the proportion of submucosal, muscle, and connective tissue cells present in the sample.
  • 51. The method of claim 43, wherein the sample is a biopsy of the tissue.
  • 52. The method of claim 43, wherein the sample is a cell suspension.
  • 53. The method of claim 43, wherein the sample comprises substantially only cells from said tissue.
  • 54. The method according to claim 53, wherein the sample comprises substantially only cells from mucosa.
  • 55. A method of predicting the prognosis a biological condition in human bladder tissue comprising, collecting a sample comprising cells from the tissue,determining an expression pattern of a bladder cell sample, comprising:collecting sample comprising bladder cells and/or expression products from bladder cells,determining the expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562, and obtaining an expression pattern of the bladder cell sample,correlating the determined expression pattern to a reference pattern,predicting the prognosis of the biological condition of said tissue.
  • 56. A method for determining the stage of a biological condition in animal tissue comprising, collecting a sample comprising cells from the tissue,determining an expression pattern of a bladder cell sample, comprising:collecting sample comprising bladder cells and/or expression products from bladder cells,determining the expression level of at least one gene in the sample, said gene being selected from the group of genes consisting of gene No. 1 to gene No. 562, and obtaining an expression pattern of the bladder cell sample,correlating the determined expression pattern to a reference pattern,determining the stage of the biological condition is said tissue.
  • 57-71. (canceled)
  • 72. An assay for predicting the prognosis of a biological condition in animal tissue, comprising at least one first marker capable of detecting an expression level of at least one gene selected from the group of genes consisting of gene No. 1 to gene No. 562.
  • 73. The assay according to claim 72, wherein the marker is a nucleotide probe.
  • 74. The assay according to claim 72, wherein the marker is an antibody.
  • 75. The assay according to claim 72, comprising at least a first marker and/or a second marker, wherein the first marker is capable of detecting a gene from a first gene group, and/or the second marker is capable of detecting a gene from a second gene group, where the gene from the first group is selected from the group consisting of gene No. 237, 238, 239, 240, 241, 242, 243, 245, 246, 247, 248, 250, 253, 254, 257, 258, 260, 263, 264, 265, 267, 270, 271, 272, 278, 283, 284, 287, 288, 290, 291, 292, 294, 297, 298, 300, 302, 303, 305, 309, 310, 315, 316, 317, 318, 319, 321, 324, 329, 335, 336, 337, 339, 340, 344, 346, 347, 354, 356, 358, 359, 362, 364, 365, 368, 365, 371, 372, 377, 378, 379, 380, 381, 382, 383, 384, 388, 391, 393, 395, 396, 397, 399, 402, 403, 404, 409, 413, 417, 419, 420, 421, 422, 423, 425, 427, 429, 430, 431, 432, 437, and 444 (progressorgener), and where the gene from the second gene group is selected from the group consisting of genes No. 233, 234, 235, 236, 244, 249, 251, 252, 255, 256, 259, 261, 262, 266, 268, 269, 273, 274, 275, 276, 277, 279, 280, 281, 282, 285, 286, 289, 293, 295, 296, 299, 301, 304, 306, 307, 308, 311, 312, 313, 314, 320, 322, 323, 325, 326, 327, 328, 330, 331, 332, 333, 334, 338, 341, 342, 343, 345, 348, 349, 350, 351, 352, 353, 355, 357, 360, 361, 363, 366, 367, 370, 373, 374, 375, 376, 385, 386, 387, 389, 390, 392, 394, 398, 400, 401, 405, 406, 407, 408, 410, 411, 412, 414, 415, 416, 418, 424, 426, 428, 433, 434, 435, 436, 438, 439, 440, 441, 442, 443, 445, and 446 (non-progressorgener).
  • 76. The assay according to claim 72, said assay further comprising means for correlating the expression level of the at least one gene to a standard expression level and/or a reference expression pattern.
Priority Claims (1)
Number Date Country Kind
PA200201685 Nov 2002 DK national
Parent Case Info

This application is a continuation of U.S. patent application Ser. No. 10/533,547 filed Nov. 16, 2005, which is a US National Phase application of PCT/DK03/00750 filed Nov. 3, 2003, both of which are incorporated herein in their entirety.

Continuations (1)
Number Date Country
Parent 10533547 Nov 2005 US
Child 12180321 US