Gene expression in biological conditions

Information

  • Patent Application
  • 20060240426
  • Publication Number
    20060240426
  • Date Filed
    November 03, 2003
    20 years ago
  • Date Published
    October 26, 2006
    17 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, but 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 tumor suppressor 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. 143, 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 promotor regions, 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 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 unregulated 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 AGeneUnigene#GeneChipProbesetBuildUnigenedescriptionClassifier1HUGeneFLAB000220_at168Hs.171921sema domain, immunoglobulin domain (Ig),stageshort basic domain, secreted, (semaphorin)3C2HUGeneFLAF000231_at168Hs.75618RAB11A, member RAS oncogene familystage3HUGeneFLD10922_s_at168Hs.99855formyl peptide receptor-like 1stage4HUGeneFLD10925_at168Hs.301921chemokine (C—C motif) receptor 1stage5HUGeneFLD11086_at168Hs.84interleukin 2 receptor, gamma (severe combinedstageimmunodeficiency)6HUGeneFLD11151_at168Hs.211202endothelin receptor type Astage7HUGeneFLD13435_at168Hs.426142phosphatidylinositol glycan, class Fstage8HUGeneFLD13666_s_at168Hs.136348osteoblast specific factor 2 (fasciclin I-like)stage9HUGeneFLD14520_at168Hs.84728Kruppel-like factor 5 (intestinal)stage10HUGeneFLD21878_at168Hs.169998bone marrow stromal cell antigen 1stage11HUGeneFLD26443_at168Hs.371369solute carrier family 1 (glial high affinity glutamatestagetransporter), member 312HUGeneFLD42046_at168Hs.194665DNA2 DNA replication helicase 2-like (yeast)stage13HUGeneFLD45370_at168Hs.74120adipose specific 2stage14HUGeneFLD49372_s_at168Hs.54460chemokine (C—C motif) ligand 11stage15HUGeneFLD50495_at168Hs.224397transcription elongation factor A (SII), 2stage16HUGeneFLD63135_at168Hs.27935tweety homolog 2 (Drosophila)stage17HUGeneFLD64053_at168Hs.198288protein tyrosine phosphatase, receptor type, Rstage18HUGeneFLD83920_at168Hs.440898ficolin (collagen/fibrinogen domain containing) 1stage19HUGeneFLD85131_s_at168Hs.433881MYC-associated zinc finger protein (purine-stagebinding transcription factor)20HUGeneFLD86062_s_at168Hs.413482chromosome 21 open reading frame 33stage21HUGeneFLD86479_at168Hs.439463AE binding protein 1stage22HUGeneFLD86957_at168Hs.307944likely ortholog of mouse septin 8stage23HUGeneFLD86959_at168Hs.105751Ste20-related serine/threonine kinasestage24HUGeneFLD86976_at168Hs.196914minor histocompatibility antigen HA-1stage25HUGeneFLD87433_at168Hs.301989stabilin 1stage26HUGeneFLD87443_at168Hs.409862sorting nexin 19stage27HUGeneFLD87682_at168Hs.134792KIAA00241 proteinstage28HUGeneFLD89077_at168Hs.75367Src-like-adaptorstage29HUGeneFLD89377_at168Hs.89404msh homeo box homolog 2 (Drosophila)stage30HUGeneFLD90279_s_at168Hs.433695collagen, type V, alpha 1stage31HUGeneFLHG1996-HT2044_at168stage32HUGeneFLHG2090-HT2152_s_at168stage33HUGeneFLHG2463-HT2559_at168stage34HUGeneFLHG3044-HT3742_s_at168stage35HUGeneFLHG3187-HT3366_s_at168stage36HUGeneFLHG3342-HT3519_s_at168stage37HUGeneFLHG371-HT26388_s_at168stage38HUGeneFLHG4069-HT4339_s_at168stage39HUGeneFLHG67-HT67_f_at168stage40HUGeneFLHG907-HT907_at168stage41HUGeneFLJ02871_s_at168Hs.436317cytochrome P450, family 4, subfamily B,stagepolypeptide 142HUGeneFLJ03040_at168Hs.111779secreted protein, acidic, cysteine-rich (osteonectin)stage43HUGeneFLJ03060_at168stage44HUGeneFLJ03068_at168stage45HUGeneFLJ03241_s_at168Hs.2025transforming growth factor, beta 3stage46HUGeneFLJ03278_at168Hs.307783platelet-derived growth factor receptor, betastagepolypeptide47HUGeneFLJ03909_at168stage48HUGeneFLJ03925_at168Hs.172631integrin, alpha M (complement componentstagereceptor 3, alpha; also known as CD11b(p170), macrophage antigen alpha polypeptide)49HUGeneFLJ04056_at168Hs.88778carbonyl reductase 1stage50HUGeneFLJ04058_at168Hs.169919electron-transfer-flavoprotein, alpha polypeptidestage(glutaric aciduria II)51HUGeneFLJ04130_s_at168Hs.75703chemokine (C—C motif) ligand 4stage52HUGeneFLJ04152_ma1_s_at168stage53HUGeneFLJ04162_at168Hs.372679Fc fragment of IgG, low affinity IIIa, receptorstagefor (CD16)54HUGeneFLJ04456_at168Hs.407909lectin, galactoside-binding, soluble, 1 (galectinstage1)55HUGeneFLJ05032_at168Hs.32393aspartyl-tRNA synthetasestage56HUGeneFLJ05070_at168Hs.151738matrix metalloproteinase 9 (gelatinase B,stage92 kDa gelatinase, 92 kDa type IV collagenase)57HUGeneFLJ05448_at168Hs.79402polymerase (RNA) II (DNA directed) polypeptidestageC, 33 kDa58HUGeneFLK01396_at168Hs.297681serine (or cysteine) proteinase inhibitor, cladestageA (alpha-1 antiproteinase, antitrypsin), member 159HUGeneFLK03430_at168stage60HUGeneFLL06797_s_at168Hs.421986chemokine (C—X—C motif) receptor 4stage61HUGeneFLL10343_at168Hs.112341protease inhibitor 3, skin-derived (SKALP)stage62HUGeneFLL13391_at168Hs.78944regulator of G-protein signalling 2, 24 kDastage63HUGeneFLL13698_at168Hs.65029growth arrest-specific 1stage64HUGeneFLL13720_at168Hs.437710growth arrest-specific 6stage65HUGeneFLL13923_at168Hs.750fibrillin 1 (Marfan syndrome)stage66HUGeneFLL15409_at168Hs.421597von Hippel-Lindau syndromestage67HUGeneFLL17325_at168Hs.195825RNA binding protein with multiple splicingstage68HUGeneFLL19872_at168Hs.170087aryl hydrocarbon receptorstage69HUGeneFLL27476_at168Hs.75608tight junction protein 2 (zona occludens 2)stage70HUGeneFLL33799_at168Hs.202097procollagen C-endopeptidase enhancerstage71HUGeneFLL40388_at168Hs.30212thyroid receptor interacting protein 15stage72HUGeneFLL40904_at168Hs.387667peroxisome proliferative activated receptor,stagegamma73HUGeneFLL41919_ma1_at168stage74HUGeneFLM11433_at168Hs.101850retinol binding protein 1, cellularstage75HUGeneFLM11718_at168Hs.283393collagen, type V, alpha 2stage76HUGeneFLM12125_at168Hs.300772tropomyosin 2 (beta)stage77HUGeneFLM14218_at168Hs.442047argininosuccinate lyasestage78HUGeneFLM15395_at168Hs.375957integrin, beta 2 (antigen CD18 (p95), lymphocytestagefunction-associated antigen 1; macrophageantigen 1 (mac-1) beta subunit)79HUGeneFLM16591_s_at168Hs.89555hemopoietic cell kinasestage80HUGeneFLM17219_at168Hs.203862guanine nucleotide binding protein (G protein),stagealpha inhibiting activity polypeptide 181HUGeneFLM20530_at168stage82HUGeneFLM23178_s_at168Hs.73817chemokine (C—C motif) ligand 3stage83HUGeneFLM28130_ma1_s_at168stage84HUGeneFLM29550_at168Hs.187543protein phosphatase 3 (formerly 2B), catalyticstagesubunit, beta isoform (calcineurin A beta)85HUGeneFLM31165_at168Hs.407546tumor necrosis factor, alpha-induced protein 6stage86HUGeneFLM32011_at168Hs.949neutrophil cytosolic factor 2 (65 kDa, chronicstagegranulomatous disease, autosomal 2)87HUGeneFLM33195_at168Hs.433300Fc fragment of IgE, high affinity I, receptor for;stagegamma polypeptide88HUGeneFLM37033_at168Hs.443057CD53 antigenstage89HUGeneFLM37766_at168Hs.901CD48 antigen (B-cell membrane protein)stage90HUGeneFLM55998_s_at168Hs.172928collagen, type I, alpha 1stage91HUGeneFLM57731_s_at168Hs.75765chemokine (C—X—C motif) ligand 2stage92HUGeneFLM62840_at168Hs.82542acyloxyacyl hydrolase (neutrophil)stage93HUGeneFLM63262_at168stage94HUGeneFLM68840_at168Hs.183109monoamine oxidase Astage95HUGeneFLM69203_s_at168Hs.75703chemokine (C—C motif) ligand 4stage96HUGeneFLM72885_ma1_s_at168stage97HUGeneFLM77349_at168Hs.421496transforming growth factor, beta-induced,stage68 kDa98HUGeneFLM82882_at168Hs.124030E74-like factor 1 (ets domain transcriptionstagefactor)99HUGeneFLM83822_at168Hs.209846LPS-responsive vesicle trafficking, beach andstageanchor containing100HUGeneFLM92934_at168Hs.410037connective tissue growth factorstage101HUGeneFLM95178_at168Hs.119000actinin, alpha 1stage102HUGeneFLS69115_at168Hs.10306natural killer cell group 7 sequencestage103HUGeneFLS77393_at168Hs.145754kruppel-like factor 3 (basic)stage104HUGeneFLS78187_at168Hs.153752cell division cycle 25Bstage105HUGeneFLU01833_at168Hs.81469nucleotide binding protein 1 (MinD homolog,stageE. coli)106HUGeneFLU07231_at168Hs.309763G-rich RNA sequence binding factor 1stage107HUGeneFLU09278_at168Hs.436852fibroblast activation protein, alphastage108HUGeneFLU09937_ma1_s_at168stage109HUGeneFLU10550_at168Hs.79022GTP binding protein overexpressed in skeletalstagemuscle110HUGeneFLU12424_s_at168Hs.108646glycerol-3-phosphate dehydrogenase 2 (mitochondrial)stage111HUGeneFLU16306_at168Hs.434488chondroitin sulfate proteoglycan 2 (versican)stage112HUGeneFLU20158_at168Hs.2488lymphocyte cytosolic protein 2 (SH2 domainstagecontaining leukocyte protein of 76 kDa)113HUGeneFLU20536_s_at168Hs.3280caspase 6, apoptosis-related cysteine proteasestage114HUGeneFLU24266_at168Hs.77448aldehyde dehydrogenase 4 family, memberstageA1115HUGeneFLU28249_at168Hs.301350FXYD domain containing ion transport regulator 3stage116HUGeneFLU28488_s_at168Hs.155935complement component 3a receptor 1stage117HUGeneFLU29680_at168Hs.227817BCL2-related protein A1stage118HUGeneFLU37143_at168Hs.152096cytochrome P450, family 2, subfamily J, polypeptide 2stage119HUGeneFLU38864_at168Hs.108139zinc finger protein 212stage120HUGeneFLU39840_at168Hs.163484forkhead box A1stage121HUGeneFLU41315_ma1_s_at168stage122HUGeneFLU44111_at168Hs.42151histamine N-methyltransferasestage123HUGeneFLU47414_at168Hs.13291cyclin G2stage124HUGeneFLU49352_at168Hs.4147542,4-dienoyl CoA reductase 1, mitochondrialstage125HUGeneFLU50708_at168Hs.1265branched chain keto acid dehydrogenase E1,stagebeta polypeptide (maple syrup urine disease)126HUGeneFLU52101_at168Hs.9999epithelial membrane protein 3stage127HUGeneFLU59914_at168Hs.153863MAD, mothers against decapentaplegic homologstage6 (Drosophila)128HUGeneFLU60205_at168Hs.393239sterol-C4-methyl oxidase-likestage129HUGeneFLU61981_at168Hs.42674mutS homolog 3 (E. coli)stage130HUGeneFLU64520_at168Hs.66708vesicle-associated membrane protein 3 (callubrevin)stage131HUGeneFLU65093_at168Hs.82071Cbp/p300-interacting transactivator, withstageGlu/Asp-rich carboxy-terminal domain, 2132HUGeneFLU68619_at168Hs.444445SWI/SNF related, matrix associated, actinstagedependent regulator of chromatin, subfamilyd, member 3133HUGeneFLU68019_at168Hs.288261MAD, mothers against decapentaplegic homologstage3 (Drosophila)134HUGeneFLU68385_at168Hs.380923likely ortholog of mouse myeloid ecotropicstageviral integration site-related gene 2135HUGeneFLU68485_at168Hs.193163bridging integrator 1stage136HUGeneFLU74324_at168Hs.90875RAB interacting factorstage137HUGeneFLU77970_at168Hs.321164neuronal PAS domain protein 2stage138HUGeneFLU83303_cds2_at168Hs.164021chemokine (C—X—C motif) ligand 6 (granulocytestagechemotactic protein 2)139HUGeneFLU88871_at168Hs.79993peroxisomal biogenesis factor 7stage140HUGeneFLU90549_at168Hs.236774high mobility group nucleosomal bindingstagedomain 4141HUGeneFLU90716_at168Hs.79187coxsackie virus and adenovirus receptorstage142HUGeneFLV00594_at168Hs.118786metallothionein 2Astage143HUGeneFLV00594_s_at168Hs.118786metallothionein 2Astage144HUGeneFLX02761_s_at168Hs.418138flbronectin 1stage145HUGeneFLX04011_at168Hs.88974cytochrome b-245, beta polypeptide (chronicstagegranulomatous disease)146HUGeneFLX04085_ma1_at168stage147HUGeneFLX07438_s_at168stage148HUGeneFLX07743_at168Hs.77436pleckstrinstage149HUGeneFLX13334_at168Hs.75627CD14 antigenstage150HUGeneFLX14046_at168Hs.153053CD37 antigenstage151HUGeneFLX14813_at168Hs.166160acetyl-Coenzyme A acyltransferase 1 (perox-stageisomal 3-oxoacyl-Coenzyme A thiolase)152HUGeneFLX15880_at168Hs.415997collagen, type VI, alpha 1stage153HUGeneFLX15882_at168Hs.420269collagen, type VI, alpha 2stage154HUGeneFLX51408_at168Hs.380138chimerin (chimaerin) 1stage155HUGeneFLX53800_s_at168Hs.89690chemokine (C—X—C motif) ligand 3stage156HUGeneFLX54489_ma1_at168stage157HUGeneFLX57351_s_at168Hs.174195interferon induced transmembrane protein 2stage(1-8D)158HUGeneFLX57579_s_at168stage159HUGeneFLX58072_at168Hs.169946GATA binding protein 3stage160HUGeneFLX62048_at168Hs.249441WEE1 homolog (S. pombe)stage161HUGeneFLX64072_s_at168Hs.375957integrin, beta 2 (antigen CD18 (p95), lymphocytestagefunction-associated antigen 1; macrophageantigen 1 (mac-1) beta subunit)162HUGeneFLX65614_at168Hs.2962S100 calcium binding protein Pstage163HUGeneFLX66945_at168Hs.748fibroblast growth factor receptor 1 (fms-relatedstagetyrosine kinase 2, Pfeiffer syndrome)164HUGeneFLX67491_f_at168Hs.355697glutamate dehydrogenase 1stage165HUGeneFLX68194_at168Hs.80919synaptophysin-like proteinstage166HUGeneFLX73882_at168Hs.254605microtubule-associated protein 7stage167HUGeneFLX78520_at168Hs.372528chloride channel 3stage168HUGeneFLX78549_at168Hs.51133PTK6 protein tyrosine kinase 6stage169HUGeneFLX78565_at168Hs.98998tenascin C (hexabrachion)stage170HUGeneFLX78669_at168Hs.79088reticulocalbin 2, EF-hand calcium bindingstagedomain171HUGeneFLX83618_at168Hs.598893-hydroxy-3-methylglutaryl-Coenzyme Astagesynthase 2 (mitochondrial)172HUGeneFLX84908_at168Hs.78060phosphorylase kinase, betastage173HUGeneFLX90908_at168Hs.147391fatty acid binding protein 6, ileal (gastrotropin)stage174HUGeneFLX91504_at168Hs.389277ADP-ribosylation factor related protein 1stage175HUGeneFLX95632_s_at168Hs.387906abl-interactor 2stage176HUGeneFLX97267_ma1_s_at168stage177HUGeneFLY00705_at168Hs.407856serine protease inhibitor, Kazal type 1stage178HUGeneFLY00787_s_at168Hs.624interleukin 8stage179HUGeneFLY00815_at168Hs.75216protein tyrosine phosphatase, receptor type, Fstage180HUGeneFLY08374_rna1_at168stage181HUGeneFLZ12173_at168Hs.334534glucosamine (N-acetyl)-6-sulfatase (Sanfilippostagedisease IIID)182HUGeneFLZ19554_s_at168Hs.435800vimentinstage183HUGeneFLZ26491_s_at168Hs.240013catechol-O-methyltransferasestage184HUGeneFLZ29331_at168Hs.372758ubiquitin-conjugating enzyme E2H (UBC8stagehomolog, yeast)185HUGeneFLZ35491_at168Hs.377484BCL2-associated athanogenestage186HUGeneFLZ48199_at168Hs.82109syndecan 1stage187HUGeneFLZ48605_at168Hs.421825inorganic pyrophosphatase 2stage188HUGeneFLZ74615_at168Hs.172928collagen, type I, alpha 1stage189HUGeneFLD87437_at168Hs.43660chromosome 1 open reading frame 16recurrence190HUGeneFLL49169_at168Hs.75678FBJ murine osteosarcoma viral oncogenerecurrencehomolog B191HUGeneFLAF006041_at168Hs.336916death-associated protein 6recurrence192HUGeneFLD83780_at168Hs.437991KIAA0196 gene productrecurrence193HUGeneFLD64154_at168Hs.90107adhesion regulating molecule 1recurrence194HUGeneFLD21337_at168Hs.408collagen, type IV, alpha 6recurrence195HUGeneFLM16938_s_at168Hs.820homeo box C6recurrence196HUGeneFLD87258_at168Hs.75111protease, serine, 11 (IGF binding)recurrence197HUGeneFLU58516_at168Hs.3745milk fat globule-EGF factor 8 proteinrecurrence198HUGeneFLU45973_at168Hs.178347skeletal muscle and kidney enriched inositolrecurrencephosphatase199HUGeneFLU62015_at168Hs.8867cysteine-rich, angiogenic inducer, 61recurrence200HUGeneFLU94855_at168Hs.381255eukaryotic translation initiation factor 3, subunitrecurrence5 epsilon, 47 kDa201HUGeneFLL34155_at168Hs.83450laminin, alpha 3recurrence202HUGeneFLU70439_s_at168Hs.84264acidic (leucine-rich) nuclear phosphoproteinrecurrence32 family, member B203HUGeneFLU66702_at168Hs.74624protein tyrosine phosphatase, receptor type, Nrecurrencepolypeptide 2204HUGeneFLHG511-HT511_at168recurrence205HUGeneFLHG3076-HT3238_s_at168recurrence206HUGeneFLM98528_at168Hs.79404DNA segment on chromosome 4 (unique) 234recurrenceexpressed sequence207HUGeneFLM63175_at168Hs.295137autocrine motility factor receptorrecurrence208HUGeneFLD49387_at168Hs.294584leukotriene B4 12-hydroxydehydrogenaserecurrence209HUGeneFLHG1879-HT1919_at168recurrence210HUGeneFLZ23064_at168Hs.380118RNA binding motif protein, X chromosomerecurrence211HUGeneFLX63469_at168Hs.77100general transcription factor IIE, polypeptide 2,recurrencebeta 34 kDa212HUGeneFLL38928_at168Hs.1181315,10-methenyltetrahydrofolate synthetase (5-recurrenceformyltetrahydrofolate cyclo-ligase)213HUGeneFLU21858_at168Hs.60679TAF9 RNA polymerase II, TATA box bindingrecurrenceprotein (TBP)-associated factor, 32 kDa214HUGeneFLM64572_at168Hs.405666protein tyrosine phosphatase, non-receptorrecurrencetype 3215HUGeneFLD83657_at168Hs.19413S100 calcium binding protein A12 (calgranulinSCCC)216HUGeneFLHG3945-HT4215_at168SCC217HUGeneFLJ00124_at168SCC218HUGeneFLL05187_at168SCC219HUGeneFLL42583_f_at168Hs.367762keratin 6ASCC220HUGeneFLL42601_f_at168Hs.367762keratin 6ASCC221HUGeneFLL42611_f_at168Hs.446417keratin 6ESCC222HUGeneFLM19888_at168Hs.1076small proline-rich protein 1B (comifin)SCC223HUGeneFLM20030_f_at168Hs.505352Human small proline rich protein (spril)SCCmRNA, clone 930.224HUGeneFLM21005_at168SCC225HUGeneFLM21302_at168Hs.505327Human small proline rich protein (spril)SCCmRNA, clone 174N.226HUGeneFLM21539_at168Hs.2421small proline-rich protein 2CSCC227HUGeneFLM86757_s_at168Hs.112408S100 calcium binding protein A7 (psoriasin 1)SCC228HUGeneFLS72493_s_at168Hs.432448keratin 16 (focal non-epidermolytic palmoplantarSCCkeratoderma)229HUGeneFLU70981_at168Hs.336046interleukin 13 receptor, alpha 2SCC230HUGeneFLV01516_f_at168Hs.367762keratin 6ASCC231HUGeneFLX53065_f_at168SCC232HUGeneFLX57766_at168Hs.143751matrix metalloproteinase 11 (stromelysin 3)SCC233EOS Hu03400773133NM_003105*: Homo sapiens sortilin-relatedprogressionreceptor, L(DLR class) A repeats-containing(SORL1), mRNA.234EOS Hu03400843133NM_003105*: Homo sapiens sortilin-relatedprogressionreceptor, L(DLR class) A repeats-containing(SORL1), mRNA.235EOS Hu03400844133NM_003105*: Homo sapiens sortilin-relatedprogressionreceptor, L(DLR class) A repeats-containing(SORL1), mRNA.236EOS Hu03400846133sortilin-related receptor, L(DLR class) A repeats-progressioncontaining (SORL1)237EOS Hu03402328133Target Exonprogression238EOS Hu03402384133NM_007181*: Homo sapiens mitogen-progressionactivated protein kinase kinase kinase kinase1 (MAP4K1), mRNA.239EOS Hu03404208133C6001282: gi|4504223|ref|NP_000172.1|progressionglucuronidase, beta [Homo sapiens]gi|114963|sp|P082240EOS Hu03404606133Target Exonprogression241EOS Hu03404826133Target Exonprogression242EOS Hu03404875133NM_022819*: Homo sapiens phospholipaseprogressionA2, group IIF (PLA2G2F), mRNA. VERSIONNM_020245.2 GI243EOS Hu03404913133NM_024408*: Homo sapiens Notch (Drosophilaprogressionhomolog 2 (NOTCH2), mRNA. VERSIONNM_024410.1 GI244EOS Hu03404977133Insulin-like growth factor 2 (somatomedin A)progression(IGF2)245EOS Hu03405036133NM_021628*: Homo sapiens arachidonateprogressionlipoxygenase 3 (ALOXE3), mRNA. VERSIONNM_020229.1 GI246EOS Hu03405371133NM_005569*: Homo sapiens LIM domainprogressionkinase 2 (LIMK2), transcript variant 2a,mRNA.247EOS Hu03405667133Target Exonprogression248EOS Hu03406002133Target Exonprogression249EOS Hu03407955133Hs.9343ESTsprogression250EOS Hu03408049133Hs.345588desmoplakin (DPI, DPII)progression251EOS Hu03408288133Hs.16886gb: zI73d06.r1 Stratagene colon (937204)progressionHomo sapiens cDNA clone 5′, mRNA sequence252EOS Hu03409513133Hs.54642methionine adenosyltransferase II, betaprogression253EOS Hu03409556133Hs.54941phosphorylase kinase, alpha 2 (liver)progression254EOS Hu03409586133Hs.55044DKFZP586H2123 proteinprogression255EOS Hu03409632133Hs.55279serine (or cysteine) proteinase inhibitor, cladeprogressionB (ovalbumin), member 5256EOS Hu03410047133Hs.379753zinc finger protein 36 (KOX 18)progression257EOS Hu03411817133Hs.72241mitogen-activated protein kinase kinase 2progression258EOS Hu03412649133Hs.74369integrin, alpha 7progression259EOS Hu03412841133Hs.101395hypothetical protein MGC11352progression260EOS Hu03413564133gb: 601146990F1 NIH_MGC_19 Homoprogressionsapiens cDNA clone 5′, mRNA sequence261EOS Hu03413786133Hs.13500ESTsprogression262EOS Hu03413840133Hs.356228RNA binding motif protein, X chromosomeprogression263EOS Hu03413929133Hs.75617collagen, type IV, alpha 2progression264EOS Hu03414223133Hs.238246hypothetical protein FLJ22479progression265EOS Hu03414732133Hs.77152minichromosome maintenance deficient (S. cerevisiae)progression7266EOS Hu03414762133Hs.77257KIAA0068 proteinprogression267EOS Hu03414840133Hs.23823hairy/enhancer-of-split related with YRPWprogressionmotif-like268EOS Hu03414843133Hs.77492heterogeneous nuclear ribonucleoprotein A0progression269EOS Hu03414895133Hs.116278Homo sapiens cDNA FLJ13571 fis, cloneprogressionPLACE1008405270EOS Hu03414907133Hs.77597polo (Drosophila)-like kinaseprogression271EOS Hu03414918133Hs.72222hypothetical protein FLJ13459progression272EOS Hu03415200133Hs.78202SWI/SNF related, matrix associated, actinprogressiondependent regulator of chromatin, subfamilya, member 4273EOS Hu03416640133Hs.79404neuron-specific proteinprogression274EOS Hu03416815133Hs.80120UDP-N-acetyl-alpha-D-progressiongalactosamine: polypeptide N-acetylgalactosaminyltransferase 1 (GaINAc-T1)275EOS Hu03416977133Hs.406103hypothetical protein FKSG44progression276EOS Hu03417615133Hs.82314hypoxanthine phosphoribosyltransferase 1progression(Lesch-Nyhan syndrome)277EOS Hu03417839133Hs.82712fragile X mental retardation, autosomal homolog 1progression278EOS Hu03417900133Hs.82906CDC20 (cell division cycle 20, S. cerevisiae,progressionhomolog)279EOS Hu03417924133Hs.82932cyclin D1 (PRAD1: parathyroid adenomatosisprogression1)280EOS Hu03418127133Hs.83532membrane cofactor protein (CD46, trophoblast-progressionlymphocyte cross-reactive antigen)281EOS Hu03418321133Hs.84087KIAA0143 proteinprogression282EOS Hu03418504133Hs.85335Homo sapiens mRNA; cDNAprogressionDKFZp564D1462 (from cloneDKFZp564D1462)283EOS Hu03418629133Hs.86859growth factor receptor-bound protein 7progression284EOS Hu03419602133Hs.91521hypothetical proteinprogression285EOS Hu03419847133Hs.184544Homo sapiens, clone IMAGE: 3355383,progressionmRNA, partial cds286EOS Hu03420079133Hs.94896PTD011 proteinprogression287EOS Hu03420116133Hs.95231FH1/FH2 domain-containing proteinprogression288EOS Hu03420307133Hs.66219ESTsprogression289EOS Hu03420613133Hs.406637ESTs, Weakly similar to A47582 B-cell growthprogressionfactor precursor [H. sapiens]290EOS Hu03420732133Hs.367762ESTsprogression291EOS Hu03421026133Hs.101067GCN5 (general control of amino-acid synthesis,progressionyeast, homolog)-like 2292EOS Hu03421075133Hs.101474KIAA0807 proteinprogression293EOS Hu03421101133Hs.101840major histocompatibility complex, class I-likeprogressionsequence294EOS Hu03421186133Hs.270563ESTs, Moderately similar to T12512 hypotheticalprogressionprotein DKFZp434G232.1 [H. sapiens]295EOS Hu03421311133Hs.283609hypothetical protein PRO2032progression296EOS Hu03421475133Hs.104640HIV-1 inducer of short transcripts bindingprogressionprotein; lymphoma related factor297EOS Hu03421505133Hs.285641KIAA1111 proteinprogression298EOS Hu03421595133Hs.301685KIAA0620 proteinprogression299EOS Hu03421628133Hs.106210hypothetical protein FLJ10813progression300EOS Hu03421649133Hs.106415peroxisome proliferative activated receptor,progressiondelta301EOS Hu03421733133Hs.1420fibroblast growth factor receptor 3 (achondro-progressionplasia, thanatophoric dwarfism)302EOS Hu03421782133Hs.108258actin binding protein; macrophin (microfilamentprogressionand actin filament cross-linker protein)303EOS Hu03421989133Hs.110457Wolf-Hirschhorn syndrome candidate 1progression304EOS Hu03422043133Hs.110953retinoic acid induced 1progression305EOS Hu03422068133Hs.104520Homo sapiens cDNA FLJ13694 fis, cloneprogressionPLACE2000115306EOS Hu03422506133Hs.300741sorcinprogression307EOS Hu03422913133Hs.121599CGI-18 proteinprogression308EOS Hu03422929133Hs.94011ESTs, Weakly similar to MGB4_HUMANprogressionMELANOMA-ASSOCIATED ANTIGEN B4[H. sapiens]309EOS Hu03422959133Hs.349256paired immunoglobulin-like receptor betaprogression310EOS Hu03423138133gb: EST385571 MAGE resequences, MAGMprogressionHomo sapiens cDNA, mRNA sequence311EOS Hu03423185133Hs.380062ornithine decarboxylase antizyme 1progression312EOS Hu03423599133Hs.31731peroxiredoxin 5progression313EOS Hu03423810133Hs.132955BCL2/adenovirus E1B 19 kD-Interacting proteinprogression3-like314EOS Hu03423960133Hs.136309SH3-containing protein SH3GLB1progression315EOS Hu03424244133Hs.143601hypothetical protein hCLA-isoprogression316EOS Hu03424415133Hs.146580enolase 2, (gamma, neuronal)progression317EOS Hu03424909133Hs.153752cell division cycle 25Bprogression318EOS Hu03424959133Hs.153937activated p21cdc42Hs kinaseprogression319EOS Hu03425093133Hs.154525KIAA1076 proteinprogression320EOS Hu03425097133Hs.154545PDZ domain containing guanine nucleotideprogressionexchange factor(GEF)1321EOS Hu03425205133Hs.155106receptor (calcitonin) activity modifying protein 2progression322EOS Hu03425221133Hs.155188TATA box binding protein (TBP)-associatedprogressionfactor, RNA polymerase II, F, 55 kD323EOS Hu03425243133Hs.155291KIAA0005 gene productprogression324EOS Hu03425380133Hs.32148AD-015 proteinprogression325EOS Hu03426028133Hs.172028a disintegrin and metalloproteinase domain 10progression(ADAM10)326EOS Hu03426125133Hs.166994FAT tumor suppressor (Drosophila) homologprogression327EOS Hu03426177133Hs.167700Homo sapiens cDNA FLJ10174 fis, cloneprogressionHEMBA1003959328EOS Hu03426252133Hs.28917ESTsprogression329EOS Hu03426468133Hs.117558ESTsprogression330EOS Hu03426469133Hs.363039methylmalonate-semialdehyde dehydrogenaseprogression331EOS Hu03426508133Hs.170171glutamate-ammonia ligase (glutamine synthase)progression332EOS Hu03426682133Hs.2056UDP glycosyltransferase 1 family, polypeptideprogressionA9333EOS Hu03426799133Hs.303154popeye protein 3progression334EOS Hu03426982133Hs.173091ubiquitin-like 3progression335EOS Hu03427239133Hs.356512ubiquitin carrier proteinprogression336EOS Hu03427351133Hs.123253hypothetical protein FLJ22009progression337EOS Hu03427681133Hs.284232tumor necrosis factor receptor superfamily,progressionmember 12 (translocating chain-associationmembrane protein)338EOS Hu03427722133Hs.180479hypothetical protein FLJ20116progression339EOS Hu03427747133Hs.180655serine/threonine kinase 12progression340EOS Hu03427999133Hs.181369ubiquitin fusion degradation 1-likeprogression341EOS Hu03428115133Hs.300855KIAA0977 proteinprogression342EOS Hu03428284133Hs.183435NM_004545: Homo sapiens NADH dehydrogenaseprogression(ubiquinone) 1 beta subcomplex, 1(7 kD, MNLL) (NDUFB1), mRNA.343EOS Hu03428318133Hs.356190ubiquitin Bprogression344EOS Hu03428712133Hs.190452KIAA0365 gene productprogression345EOS Hu03428901133Hs.146668KIAA1253 proteinprogression346EOS Hu03429124133Hs.196914minor histocompatibility antigen HA-1progression347EOS Hu03429187133Hs.163872ESTs, Weakly similar to S65657 alpha-1C-progressionadrenergic receptor splice form 2 [H. sapiens]348EOS Hu03429311133Hs.198998conserved helix-loop-helix ubiquitous kinaseprogression349EOS Hu03429561133Hs.250646baculoviral IAP repeat-containing 6progression350EOS Hu03429802133Hs.5367ESTs, Weakly similar to I38022 hypotheticalprogressionprotein [H. sapiens]351EOS Hu03429953133Hs.226581COX15 (yeast) homolog, cytochrome c oxidaseprogressionassembly protein352EOS Hu03430604133Hs.247309succinate-CoA ligase, GDP-forming, betaprogressionsubunit353EOS Hu03430677133Hs.359784desmoglein 2progression354EOS Hu03430746133Hs.406256ESTsprogression355EOS Hu03431604133Hs.264190vacuolar protein sorting 35 (yeast homolog)progression356EOS Hu03431842133Hs.271473epithelial protein up-regulated in carcinoma,progressionmembrane associated protein 17357EOS Hu03431857133Hs.271742ADP-ribosyltransferase (NAD; poly (ADP-progressionribose) polymerase)-like 3358EOS Hu03432258133Hs.293039ESTsprogression359EOS Hu03432327133Hs.274363neuroglobinprogression360EOS Hu03432554133Hs.278411NCK-associated protein 1progression361EOS Hu03432864133Hs.359682calpastatinprogression362EOS Hu03433052133Hs.293003ESTs, Weakly similar to PC4259 ferritin associatedprogressionprotein [H. sapiens]363EOS Hu03433282133Hs.49007hypothetical proteinprogression364EOS Hu03433844133Hs.179647Homo sapiens cDNA FLJ12195 fis, cloneprogressionMAMMA1000865365EOS Hu03433914133Hs.112160Homo sapiens DNA helicase homolog (PIF1)progressionmRNA, partial cds366EOS Hu03434055133Hs.3726x 003 proteinprogression367EOS Hu03434263133Hs.79187ESTsprogression368EOS Hu03434547133Hs.106124ESTsprogression369EOS Hu03434831133Hs.273397KIAA0710 gene productprogression370EOS Hu03434978133Hs.4310eukaryotic translation initiation factor 1Aprogression371EOS Hu03435158133Hs.65588DAZ associated protein 1progression372EOS Hu03435320133Hs.117864ESTsprogression373EOS Hu03435521133Hs.6361mitogen-activated protein kinase kinase 1progressioninteracting protein 1374EOS Hu03436472133Hs.46366KIAA0948 proteinprogression375EOS Hu03436576133Hs.77542ESTsprogression376EOS Hu03437223133Hs.330716Homo sapiens cDNA FLJ14368 fis, cloneprogressionHEMBA1001122377EOS Hu03437256133Hs.97871Homo sapiens, clone IMAGE: 3845253,progressionmRNA, partial cds378EOS Hu03437524133Hs.385719ESTsprogression379EOS Hu03438013133Hs.15670ESTsprogression380EOS Hu03438644133Hs.129037ESTsprogression381EOS Hu03438818133Hs.30738ESTsprogression382EOS Hu03438942133Hs.6451PRO0659 proteinprogression383EOS Hu03439010133Hs.75216Homo sapiens cDNA FLJ13713 fis, cloneprogressionPLACE2000398, moderately similar to LARPROTEIN PRECURSOR (LEUKOCYTEANTIGEN RELATED) (EC 3.1.3.48)384EOS Hu03439130133Hs.375195ESTsprogression385EOS Hu03439578133Hs.350547nuclear receptor co-repressor/HDAC3 complexprogressionsubunit386EOS Hu03439632133Hs.334437hypothetical protein MGC4248progression387EOS Hu03440014133Hs.6856ash2 (absent, small, or homeotic, Drosophila,progressionhomolog)-like388EOS Hu03440100133Hs.158549ESTs, Weakly similar to T2D3_HUMANprogressionTRANSCRIPTION INITIATION FACTORTFIID 135 KDA SUBUNIT [H. sapiens]389EOS Hu03440197133Hs.317714pallid (mouse) homolog, pallidinprogression390EOS Hu03440357133Hs.20950phospholysine phosphohistidine inorganicprogressionpyrophosphate phosphatase391EOS Hu03441650133Hs.132545ESTsprogression392EOS Hu03442220133Hs.8148selenoprotein Tprogression393EOS Hu03442549133Hs.8375TNF receptor-associated factor 4progression394EOS Hu03443407133Hs.348514ESTs, Moderately similar to 2109260A B cellprogressiongrowth factor [H. sapiens]395EOS Hu03443471133Hs.398102Homo sapiens clone FLB3442 PRO0872progressionmRNA, complete cds396EOS Hu03443679133Hs.9670hypothetical protein FLJ10948progression397EOS Hu03443893133Hs.115472ESTs, Weakly similar to 2004399A chromosomalprogressionprotein [H. sapiens]398EOS Hu03444037133Hs.380932CHMP1.5 proteinprogression399EOS Hu03444312133Hs.351142ESTsprogression400EOS Hu03444336133Hs.10882HMG-box containing protein 1progression401EOS Hu03444604133Hs.11441chromosome 1 open reading frame 8progression402EOS Hu03445084133Hs.250848hypothetical protein FLJ14761progression403EOS Hu03445462133Hs.288649hypothetical protein MGC3077progression404EOS Hu03445692133Hs.182099ESTsprogression405EOS Hu03445831133Hs.13351LanC (bacterial lantibiotic synthetase componentprogressionC)-like 1406EOS Hu03446556133Hs.15303KIAA0349 proteinprogression407EOS Hu03446847133Hs.82845Homo sapiens cDNA: FLJ21930 fis, cloneprogressionHEP04301, highly similar to HSU90916 Humanclone 23815 mRNA sequence408EOS Hu03447343133Hs.236894ESTs, Highly similar to S02392 alpha-2-progressionmacroglobulin receptor precursor [H. sapiens]409EOS Hu03447400133Hs.18457hypothetical protein FLJ20315progression410EOS Hu03448357133Hs.108923RAB38, member RAS oncogene familyprogression411EOS Hu03448524133Hs.21356hypothetical protein DKFZp762K2015progression412EOS Hu03448625133Hs.178470hypothetical protein FLJ22662progression413EOS Hu03448780133Hs.267749Human DNA sequence from clone 366N23 onprogressionchromosome 6q27. Contains two genes similarto consecutive parts of the C. elegansUNC-93 (protein 1, C46F11.1) gene, aKIAA0173 and Tubulin-Tyrosine Ligase LIKEgene, a Mitotic Feedback Control ProteinMADP2 H414EOS Hu03448813133Hs.22142cytochrome b5 reductase b5R.2progression415EOS Hu03449268133Hs.23412hypothetical protein FLJ20160progression416EOS Hu03449626133Hs.112860zinc finger protein 258progression417EOS Hu03450693133Hs.25625hypothetical protein FLJ11323progression418EOS Hu03450997133Hs.35254hypothetical protein FLB6421progression419EOS Hu03451164133Hs.60659ESTs, Weakly similar to T46471 hypotheticalprogressionprotein DKFZp434L0130.1 [H. sapiens]420EOS Hu03451225133Hs.57655ESTsprogression421EOS Hu03451867133Hs.27192hypothetical protein dJ1057B20.2progression422EOS Hu03451970133Hs.211046ESTsprogression423EOS Hu03452012133Hs.279766kinesin family member 4Aprogression424EOS Hu03452170133Hs.28285patched related protein translocated in renalpragressioncancer425EOS Hu03452517133gb: RC-BT068-130399-068 BT068 Homoprogressionsapiens cDNA, mRNA sequence426EOS Hu03452829133Hs.63368ESTs, Weakly similar to TRHY_HUMANprogressionTRICHOHYALI [H. sapiens]427EOS Hu03452929133Hs.172816neuregulin 1progression428EOS Hu03453395133Hs.377915mannosidase, alpha, class 2A, member 1progression429EOS Hu03454639133gb: RC2-ST0158-091099-011-d05 ST0158progressionHomo sapiens cDNA, mRNA sequence430EOS Hu03456332133Hs.399939gb: nc39d05.r1 NCI_CGAP_Pr2 Homo sapiensprogressioncDNA clone, mRNA sequence431EOS Hu03457228133Hs.195471Human cosmid CRI-JC2015 at D10S289 inprogression10sp13432EOS Hu03458132133Hs.103267hypothetical protein FLJ22548 similar to geneprogressiontrap PAT 12433EOS Hu03408688133Hs.152925KIAA1268 proteinprogression434EOS Hu03410691133Hs.65450reticulon 4progression435EOS Hu03420269133Hs.96264alpha thalassemia/mental retardation syndromeprogressionX-linked (RAD54 (S. cerevisiae) homolog)436EOS Hu03422119133Hs.111862KIAA0590 gene productprogression437EOS Hu03422765133Hs.1578baculoviral IAP repeat-containing 5 (survivin)progression438EOS Hu03422984133Hs.351597ESTsprogression439EOS Hu03428016133Hs.181461ariadne homolog, ubiquitin-conjugating enzymeprogressionE2 binding protein, 1 (Drosophila)440EOS Hu03437325133Hs.5548F-box and leucine-rich repeat protein 5progression441EOS Hu03444773133Hs.11923hypothetical protein DJ167A19.1progression442EOS Hu03445926133Hs.334826splicing factor 3b, subunit 1, 155 kDaprogression443EOS Hu03452714133Hs.30340KIAA1165: likely ortholog of mouse Nedd4progressionWW domain-binding protein 5A444EOS Hu03452866133Hs.268016ESTsprogression445EOS Hu03453963133Hs.28959cDNA FLJ36513 fis, clone TRACH2001523progression446EOS Hu03457329133Hs.359682calpastatinprogression447U133A200600_at168Hs.170328NM_001910; cathepsin E isoform a preproproteinCISNM_148964; cathepsin E isoform b preproprotein448U133A200762_at168Hs.173381NM_019894; transmembrane protease, serineCIS4 isoform 1 NM_183247; transmembraneprotease, serine 4 isoform 2449U133A201088_at168Hs.159557NM_000228; laminin subunit beta 3 precursorCIS450U133A201291_s_at168Hs.156346NM_030570; uroplakin 3B isoform aCISNM_182683; uroplakin 3B isoform cNM_182684; uroplakin 3B isoform b451U133A201560_at168Hs.25035NM_005547; involucrinCIS452U133A201616_s_at168Hs.443811NM_004692; NM_032727; intemexin neuronalCISintermediate filament protein, alpha453U133A201641_at168Hs.118110NM_016233; peptidylarginine deiminase typeCISIII454U133A201744_s_at168Hs.406475NM_014417; BCL2 binding component 3CIS455U133A201842_s_at168Hs.76224NM_020142; NADH: ubiquinone oxidoreductaseCISMLRQ subunit homolog456U133A201858_s_at168Hs.1908NM_018058; cartilage acidic protein 1CIS457U133A201859_at168Hs.1908NM_000497; cytochrome P450, subfamily XIBCIS(steroid 11-beta-hydroxylase), polypeptide 1precursor458U133A202746_at168Hs.17109NM_007193; annexin A10CIS459U133A202917_s_at168Hs.416073NM_001958; eukaryotic translation elongationCISfactor 1 alpha 2460U133A203009_at168Hs.155048NM_005581; Lutheran blood group (AubergerCISb antigen included)461U133A203287_at168Hs.18141NM_005581; Lutheran blood group (AubergerCISb antigen included)462U133A203477_at168Hs.409034NM_030570; uroplakin 3B isoform aCISNM_182683; uroplakin 3B isoform cNM_182684; uroplakin 3B isoform b463U133A203649_s_at168Hs.76422NM_000300; phospholipase A2, group IIACIS(platelets, synovial fluid)464U133A203759_at168Hs.75268NM_007193; annexin A10CIS465U133A203792_x_at168Hs.371617NM_007144; ring finger protein 110CIS466U133A203842_s_at168Hs.172740NM_014417; BCL2 binding component 3CIS467U133A203980_at168Hs.391561NM_001442; fatty acid binding protein 4,CISadipocyte468U133A204141_at168Hs.300701NM_017689; hypothetical protein FLJ20151CIS469U133A204380_s_at168Hs.1420NM_007144; ring finger protein 110CIS470U133A204465_s_at168Hs.76888NM_004692; NM_032727; intemexin neuronalCISintermediate filament protein, alpha471U133A204487_s_at168Hs.367809NM_001248; ectonucleoside triphosphateCISdiphosphohydrolase 3472U133A204508_s_at168Hs.279916NM_017689; hypothetical protein FLJ20151CIS473U133A204540_at168Hs.433839NM_001958; eukaryotic translation elongationCISfactor 1 alpha 2474U133A204688_at168Hs.409798NM_016233; peptidylarginine deiminase typeCISIII475U133A204952_at168Hs.377028NM_000445; plectin 1, intermediate filamentCISbinding protein 500 kDa476U133A204990_s_at168Hs.85266NM_000213; integrin, beta 4CIS477U133A205073_at168Hs.152096NM_019894; transmembrane protease, serineCIS4 isoform 1 NM_183247; transmembraneprotease, serine 4 isoform 2478U133A205382_s_at168Hs.155597NM_000213; integrin, beta 4CIS479U133A205453_at168Hs.290432NM_002145; homeo box B2CIS480U133A205455_at168Hs.2942NM_006760; uroplakin 2CIS481U133A205927_s_at168Hs.1355NM_001910; cathepsin E isoform a preproproteinCISNM_148964; cathepsin E isoform b preproprotein482U133A206122_at168Hs.95582NM_006942; SRY-box 15CIS483U133A206191_at168Hs.47042NM_001248; ectonucleoside triphosphateCISdiphosphohydrolase 3484U133A206392_s_at168Hs.82547NM_005522; homeobox A1 protein isoform aCISNM_153620; homeobox A1 protein isoform b485U133A206393_at168Hs.83760NM_003282; troponin I, skeletal, fastCIS486U133A206465_at168Hs.277543NM_015162; lipidosinCIS487U133A206561_s_at168Hs.116724NM_015162; lipidosinCIS488U133A206658_at168Hs.284211NM_030570; uroplakin 3B isoform aCISNM_182683; uroplakin 3B isoform cNM_182684; uroplakin 3B isoform b489U133A207173_x_at168Hs.443435NM_000213; integrin, beta 4CIS490U133A207862_at168Hs.379613NM_006760; uroplakin 2CIS491U133A209138_x_at168Hs.505407NM_015162; lipidosinCIS492U133A209270_at168Hs.436983NM_000228; laminin subunit beta 3 precursorCIS493U133A209340_at168Hs.21293NM_007144; ring finger protein 110CIS494U133A209591_s_at168Hs.170195NM_000228; laminin subunit beta 3 precursorCIS495U133A209732_at168Hs.85201NM_001248; ectonucleoside triphosphateCISdiphosphohydrolase 3496U133A210143_at168Hs.188401NM_007193; annexin A10CIS497U133A210735_s_at168Hs.5338NM_017689; hypothetical protein FLJ20151CIS498U133A210761_s_at168Hs.86859NM_020142; NADH: ubiquinone oxidoreductaseCISMLRQ subunit homolog499U133A211002_s_at168Hs.82237NM_001958; eukaryotic translation elongationCISfactor 1 alpha 2500U133A211161_s_at168NM_000300; phospholipase A2, group IIACIS(platelets, synovial fiuld)501U133A211430_s_at168Hs.413826NM_001910; cathepsin E isoform a preproproteinCISNM_148964; cathepsin E isoform b preproprotein502U133A211671_s_at168Hs.126608NM_007144; ring finger protein 110CIS503U133A211692_s_at168Hs.87246NM_014417; BCL2 binding component 3CIS504U133A211896_s_at168Hs.156316NM_005581; Lutheran blood group (AubergerCISb antigen included)505U133A212077_at168Hs.443811NM_003282; troponin I, skeletal, fastCIS506U133A212192_at168Hs.109438NM_020142; NADH: ubiquinone oxidoreductaseCISMLRQ subunit homolog507U133A212195_at168Hs.71968NM_000445; plectin 1, intermediate filamentCISbinding protein 500 kDa508U133A212386_at168Hs.359259NM_005547; involucrinCIS509U133A212667_at168Hs.111779NM_000299; plakophilin 1CIS510U133A212671_s_at168Hs.387679NM_002145; homeo box B2CIS511U133A212998_x_at168Hs.375115NM_000497; cytochrome P450, subfamily XIBCIS(steroid 11-beta-hydroxylase), polypeptide 1precursor512U133A213891_s_at168Hs.359289NM_007193; annexin A10CIS513U133A213975_s_at168Hs.234734NM_005522; homeobox A1 protein isoform aCISNM_153620; homeobox A1 protein isoform b514U133A214352_s_at168Hs.412107NM_006760; uroplakin 2CIS515U133A214599_at168Hs.157091NM_005547; involucrinCIS516U133A214630_at168Hs.184927NM_000497; cytochrome P450, subfamily XIBCIS(steroid 11-beta-hydroxylase), polypeptide 1precursor517U133A214639_s_at168Hs.67397NM_005522; homeobox A1 protein isoform aCISNM_153620; homeobox A1 protein isoform b518U133A214651_s_at168Hs.127428NM_002145; homeo box B2CIS519U133A214669_x_at168Hs.377975NM_001442; fatty acid binding protein 4,CISadipocyte520U133A214677_x_at168Hs.449601NM_006942; SRY-box 15CIS521U133A214752_x_at168Hs.195464NM_006942; SRY-box 15CIS522U133A215076_s_at168Hs.443625NM_016233; peptidylarginine deiminase typeCISIII523U133A215121_x_at168Hs.356861NM_018058; cartilage acidic protein 1CIS524U133A215176_x_at168Hs.503443NM_001248; ectonucleoside triphosphateCISdiphosphohydrolase 3525U133A215379_x_at168Hs.449601NM_006760; uroplakin 2CIS526U133A215812_s_at168Hs.499113NM_018058; cartilage acidic protein 1CIS527U133A216641_s_at168Hs.18141NM_005547; involucrinCIS528U133A216971_s_at168Hs.79706NM_000445; plectin 1, intermediate filamentCISbinding protein 500 kDa529U133A217028_at168Hs.421986NM_003282; troponin I, skeletal, fastCIS530U133A217040_x_at168Hs.95582NM_001910; cathepsin E isoform a preproproteinCISNM_148964; cathepsin E isoform b preproprotein531U133A217388_s_at168Hs.444471NM_000228; laminin subunit beta 3 precursorCIS532U133A217626_at168Hs.201967NM_000299; plakophilin 1CIS533U133A218484_at168Hs.221447NM_020142; NADH: ubiquinone oxidoreductaseCISMLRQ subunit homolog534U133A218656_s_at168Hs.93765NM_001442; fatty acid binding protein 4,CISadipocyte535U133A218718_at168Hs.43080NM_000445; plectin 1, intermediate filamentCISbinding protein 500 kDa536U133A218918_at168Hs.8910NM_000300; phospholipase A2, group IIACIS(platelets, synovial fluid)537U133A218960_at168Hs.414005NM_019894; transmembrane protease, serineCIS4 isoform 1 NM_183247; transmembraneprotease, serine 4 isoform 2538U133A219410_at168Hs.104800NM_004692; NM_032727; internexin neuronalCISintermediate filament protein, alpha539U133A219922_s_at168Hs.289019NM_030570; uroplakin 3B isoform aCISNM_182683; uroplakin 3B isoform cNM_182684; uroplakin 3B isoform b540U133A220026_at168Hs.227059NM_001442; fatty acid binding protein 4,CISadipocyte541U133A220779_at168Hs.149195NM_016233; peptidylarginine deiminase typeCISIII542U133A221204_s_at168Hs.326444NM_018058; cartilage acidic protein 1CIS543U133A221660_at168Hs.247831NM_000300; phospholipase A2, group IIACIS(platelets, synovial fluid)544U133A221671_x_at168Hs.377975NM_000299; plakophilin 1CIS545U133A221854_at168Hs.313068NM_000299; plakophilin 1CIS546U133A221872_at168Hs.82547NM_001958; eukaryotic translation elongationCISfactor 1 alpha 2547U133A200958_s_at168Hs.164067NM_005625; syndecan binding proteinCIS(syntenin)548U133A201877_s_at168Hs.249955NM_002719; gamma isoform of regulatoryCISsubunit B56, protein phosphatase 2A isoforma NM_178586; gamma isoform, of regulatorysubunit B56, protein phosphatase 2A isoformb NM_178587; gamma isoform of regulatorysubunit B56, protein phosphatase 2A isoformc NM_178588; gamma isoform of regulatorysubunit B56, protein phosphatase 2A isoform d549U133A201887_at168Hs.285115NM_001560; interleukin 13 receptor, alpha 1CISprecursor550U133A202076_at168Hs.289107NM_001166; baculoviral IAP repeat-CIScontaining protein 2551U133A202777_at168Hs.104315NM_007373; soc-2 suppressor of clear homologCIS552U133A204640_s_at168Hs.129951NM_003563; speckle-type POZ proteinCIS553U133A209004_s_at168Hs.5548NM_012161; F-box and leucine-rich repeatCISprotein 5 isoform 1 NM_033535; F-box andleucine-rich repeat protein 5 isoform 2554U133A209241_x_at168Hs.112028NM_015716; misshapen/NIK-related kinaseCISisoform 1 NM_153827; misshapen/NIK-relatedkinase isoform 3 NM_170663; misshapen/NIK-related kinase isoform 2555U133A209579_s_at168Hs.35947NM_003925; methyl-CpG binding domainCISprotein 4556U133A209630_s_at168Hs.444354NM_012164; F-box and WD-40 domain protein 2CIS557U133A212784_at168Hs.388236NM_015125; capicua homologCIS558U133A212802_s_at168Hs.287266CIS559U133A212899_at168Hs.129836NM_015076; cyclin-dependent kinase (CDC2-CISlike) 11560U133A213633_at168Hs.97858NM_018957; SH3-domain binding protein 1CIS561U133A217941_s_at168Hs.8117NM_018695; erbb2 interacting proteinCIS562U133A218150_at168Hs.342849NM_012097; ADP-ribosylation factor-like 5CISisoform 1 NM_177985; ADP-ribosylationfactor-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 Carcinoma 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 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., 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 present 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 reveals 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 expresison 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 expresison 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 expresison 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 expresison 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-progession 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 expresison 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 expresison 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.TM.-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 porous surfaces, 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 nucleoc 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 calorimetric 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-6,7,8trimethoxy- and the dimethylamino)calbenz 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 Iophine 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% H202 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 dose 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, i.e. 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. Subsequent 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 1Clinical data on all patients involved in the studyFollow-ProgressedTime toup timeGroupSampleHist.to:progressionmonthsTraining setNo prog.150-6Ta gr344No prog.997-1Ta gr224No prog.833-2Ta gr335No prog.1070-1 Ta gr333No prog.968-1Ta gr226No prog.625-1T1 gr312No prog.880-1T1 gr347No prog.815-1Ta gr249No prog.861-1Ta gr245No prog.669-1Ta gr255No prog.368-4Ta gr216No prog.898-1Ta gr217No prog.576-6Ta gr236Prog.747-3Ta gr2T1 gr36Prog.956-2Ta gr3T1 gr327Prog.1083-1 Ta gr2T1 gr31Prog.686-3Ta gr2T1 gr26Prog. 795-13Ta gr2T1 gr34Prog.865-1Ta gr2T1 gr25Prog.112-2Ta gr3T1 gr37Prog.825-3Ta gr3T1 gr36Prog.679-2Ta gr2T2+ gr331Prog.941-4Ta gr3T2+ gr310Prog.607-1T1 gr2T2+ gr33Prog.1017-1 T1 gr3T2+ gr38Prog.1276-1 T1 gr3T2+ gr37Prog.501-1T1 gr3T2+ gr326Prog.744-1T1 gr3T2+ gr314Prog.839-1T1 gr3T2+ gr312Test setNo prog.1008-1 Ta gr255No prog.1060-1 Ta gr248No prog.1086-1 Ta gr234No prog.1105-1 Ta gr231No prog.1145-1 Ta gr239No prog.1352-1 Ta gr226No prog.829-1Ta gr237No prog.942-1Ta gr237No prog.780-1Ta gr250Prog1327-1 Ta gr2T1 gr38Prog.1062-2 Ta gr3T1 gr34Prog.1354-1 Ta gr3T1 gr38Prog.1093-1 Ta gr3T1 gr35Prog.925-7Ta gr2T1 gr34Prog. 962-10Ta gr0T2+ gr31Prog.970-1Ta gr3T2+ gr31Prog.1027-1 Ta gr3T2+ gr32Prog.1252-1 T1 gr3T2+ gr35Prog.1191-1 T1 gr4T2+ gr41


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 probe-sets 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 (Hirawa 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 2The 200 best markers of progressionEosUnigene5%ExemplarHu03 IDBuild 133DescriptionT-testpermaccession#416640Hs.79404neuron-specific protein6.035.62BE262478442220Hs.8148selenoprotein T5.985.06AL037800426982Hs.173091ubiquitin-like 35.94.88AA149707416815Hs.80120UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-5.524.67U41514acetylgalactosaminyltransferase 1 (GalNAc-T1)435521Hs.6361mitogen-activated protein kinase kinase 1 interacting protein 15.244.51W23814447343Hs.236894ESTs, Highly similar to S02392 alpha-2-macroglobulin receptor5.234.44AA256641precursor [H. sapiens]452829Hs.63368ESTs, Weakly similar to TRHY_HUMAN TRICHOHYALI4.954.39AI955579[H. sapiens]414895Hs.116278Homo sapiens cDNA FLJ13571 fis, clone PLACE10084054.944.31AW894856426252Hs.28917ESTs4.94.26BE176980444604Hs.11441chromosome 1 open reading frame 84.894.17AW327695409632Hs.55279serine (or cysteine) proteinase inhibitor, clade B (ovalbumin),4.894.13W74001member 5446556Hs.15303KIAA0349 protein4.874.08AB002347426799Hs.303154popeye protein 34.864.03H14843428115Hs.300855KIAA0977 protein4.864.00AB023194419847Hs.184544Homo sapiens, clone IMAGE: 3355383, mRNA, partial cds4.823.97AW390601417839Hs.82712fragile X mental retardation, autosomal homolog 14.83.93AI815732428284Hs.183435NM_004545: Homo sapiens NADH dehydrogenase4.783.92AA535762(ubiquinone) 1 beta subcomplex, 1 (7 kD, MNLL) (NDUFB1),mRNA.422929Hs.94011ESTs, Weakly similar to MGB4_HUMAN MELANOMA-4.773.90AA356694ASSOCIATED ANTIGEN B4 [H. sapiens]414762Hs.77257KIAA0068 protein4.723.86AW068349453395Hs.377915mannosidase, alpha, class 2A, member 14.713.84D63998421311Hs.283609hypothetical protein PRO20324.653.82N71848446847Hs.82845Homo sapiens cDNA: FLJ21930 fis, clone HEP04301, highly4.653.82T51454similar to HSU90916 Human clone 23815 mRNA sequence413840Hs.356228RNA binding motif protein, X chromosome4.623.79AI301558418321Hs.84087KIAA0143 protein4.623.78D63477430604Hs.247309succinate-CoA ligase, GDP-forming, beta subunit4.613.74AV650537423185Hs.380062ornithine decarboxylase antizyme 14.613.74BE299590417615Hs.82314hypoxanthine phosphoribosyltransferase 1 (Lesch-Nyhan4.63.70BE548641syndrome)418504Hs.85335Homo sapiens mRNA; cDNA DKFZp564D1462 (from clone4.593.68BE159718DKFZp564D1462)400846sortilin-related receptor, L(DLR class) A repeats-containing4.573.66(SORL1)426028Hs.172028a disintegrin and metalloproteinase domain 10 (ADAM10)4.533.65NM_001110425243Hs.155291KIAA0005 gene product4.473.63N89487434978Hs.4310eukaryotic translation initiation factor 1A4.453.62AA321238409513Hs.54642methionine adenosyltransferase II, beta4.433.59AW966728433282Hs.49007hypothetical protein4.433.56BE539101421628Hs.106210hypothetical protein FLJ108134.373.56AL121317452170Hs.28285patched related protein translocated in renal cancer4.373.54AF064801440014Hs.6856ash2 (absent, small, or homeotic, Drosophila, homolog)-like4.373.52AW960782431857Hs.271742ADP-ribosyltransferase (NAD; poly (ADP-ribose) polymerase)-4.363.52W19144like 3417924Hs.82932cyclin D1 (PRAD1: parathyrold adenomatosis 1)4.353.51AU077231421733Hs.1420fibroblast growth factor receptor 3 (achondroplasia, thanatophoric4.343.50AL119671dwarfism)440197Hs.317714pallid (mouse) homolog, pallidin4.323.49AW340708434055Hs.3726x 003 protein4.323.48AF168712445831Hs.13351LanC (bacterial lantibiotic synthetase component C)-like 14.313.46NM_006055439632Hs.334437hypothetical protein MGC42484.293.45AW410714448813Hs.22142cytochrome b5 reductase b5R.24.283.44AF169802449268Hs.23412hypothetical protein FLJ201604.283.43AW369278429311Hs.198998conserved helix-loop-helix ubiquitous kinase4.283.42AF080157423599Hs.31731peroxiredoxin 54.273.41AI805664422913Hs.121599CGI-18 protein4.263.40NM_015947418127Hs.83532membrane cofactor protein (CD46, trophoblast-lymphocyte4.263.39BE243982cross-reactive antigen)425221Hs.155188TATA box binding protein (TBP)-associated factor, RNA4.253.38AV649864polymerase II, F, 55 kD426682Hs.2056UDP glycosyltransferase 1 family, polypeptide A94.233.37AV660038421101Hs.101840major histocompatibility complex, class I-like sequence4.233.37AF010446444037Hs.380932CHMP1.5 protein4.223.35AV647686443407Hs.348514ESTs, Moderately similar to 2109260A B cell growth factor4.213.35AA037683[H. sapiens]448625Hs.178470hypothetical protein FLJ226624.213.34AW970786450997Hs.35254hypothetical protein FLB64214.163.34AW580830444336Hs.10882HMG-box containing protein 14.153.33AF019214416977Hs.406103hypothetical protein FKSG444.143.32AW130242420613Hs.406637ESTs, Weakly similar to A47582 B-cell growth factor precursor4.133.31AI873871[H. sapiens]414843Hs.77492heterogeneous nuclear ribonucleoprotein A04.13.30BE386038408288Hs.16886gb: zI73d06.r1 Stratagene colon (937204) Homo sapiens4.093.29AA053601cDNA clone 5′, mRNA sequence422043Hs.110953retinoic acid induced 14.093.29AL133649432864Hs.359682calpastatin4.083.28D16217410047Hs.379753zinc finger protein 36 (KOX 18)4.063.28AI167810400773NM_003105*: Homo sapiens sortilin-related receptor, L(DLR4.063.27class) A repeats-containing (SORL1), mRNA.423960Hs.136309SH3-containing protein SH3GLB14.053.27AA164516449626Hs.112860zinc finger protein 2584.043.27AA774247429953Hs.226581COX15 (yeast) homolog, cytochrome c oxidase assembly4.043.24NM_004376protein428901Hs.146668KIAA1253 protein4.023.24AI929568420079Hs.94896PTD011 protein3.993.22NM_014051436576Hs.77542ESTs3.983.21AI458213412841Hs.101395hypothetical protein MGC113523.973.21AI751157431604Hs.264190vacuolar protein sorting 35 (yeast homolog)3.963.21AF175265428318Hs.356190ubiquitin B3.963.19BE300110430677Hs.359784desmoglein 23.953.19Z26317407955Hs.9343ESTs3.943.18BE536739426177Hs.167700Homo sapiens cDNA FLJ10174 fis, clone HEMBA10039593.923.17AA373452429802Hs.5367ESTs, Weakly similar to I38022 hypothetical protein3.923.17H09548[H. sapiens]423810Hs.132955BCL2/adenovirus E1B 19 kD-interacting protein 3-like3.923.16AL132665421475Hs.104640HIV-1 Inducer of short transcripts binding protein; lymphoma3.913.15AF000561related factor436472Hs.46366KIAA0948 protein3.913.14AL045404434263Hs.79187ESTs3.93.13N34895400843NM_003105*: Homo sapiens sortilin-related receptor, L(DLR3.93.13class) A repeats-containing (SORL1), mRNA.440357Hs.20950phospholysine phosphohistidine inorganic pyrophosphate3.893.12AA379353phosphatase437223Hs.330716Homo sapiens cDNA FLJ14368 fis, clone HEMBA10011223.883.12C15105426125Hs.166994FAT tumor suppressor (Drosophila) homolog3.863.11X87241432554Hs.278411NCK-associated protein 13.883.10AI479813422506Hs.300741sorcin3.853.10R20909413786Hs.13500ESTs3.833.09AW613780429561Hs.250646baculoviral IAP repeat-containing 63.833.08AF265555404977Insulin-like growth factor 2 (somatomedin A) (IGF2)3.833.08427722Hs.180479hypothetical protein FLJ201163.823.08AK000123400844NM_003105*: Homo sapiens sortilin-related receptor, L(DLR3.823.08class) A repeats-containing (SORL1), mRNA.426469Hs.363039methylmalonate-semialdehyde dehydrogenase3.813.07BE297886439578Hs.350547nuclear receptor co-repressor/HDAC3 complex subunit3.813.06AW263124426508Hs.170171glutamate-ammonia ligase (glutamine synthase)3.83.06W23184448524Hs.21356hypothetical protein DKFZp762K20153.793.06AB032948448357Hs.108923RAB38, member RAS oncogene family3.793.06N20169425097Hs.154545PDZ domain containing guanine nucleotide exchange factor3.773.05NM_014247(GEF)1421649Hs.106415peroxisome proliferative activated receptor, delta5.765.50AA721217427747Hs.180655serine/threonine kinase 125.415.03AW411425439010Hs.75216Homo sapiens cDNA FLJ13713 fis, clone PLACE2000398,4.574.80AW170332moderately similar to LAR PROTEIN PRECURSOR (LEUKOCYTEANTIGEN RELATED) (EC 3.1.3.48)438818Hs.30738ESTs4.494.59AW979008438013Hs.15670ESTs4.424.50AI002106452929Hs.172816neuregulin 14.374.40AW954938404826Target Exon4.224.32429124Hs.196914minor histocompatibility antigen HA-14.24.26AW505086421505Hs.285641KIAA1111 protein4.164.24AW249934428712Hs.190452KIAA0365 gene product4.144.19AW085131427239Hs.356512ubiquitin carrier protein4.114.10BE270447421595Hs.301685KIAA0620 protein4.14.07AB014520433844Hs.179647Homo sapiens cDNA FLJ12195 fis, clone MAMMA10008654.044.02AA610175443679Hs.9670hypothetical protein FLJ109484.014.00AK001810422959Hs.349256paired immunoglobulin-like receptor beta4.013.98AV647015452012Hs.279766kinesin family member 4A3.983.96AA307703435320Hs.117864ESTs3.973.91AA677934456332Hs.399939gb: nc39d05.r1 NCI_CGAP_Pr2 Homo sapiens cDNA clone,3.953.88AA228357mRNA sequence427999Hs.181369ubiquitin fusion degradation 1-like3.943.86AI435128427681Hs.284232tumor necrosis factor receptor superfamily, member 123.933.81AB018263(translocating chain-association membrane protein)413929Hs.75617collagen, type IV, alpha 23.933.79BE501689420116Hs.95231FH1/FH2 domain-containing protein3.93.77NM_013241433914Hs.112160Homo sapiens DNA heilcase homolog (PlF1) mRNA, partial3.883.75AF108138cds420732Hs.367762ESTs3.873.74AA789133452517gb: RC-BT068-130399-068 BT068 Homo sapiens cDNA,3.843.70AI904891mRNA sequence437524Hs.385719ESTs3.823.68AI627565435158Hs.65588DAZ associated protein 13.83.66AW663317448780Hs.267749Human DNA sequence from clone 366N23 on chromosome3.83.65W920716q27. Contains two genes similar to consecutive parts of theC. elegans UNC-93 (protein 1, C46F11.1) gene, a KIAA0173and Tubulin-Tyrosine Ligase LIKE gene, a Mitotic FeedbackControl Protein MADP2 H445084Hs.250848hypothetical protein FLJ147613.793.64H38914423138gb: EST385571 MAGE resequences, MAGM Homo sapiens3.753.60AW973426cDNA, mRNA sequence419602Hs.91521hypothetical protein3.743.59AW248434442549Hs.8375TNF receptor-associated factor 43.743.58AI751601460893Hs.25625hypothetical protein FLJ113233.733.55AK002185414223Hs.238246hypothetical protein FLJ224793.733.55AA954566444312Hs.351142ESTs3.723.53R44007425205Hs.155106receptor (calcitonin) activity modifying protein 23.713.51NM_005854432327Hs.274363neuroglobin3.713.49R36571451970Hs.211046ESTs3.673.48AI825732408049Hs.345588desmoplakin (DPI, DPII)3.673.45AW076098440100Hs.158549ESTs, Weakly similar to T2D3_HUMAN TRANSCRIPTION3.663.45BE382685INITIATION FACTOR TFIID 135 KDA SUBUNIT [H. sapiens]426468Hs.117558ESTs3.653.43AA379306402384NM_007181*: Homo sapiens mitogen-activated protein3.643.43kinase kinase kinase kinase 1 (MAP4K1), mRNA.458132Hs.103267hypothetical protein FLJ22548 similar to gene trap PAT 123.643.42AW247012447400Hs.18457hypothetical protein FLJ203153.643.42AK000322443893Hs.115472ESTs, Weakly similar to 2004399A chromosomal protein3.633.41BE079602[H. sapiens]424959Hs.153937activated p21cdc42Hs kinase3.623.40NM_005781409586Hs.55044DKFZP586H2123 protein3.63.39AL050214445692Hs.182099ESTs3.63.37AI248322433052Hs.293003ESTs, Weakly similar to PC4259 ferritin associated protein3.63.36AW971983[H. sapiens]421782Hs.108258actin binding protein; macrophin (microfilament and actin3.593.35AB029290filament cross-linker protein)414907Hs.77597polo (Drosophila)-like kinase3.583.34X90725454639gb: RC2-ST0158-091099-011-d05 ST0158 Homo sapiens3.573.33AW811633cDNA, mRNA sequence434547Hs.106124ESTs3.563.32R26240439130Hs.375195ESTs3.553.32AA306090413564gb: 601146990F1 NIH_MGC_19 Homo sapiens cDNA clone3.543.31BE2601205′, mRNA sequence443471Hs.398102Homo sapiens clone FLB3442 PRO0872 mRNA, complete3.533.31AW236939cds424415Hs.146580enolase 2, (gamma, neuronal)3.523.30NM_001975405036NM_021628*: Homo sapiens arachidonate lipoxygenase 33.523.29(ALOXE3), mRNA. VERSION NM_020229.1 GI422068Hs.104520Homo sapiens cDNA FLJ13694 fis, clone PLACE20001153.523.29AI807519424244Hs.143601hypothetical protein hCLA-iso3.523.28AV647184451867Hs.27192hypothetical protein dJ1057B20.23.513.26W74157429187Hs.163872ESTs, Weakly similar to S65657 alpha-1C-adrenergic receptor3.493.26AA447648splice form 2 [H. sapiens]415200Hs.78202SWI/SNF related, matrix associated, actin dependent regulator3.483.25AL040328of chromatin, subfamily a, member 4405667Target Exon3.483.25421075Hs.101474KIAA0807 protein3.473.23AB018350424909Hs.153752cell division cycle 25B3.463.22S78187451164Hs.60659ESTs, Weakly similar to T46471 hypothetical protein3.463.21AA015912DKFZp434L0130.1 [H. sapiens]438644Hs.129037ESTs3.463.20AI126162432258Hs.293039ESTs3.453.19AW973078411817Hs.72241mitogen-activated protein kinase kinase 23.453.19BE302900414918Hs.72222hypothetical protein FLJ134593.453.18AI219207437256Hs.97871Homo sapiens, clone IMAGE: 3845253, mRNA, partial cds3.433.17AL137404404208C6001282: gi|4504223|ref|NP_000172.1| glucuronidase, beta3.423.16[Homo sapiens] gi|114963|sp|P082421989Hs.110457Wolf-Hirschhorn syndrome candidate 13.43.15AJ007042438942Hs.6451PRO0659 protein3.393.14AW875398412649Hs.74369integrin, alpha 73.383.14NM_002206414840Hs.23823hairy/enhancer-of-split related with YRPW motif-like3.373.13R27319434831Hs.273397KIAA0710 gene product3.353.12AA248060431842Hs.271473epithelial protein up-regulated in carcinoma, membrane3.343.11NM_005764associated protein 17402328Target Exon3.343.10405371NM_005569*: Homo sapiens LIM domain kinase 2 (LIMK2),3.333.10transcript variant 2a, mRNA.441650Hs.132545ESTs3.323.09AI261960418629Hs.86859growth factor receptor-bound protein 73.33.09BE247550406002Target Exon3.33.08420307Hs.66219ESTs3.293.08AW502869425093Hs.154525KIAA1076 protein3.283.07AB028999427351Hs.123253hypothetical protein FLJ220093.283.07AW402593417900Hs.82906CDC20 (cell division cycle 20, S. cerevisiae, homolog)3.283.06BE250127457228Hs.195471Human cosmid CRI-JC2015 at D10S289 in 10sp133.273.05U15177421026Hs.101067GCN5 (general control of amino-acid synthesis, yeast, homolog)-3.273.04AL047332like 2430746Hs.406256ESTs3.273.03AW977370409556Hs.54941phosphorylase kinase, alpha 2 (liver)3.273.03D38616451225Hs.57655ESTs3.263.03AI433694404913NM_024408*: Homo sapiens Notch (Drosophila) homolog 23.253.02(NOTCH2), mRNA. VERSION NM_024410.1 GI404875NM_022819*: Homo sapiens phospholipase A2, group IIF3.233.02(PLA2G2F), mRNA. VERSION NM_020245.2 GI404606Target Exon3.233.01414732Hs.77152minichromosome maintenance deficient (S. cerevisiae) 73.223.01AW410976425380Hs.32148AD-015 protein3.223.00AA356389421186Hs.270563ESTs, Moderately similar to T12512 hypothetical protein3.212.98AI798039DKFZp434G232.1 [H.sapiens]445462Hs.288649hypothetical protein MGC30773.22.97AA378776


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 prediction 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 3The 45 optimal genes for disease progression prediction.EosUnigene5%ExemplarHu03 IDBuild 133DescriptionT-TestpermGene NameAccessionCV439010Hs.75216protein tyrosine phosphatase, receptor4.574.39PTPRFAW17033229type, F429124Hs.196914minor histocompatibility antigen HA-14.204.09HA-1AW50508629421649Hs.106415peroxisome proliferative activated receptor,5.765.64PPARDAA72121729delta433914Hs.112160DNA helicase homolog (PIF1)3.883.61PIF1AF10813829429187Hs.163872ESTs, Weakly similar to hypothetical3.493.17AA44764828protein FLJ20489422765Hs.1578baculoviral IAP repeat-containing 52.682.56BIRC5AW40970128(survivin)433844Hs.179647ESTs4.043.80AA61017526450893Hs.25625Hypothetical protein FLJ113233.733.46FLJ11323AK00218525452866Hs.268016ESTs3.103.02R2696924424909Hs.153752cell division cycle 25B3.463.16CDC25BS7818724452929Hs.172816neuregulin 14.374.23NRG1AW95493823420116Hs.95231formin homology 2 domain containing 13.903.63FHOD1NM_01324122453963Hs.28959cDNA FLJ36513 fis, clone3.442.88AA04031129TRACH2001523429561Hs.250646baculoviral IAP repeat-containing 63.833.03BIRC6AF26555529(apollon)418127Hs.83532membrane cofactor protein (CD46,4.263.37MCPBE24398229trophoblast-lymphocyte cross-reactiveantigen)422119Hs.111862KIAA0590 gene product2.331.95KIAA0590AI27782929435521Hs.6361mitogen-activated protein kinase kinase5.244.53MAP2K1IP1W23814291 interacting protein 1409632Hs.55279serine (or cysteine) proteinase Inhibitor,4.894.11SERPINB5W7400129clade B (ovalbumin), member 5452829Hs.63368ESTs4.954.31AI95557929416640Hs.79404DNA segment on chromosome 46.035.51D4S234EBE26247829(unique) 234 expressed sequence425097Hs.154545PDZ domain containing guanine nucleotide3.773.18PDZ-GEF1NM_01424728exchange factor(GEF)1445926Hs.334826splicing factor 3b, subunit 1, 155 kDa2.402.03SF3B1AF05428428437325Hs.5548F-box and leucine-rich repeat protein 52.482.09FBXL5AF14248128448813Hs.22142cytochrome b5 roductase b5R.24.283.41LOC51700AF16980228426799Hs.303154ESTs4.864.04H1484328446847Hs.82845ESTs4.653.79T5145428428016Hs.181461ariadne homolog, ubiquitin-conjugating3.773.15ARIH1AJ24319027enzyme E2 binding protein, 1 (Drosophila)418321Hs.84087KIAA0143 protein4.623.76KIAA0143D6347727422984Hs.351597ESTs3.502.93W2861426408688Hs.152925KIAA1268 protein3.522.95KIAA1268AI63452226440357Hs.20950phospholysine phosphohistidine Inorganic3.893.07LHPPAA37935326pyrophosphate phosphatase420269Hs.96264alpha thalassemia/mental retardation3.392.85ATRXU7293726syndrome X-linked (RAD54 (S. cerevisiae)homolog)423185?omithine decarboxylase antizyme 14.613.71OAZ1BE29959026443407Hs.348514clone IMAGE: 4052238, mRNA, partial4.213.32AA03768325cds457329Hs.359682calpastatin3.592.99CASTAI63486025452714Hs.30340KIAA1165: likely ortholog of mouse3.623.01KIAA1165AW77099425Nedd4 WW domain-binding protein 5A444773Hs.11923hypothetical protein DJ167A19.13.713.11DJ167A19.1BE15625624418504Hs.85335ESTs4.593.67BE15971824444604Hs.11441Chromosome 1 open reading frame 84.894.17C1orf8AW32769523410691Hs.65450reticulon 4RTN4AW23922623430604Hs.247309succinate-CoA ligase, GDP-forming,4.613.72SUCLG2AV65053723beta subunit421311Hs.283609muscleblind-ilke protein MBLL394.653.82MBLL39N7184823439632Hs.334437hypothetical protein MGC42484.293.42MGC4248AW41071422417924Hs.82932cyclin D1 (PRAD1: parathyroid adenomatosis4.353.49CCND1AU077231221)453395Hs.377915mannosidase, alpha, class 2A, member 14.713.84MAN2A1D6399822


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 probe-sets 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 previousley (Dyrskjot et al. 2003).


Custom Oligonucleotide Microarray Procedures


Three 60 mer 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 4Disease course information of all patients involved-class discovery.Tumour examinedReviewedCarcinomaGroupPatientPrevious tumourson arrayPatternhistologySubsequent tumoursin situ*A709-1Ta gr 2 (200297)PapillaryTa gr3no968-1Ta gr 2 (011098)Papillary+Ta gr 2 (150101)no934-1Ta gr 2 (220798)Papillary+no928-1Ta gr 2 (240698)Papillary+no930-1Ta gr 2 (300698)Papillary+noB989-1Ta gr 3 (281098)Papillary+no1264-1 Ta gr 3 (130600)Papillary+Ta gr 2 (231000)noTa gr 2 (220101)Ta gr 2 (300401)876-5Ta gr 2 (230398)Ta gr 3 (170400)Papillary+noTa gr 2 (271098)Ta gr 2 (090699)Ta gr 2 (011199)669-7Ta gr 2 (101296)Ta gr 3 (230899)PapillaryTa gr2Ta gr 2 (120100)noTa 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)716-2Ta gr 2 (070397)Ta gr 3 (230497)Papillary+Ta gr 2 (040697)noTa gr 1 (170698)C1070-1Ta gr 3 (150399)Papillary+Ta gr 3 (291099)Subsequent visit956-2Ta gr 3 (061299)Papillary+Ta gr 3 (061200)Sampling visit1062-2Ta gr 3 (120799)Papillary+T1 gr 3 (161199)Sampling visit1166-1Ta gr 3 (271099)Papillary+Sampling visit1330-1Ta gr 3 (311000)Papillary+Sampling visitD112-10Ta gr 2 (070794)Ta gr 3 (060198)Papillary+Ta gr 3 (110698)Previous visitTa 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)320-7T1 gr 3 (011194)Ta gr 3 (290997)Papillary+Ta gr 3 (290198)Sampling visitT1 gr 3 (150896)Ta gr 3 (290698)Ta gr 3 (100897)747-7Ta gr 2 (010597)Ta gr 3 (161298)Papillary+Ta gr 2 (050599)Sampling visitTa 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)967-3T1 gr 3 (280998)Ta gr 3 (140699)Papillary+T1 gr 3 (080999)Sampling visitT1 gr 3 (250199)E625-1T1 gr 3 (200996)Papillary+No847-1T1 gr 3 (210198)Papillary+No1257-1T1 gr 3 (240500)Solid+Sampling visit919-1T1 gr 3 (220698)Papillary+No880-1T1 gr 3 (300398)Papillary+Ta gr 2 (091198)NoTa gr 1 (090399)Ta gr 2 (050900)Ta gr 2 (190301)812-1T1 gr 3 (061098)Papillary+No1269-1T1 gr 3 (230600)PapillaryNo1083-2Ta gr 2 (280499)T1 gr 3 (120599)PapillaryNo1238-1T1 gr 3 (020500)Papillary+T2 gr 3 (211100)NoTa gr 2 (211100)1065-1T1 gr 3 (160399)PapillarySubsequent visit1134-1T1 gr 3 (181099)PapillaryT2 gr3T1 gr 3 (280200)Sampling visitT1 gr 3 (020500)T1 gr 3 (131100)F1164-1T2+ gr 4 (101299)Solidgr 3No1032-1T2+ gr ? (050199)MixedNot measured1117-1T2+ gr 3 (010999)Solid+Sampling visit1178-1T2+ gr 3 (200100)Solid+Not measured1078-1T2+ gr 3 (120499)Solid+Not measured875-1T2+ gr 3 (180398)Solid+No1044-1T2+ gr 3 (010299)Solid+T2+ gr 3 (060999)Not measured1133-1T2+ gr 3 (081099)Solid+Not measured1068-1T2+ gr 3 (220399)Solid+No937-1T2+ gr 3 (280798)SolidNot 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 subfraction 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. 70. 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 5Genes for classifying samples with squamous metaplasiaUniGeneChip acc. #Build 162descriptionD83657_atHs.19413NM_005621; S100calcium-binding protein A12HG3945-HT4215_atJ00124_atL05187_atL05188_f_atHs.505327L10343_atHs.112341NM_002638; skin-derivedprotease inhibitor 3preproproteinL42583_f_atHs.367762NM_005554; keratin 6AL42601_f_atHs.367762NM_005554; keratin 6AL42611_f_atHs.446417NM_173086;keratin 6 isoform K6eM19888_atHs.1076NM_003125; smallproline-rich protein 1B (cornifin)M20030_f_atHs.505352M21005_atM21302_atHs.505327M21539_atHs.2421NM_006518;small proline-rich protein 2CM86757_s_atHs.112408NM_002963;S100 calcium-binding protein A7S72493_s_atHs.432448NM_005557;keratin 16U70981_atHs.336046NM_000640; interleukin13 receptor, alpha 2 precursorV01516_f_atHs.367762NM_005554;keratin 6AX53065_f_atX57766_atHs.143751NM_005940; matrixmetalloproteinase 11 preproproteinZ19574_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 6Table 6. Summary of stage related gene expressionFunctional gene clustersaNuclearExtracellularImmuneTumour stageTranscriptionprocessesProliferationMatrix remodellingmatrixsystemTa gr2↓↓Ta gr3↑↑↑↑↑↑↑↓↓T1 gr3b↑↑bbT2 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 usefull. 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 7Feature: Accession number on HuGene fl array. Number: Number of times used inthe 80 genes cross validation loops. Test (B/W): see below.UnigeneTestFeatureBuild 162DescriptionNumber(B/W)AF000231_atHs.75618NM_004663; Ras-related protein Rab-11A3326.77D13666_s_atHs.136348NM_006475; osteoblast specific factor 2 (fasciclin I-like)3327.71D49372_s_atHs.54460NM_002986; small inducible cytokine A11 precursor3125.78D83920_atHs.440898NM_002003; ficolin 1 precursor3331.18D86479_atHs.439463NM_001129; adipocyte enhancer binding protein 1 precursor3328.29D89077_atHs.75367NM_006748; Src-like-adaptor3330.03D89377_atHs.89404NM_002449; msh homeo box homolog 23351.50HG4069-HT4339_s_at2725.06HG67-HT67_f_at3327.81HG907-HT907_at3325.76J02871_s_atHs.436317NM_000779; cytochrome P450, famliy 4, subfamily B, polypeptide 13332.61J03278_atHs.307783NM_002609; platelet-derived growth factor receptor beta3328.02precursorJ04058_atHs.169919NM_000126; electron transfer flavoprotein, alpha polypeptide3329.46J05032_atHs.32393NM_001349; aspartyl-tRNA synthetase3338.21J05070_atHs.151738NM_004994; matrix metalloproteinase 9 preproprotein3335.34J05448_atHs.79402NM_002694; DNA directed RNA polymerase II polypeptide3226.51C NM_032940; DNA directed RNA polymerase II polypeptide CK01396_atHs.297681NM_000295; serine (or cysteine) proteinase inhibitor, clade3328.66A (alpha-1 antiproteinase, antitrypsin), member 1L13720_atHs.437710NM_000820; growth arrest-specific 63329.69M12125_atHs.300772NM_003289; tropomyosin 2 (beta)2824.89M15395_atHs.375957NM_000211; integrin beta chain, beta 2 precursor3329.40M16591_s_atHs.89555NM_002110; hemopoietic cell kinase isoform p61HCK3332.34M20530_at3330.28M23178_s_atHs.73817NM_002983; chemokine (C—C motif) ligand 33335.36M32011_atHs.949NM_000433; neutrophil cytosolic factor 23341.88M33195_atHs.433300NM_004106; Fc fragment of IgE, high affinity I, receptor for,3330.40gamma polypeptide precursorM55998_s_atHs.172928NM_000088; alpha 1 type I collagen preproprotein3326.83M57731_s_atHs.75765NM_002089; chemokine (C—X—C motif) ligand 23331.84M68840_atHs.183109NM_000240; monoamine oxidase A3332.39M69203_s_atHs.75703NM_002984; chemokine (C—C motif) ligand 4 precursor3336.21M72885_rna1_s_at3327.94M83822_atHs.209846NM_006726; LPS-responsive vesicle trafficking, beach and3326.44anchor containingS77393_atHs.145754NM_016531; Kruppel-like factor 3 (basic)3349.85U01833_atHs.81469NM_002484; nucleotide binding protein 1 (MinD homolog, E. coli)3330.62U07231_atHs.309763NM_002092; G-rich RNA sequence binding factor 13339.10U09937_rna1_s_at3330.88U10550_atHs.79022NM_005261; GTP-binding mitogen-induced T-cell protein2825.26NM_181702; GTP-binding mitogen-induced T-cell proteinU20158_atHs.2488NM_005565; lymphocyte cytosolic protein 23332.41U41315_rna1_s_at3343.56U47414_atHs.13291NM_004354; cyclin G23344.42U49352_atHs.414754NM_001359; 2,4-dienoyl CoA reductase 1 precursor3337.04U50708_atHs.1265NM_000056; branched chain keto acid dehydrogenase E1,3342.89beta polypeptide precursor NM_183050; branched chainketo acid dehydrogenase E1, beta polypeptide precursorU52101_atHs.9999NM_001425; epithelial membrane protein 33329.86U64520_atHs.66708NM_004781; vesicle-associated membrane protein 3 (cellubrevin)3330.17U65093_atHs.82071NM_006079; Cbp/p300-interacting transactivator, with3332.07Glu/Asp-rich carboxy-terminal domain, 2U68019_atHs.288261NM_005902; MAD, mothers against decapentaplegic homolog 33126.70U68385_atHs.3809233331.56U74324_atHs.90875NM_002871; RAB-interacting factor3330.26U77970_atHs.321164NM_002518; neuronal PAS domain protein 2 NM_032235;3350.37U90549_atHs.236774NM_006353; high mobility group nucleosomal binding domain 43332.16X04085_rna1_at2825.13X07743_atHs.77436NM_002664; pleckstrin3328.13X13334_atHs.75627NM_000591; CD14 antigen precursor3335.79X14046_atHs.153053NM_001774; CD37 antigen3024.70X15880_atHs.415997NM_001848; collagen, type VI, alpha 1 precursor3331.51X15882_atHs.420269NM_001849; alpha 2 type VI collagen isoform 2C2 precursor3332.32NM_058174; alpha 2 type VI collagen isoform 2C2a precursorNM_058175; alpha 2 type VI collagen isoform 2C2aprecursorX51408_atHs.380138NM_001822; chimerin (chimaerin)3330.51X53800_s_atHs.89690NM_002090; chemokine (C—X—C motif) ligand 33333.63X54489_rna1_at3333.57X57579_s_at3341.43X64072_s_atHs.375957NM_000211; integrin beta chain, beta 2 precursor3343.21X67491_f_atHs.355697NM_005271; glutamate dehydrogenase 13330.97X68194_atHs.80919NM_006754; synaptophysin-like protein isoform a3346.53NM_182715; synaptophysin-like protein isoform bX73882_atHs.254605NM_003980; microtubule-associated protein 73353.16X78520_atHs.372528NM_001829; chloride channel 33347.38Y00787_s_atHs.624NM_000584; interleukin 8 precursor3227.54Z12173_atHs.334534NM_002076; glucosamine (N-acetyl)-6-sulfatase precursor3025.44Z19554_s_atHs.435800NM_003380; vimentin2724.59Z26491_s_atHs.240013NM_000754; catechol-O-methyltransferase isoform MB-3226.92COMT NM_007310; catechol-O-methyltransferase isoformS-COMTZ29331_atHs.372758NM_003344; ubiquitin-conjugating enzyme E2H isoform 13333.49NM_182697; ubiquitin-conjugating enzyme E2H isoform 2Z48605_atHs.421825NM_006903; inorganic pyrophosphatase 2 isoform 23344.45NM_176865; NM_176866; inorganic pyrophosphatase 2isoform 3 NM_176867; inorganic pyrophosphatase 2 isoform4 NM_176869; inorganic pyrophosphatase 2 Isoform 1Z74615_atHs.172928NM_000088; alpha 1 type I collagen preproprotein3355.18


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 psudogroups 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.
embedded imageembedded image


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 9320 genes for classifierUniGeneChip acc. #Build 162descriptionAB000220_atHs.171921NM_006379; semaphorin3CAB000220_atHs.171921NM_006379; semaphorin3CAC002073_cds1_atAF000231_atHs.75618NM_004663; Ras-related protein Rab-11AD10922_s_atHs.99855NM_001462; formylpeptide receptor-like 1D10925_atHs.301921NM_001295;chemokine (C—C motif)receptor 1D11086_atHs.84NM_000206; Interleukin2 receptor, gammachain, precursorD11151_atHs.211202NM_001957; endothelinreceptor type AD13435_atHs.426142NM_002643; phosphatidylinositolglycan,class F isoform 1NM_173074; phosphatidylinositolglycan,class F isoform 2D13666_s_atHs.136348NM_006475; osteoblastspecific factor 2 (fasciclinI-like)D14520_atHs.84728NM_001730; Kruppel-like factor 5D21878_atHs.169998NM_004334; bonemarrow stromal cellantigen 1 precursorD26443_atHs.371369NM_004172; solutecarrier family 1 (glialhigh affinity glutamatetransporter), member 3D28589_atHs.17719D42046_atHs.194665D45370_atHs.74120NM_006829; adiposespecific 2D49372_s_atHs.54460NM_002986; smallinducible cytokine A11precursorD50495_atHs.224397NM_003195; transcriptionelongation factor A(SII), 2D63135_atHs.27935NM_032646; tweetyhomolog 2D64053_atHs.198288NM_002849; proteintyrosine phosphatase,receptor type, R isoform1 precursorNM_130846; proteintyrosine phosphatase,receptor type, R isoform 2D83920_atHs.440898NM_002003; ficolin 1precursorD85131_s_atHs.433881NM_002383; MYC-associated zinc fingerproteinD86082_s_atHs.413482NM_004649; chromosome21 open readingframe 33D86479_atHs.439463NM_001129; adipocyteenhancer binding protein1 precursorD86957_atHs.307944D86959_atHs.105751NM_014720; Ste20-related serine/threoninekinaseD86976_atHs.196914D87433_atHs.301989NM_015136; stabilin 1D87443_atHs.409862NM_014758; sortingnexin 19D87682_atHs.134792D89077_atHs.75367NM_006748; Src-like-adaptorD89377_atHs.89404NM_002449; mshhomeo box homolog 2D90279_s_atHs.433695NM_000093; alpha 1type V collagen preproproteinHG1996-HT2044_atHG2090-HT2152_s_atHG2463-HT2559_atHG2994-HT4850_s_atHG3044-HT3742_s_atHG3187-HT3366_s_atHG3342-HT3519_s_atHG371-HT26388_s_atHG4069-HT4339_s_atHG67-HT67_f_atHG907-HT907_atJ02871_s_atHs.436317NM_000779; cytochromeP450, family 4,subfamily B, polypeptide 1J03040_atHs.111779NM_003118; secretedprotein, acidic, cysteine-rich (osteonectin)J03060_atJ03068_atJ03241_s_atHs.2025NM_003239; transforminggrowth factor, beta 3J03278_atHs.307783NM_002609; platelet-derived growth factorreceptor beta precursorJ03909_atJ03925_atHs.172631NM_000632; integrinalpha M precursorJ04056_atHs.88778NM_001757; carbonylreductase 1J04058_atHs.169919NM_000126; electrontransfer flavoprotein,alpha polypeptideJ04093_s_atHs.278896NM_019075; UDPglycosyltransferase 1family, polypeptide A10J04130_s_atHs.75703NM_002984;chemokine (C—C motif)ligand 4 precursorJ04152_ma1_s_atJ04162_atHs.372679NM_000569; Fc fragmentof IgG, low affinityIIIa, receptor for (CD16)J04456_atHs.407909NM_002305; beta-galactosidase bindinglectin precursorJ05032_atHs.32393NM_001349; aspartyl-tRNA synthetaseJ05036_s_atHs.1355NM_001910; cathepsinE isoform a preproproteinNM_148964; cathepsinE isoform bpreproproteinJ05070_atHs.151738NM_004994; matrixmetalloproteinase 9preproproteinJ05448_atHs.79402NM_002694; DNAdirected RNA polymeraseII polypeptide CNM_032940; DNAdirected RNA polymeraseII polypeptide CK01396_atHs.297681NM_000295; serine (orcysteine) proteinaseInhibitor, clade A (alpha-1 antiproteinase,antitrypsin), member 1K03430_atL06797_s_atHs.421986NM_003467;chemokine (C—X—Cmotif) receptor 4L10343_atHs.112341NM_002638; skin-derived protease inhibitor3 preproproteinL11708_atHs.155109NM_002153; hydroxysteroid(17-beta) dehydrogenase 2L13391_atHs.78944NM_002923; regulatorof G-protein signalling2, 24 kDaL13698_atHs.65029NM_002048; growtharrest-specific 1L13720_atHs.437710NM_000820; growtharrest-specific 6L13923_atHs.750NM_000138; fibrillin 1AB000220_atHs.171921NM_006379; semaphorin3CAC002073_cds1_atAF000231_atHs.75618NM_004663; Ras-related protein Rab-11AD10922_s_atHs.99855NM_001462; formylpeptide receptor-like 1D10925_atHs.301921NM_001295;chemokine (C—C motif)receptor 1D11086_atHs.84NM_000206; interleukin2 receptor, gammachain, precursorD11151_atHs.211202NM_001957; endothelinreceptor type AD13435_atHs.426142NM_002643; phosphatidylinositolglycan,class F isoform 1NM_173074; phosphatidylinositolglycan,class F isoform 2D13666_s_atHs.136348NM_006475; osteoblastspecific factor 2 (fasciclinI-like)D14520_atHs.84728NM_001730; Kruppel-like factor 5D21878_atHs.169998NM_004334; bonemarrow stromal cellantigen 1 precursorD26443_atHs.371369NM_004172; solutecarrier family 1 (glialhigh affinity glutamatetransporter), member 3D28589_atHs.17719D42046_atHs.194665D45370_atHs.74120NM_006829; adiposespecific 2D49372_s_atHs.54460NM_002986; smallinducible cytokine A11precursorD50495_atHs.224397NM_003195; transcriptionelongation factor A(SII), 2D63135_atHs.27935NM_032646; tweetyhomolog 2D64053_atHs.198288NM_002849; proteintyrosine phosphatase,receptor type, R isoform1 precursorNM_130846; proteintyrosine phosphatase,receptor type, R isoform 2D83920_atHs.440898NM_002003; ficolin 1precursorD85131_s_atHs.433881NM_002383; MYC-associated zinc fingerproteinD86062_s_atHs.413482NM_004649; chromosome21 open readingframe 33D86479_atHs.439463NM_001129; adipocyteenhancer binding protein1 precursorD86957_atHs.307944D86959_atHs.105751NM_014720; Ste20-related serine/threoninekinaseD86976_atHs.196914D87433_atHs.301989NM_015136; stabilin 1D87443_atHs.409862NM_014758; sortingnexin 19D87682_atHs.134792D89077_atHs.75367NM_006748; Src-like-adaptorD89377_atHs.89404NM_002449; mshhomeo box homolog 2D90279_s_atHs.433695NM_000093; alpha 1type V collagen preproproteinHG1996-HT2044_atHG2090-HT2152_s_atHG2463-HT2559_atHG2994-HT4850_s_atHG3044-HT3742_s_atHG3187-HT3366_s_atHG3342-HT3519_s_atHG371-H726388_s_atHG4069-HT4339_s_atHG67-HT67_f_atHG907-HT907_atJ02871_s_atHs.436317NM_000779; cytochromeP450, family 4,subfamily B, polypeptide 1J03040_atHs.111779NM_003118; secretedprotein, acidic, cysteine-rich (osteonectin)J03060_atJ03068_atJ03241_s_atHs.2025NM_003239; transforminggrowth factor, beta 3J03278_atHs.307783NM_002609; platelet-derived growth factorreceptor beta precursorJ03909_atJ03925_atHs.172631NM_000632; integrinalpha M precursorJ04056_atHs.88778NM_001757; carbonylreductase 1J04058_atHs.169919NM_000126; electrontransfer flavoprotein,alpha polypeptideJ04093_s_atHs.278896NM_019075; UDPglycosyltransferase 1family, polypeptide A10J04130_s_atHs.75703NM_002984;chemokine (C—C motif)ligand 4 precursorJ04152_rna1_s_atJ04162_atHs.372679NM_000569; Fc fragmentof IgG, low affinityIIIa, receptor for (CD16)J04456_atHs.407909NM_002305; beta-galactosidase bindinglectin precursorJ05032_atHs.32393NM_001349; aspartyl-tRNA synthetaseJ05036_s_atHs.1355NM_001910; cathepsinE isoform a preproproteinNM_148964; cathepsinE isoform bpreproproteinJ05070_atHs.151738NM_004994; matrixmetalloproteinase 9preproproteinJ05448_atHs.79402NM_002694; DNAdirected RNA polymeraseII polypeptide CNM_032940; DNAdirected RNA polymeraseII polypeptide CK01396_atHs.297681NM_000295; serine (orcysteine) proteinaseinhibitor, clade A (alpha-1 antiproteinase,antitrypsin), member 1K03430_atL06797_s_atHs.421986NM_003467;chemokine (C—X—Cmotif) receptor 4L10343_atHs.112341NM_002638; skin-derived protease inhibitor3 preproproteinL11708_atHs.155109NM_002153; hydroxysteroid(17-beta) dehydrogenase 2L13391_atHs.78944NM_002923; regulatorof G-protein signalling2, 24 kDaL13698_atHs.65029NM_002048; growtharrest-specific 1L13720_atHs.437710NM_000820; growtharrest-specific 6L13923_atHs.750NM_000138; fibrillin 1AB000220_atHs.171921NM_006379; semaphorin3CAC002073_cds1_atAF000231_atHs.75618NM_004663; Ras-related protein Rab-11AD10922_s_atHs.99855NM_001462; formylpeptide receptor-like 1D10925_atHs.301921NM_001295;chemokine (C—C motif)receptor 1D11086_atHs.84NM_000206; interleukin2 receptor, gammachain, precursorD11151_atHs.211202NM_001957; endothelinreceptor type AD13435_atHs.426142NM_002643; phosphatidylinositolglycan,class F isoform 1NM_173074; phosphatidylinositolglycan,class F isoform 2D13666_s_atHs.136348NM_006475; osteoblastspecific factor 2 (fasciclinI-like)D14520_atHs.84728NM_001730; Kruppel-like factor 5D21878_atHs.169998NM_004334; bonemarrow stromal cellantigen 1 precursorD26443_atHs.371369NM_004172; solutecarrier family 1 (glialhigh affinity glutamatetransporter), member 3D28589_atHs.17719D42046_atHs.194665D45370_atHs.74120NM_006829; adiposespecific 2D49372_s_atHs.54460NM_002986; smallinducible cytokine A11precursorD50495_atHs.224397NM_003195; transcriptionelongation factor A(SII), 2D63135_atHs.27935NM_032646; tweetyhomolog 2D64053_atHs.198288NM_002849; proteintyrosine phosphatase,receptor type, R isoform1 precursorNM_130846; proteintyrosine phosphatase,receptor type, R isoform 2D83920_atHs.440898NM_002003; ficolin 1precursorD85131_s_atHs.433881NM_002383; MYC-associated zinc fingerproteinD86062_s_atHs.413482NM_004849; chromosome21 open readingframe 33D86479_atHs.439463NM_001129; adipocyteenhancer binding protein1 precursorD86957_atHs.307944D86959_atHs.105751NM_014720; Ste20-related serine/threoninekinaseD86976_atHs.196914D87433_atHs.301989NM_015136; stabilin 1D87443_atHs.409862NM_014758; sortingnexin 19D87682_atHs.134792D89077_atHs.75367NM_006748; Src-like-adaptorD89377_atHs.89404NM_002449; mshhomeo box homolog 2D90279_s_atHs.433695NM_000093; alpha 1type V collagen preproproteinHG1996-HT2044_atHG2090-HT2152_s_atHG2463-HT2559_atHG2994-HT4850_s_atHG3044-HT3742_s_atHG3187-HT3366_s_atHG3342-HT3519_s_atHG371-HT26388_s_atHG4069-HT4339_s_atHG67-HT67_f_atHG907-HT907_atJ02871_s_atHs.436317NM_000779; cytochromeP450, family 4,subfamily B, polypeptide 1J03040_atHs.111779NM_003118; secretedprotein, acidic, cysteine-rich (osteonectin)J03060_atJ03068_atJ03241_s_atHs.2025NM_003239; transforminggrowth factor, beta 3J03278_atHs.307783NM_002609; platelet-derived growth factorreceptor beta precursorJ03909_atJ03925_atHs.172631NM_000632; integrinalpha M precursorJ04056_atHs.88778NM_001757; carbonylreductase 1J04058_atHs.169919NM_000126; electrontransfer flavoprotein,alpha polypeptideJ04093_s_atHs.278896NM_019075; UDPglycosyltransferase 1family, polypeptide A10J04130_s_atHs.75703NM_002984;chemokine (C—C motif)ligand 4 precursorJ04152_rna1_s_atJ04162_atHs.372679NM_000569; Fc fragmentof IgG, low affinityIIIa, receptor for (CD16)J04456_atHs.407909NM_002305; beta-galactosidase bindinglectin precursorJ05032_atHs.32393NM_001349; aspartyl-tRNA synthetaseJ05036_s_atHs.1355NM_001910; cathepsinE isoform a preproproteinNM_148964; cathepsinE isoform bpreproproteinJ05070_atHs.151738NM_004994; matrixmetalloproteinase 9preproproteinJ05448_atHs.79402NM_002694; DNAdirected RNA polymeraseII polypeptide CNM_032940; DNAdirected RNA polymeraseII polypeptide CK01396_atHs.297681NM_000295; serine (orcysteine) proteinaseinhibitor, clade A (alpha-1 antiproteinase,antitrypsin), member 1K03430_atL06797_s_atHs.421986NM_003467;chemokine (C—X—Cmotif) receptor 4L10343_atHs.112341NM_002638; skin-derived protease inhibitor3 preproproteinL11708_atHs.155109NM_002153; hydroxysteroid(17-beta) dehydrogenase 2L13391_atHs.78944NM_002923; regulatorof G-protein signalling2, 24 kDaL13698_atHs.65029NM_002048; growtharrest-specific 1L13720_atHs.437710NM_000820; growtharrest-specific 6L13923_atHs.750NM_000138; fibrillin 1AB000220_atHs.171921NM_006379; semaphorin3CAC002073_cds1_atAF000231_atHs.75618NM_004663; Ras-related protein Rab-11AD10922_s_atHs.99855NM_001462; formylpeptide receptor-like 1D10925_atHs.301921NM_001295;chemokine (C—C motif)receptor 1D11086_atHs.84NM_000206; interleukin2 receptor, gammachain, precursorD11151_atHs.211202NM_001957; endothelinreceptor type AD13435_atHs.426142NM_002643; phosphatidylinositolglycan,class F isoform 1NM_173074; phosphatidylinositolglycan,class F isoform 2D13666_s_atHs.136348NM_006475; osteoblastspecific factor 2 (fasciclinI-like)D14520_atHs.84728NM_001730; Kruppel-like factor 5D21878_atHs.169998NM_004334; bonemarrow stromal cellantigen 1 precursorD26443_atHs.371369NM_004172; solutecarrier family 1 (glialhigh affinity glutamatetransporter), member 3D28589_atHs.17719D42046_atHs.194665D45370_atHs.74120NM_006829; adiposespecific 2D49372_s_atHs.54460NM_002986; smallinducible cytokine A11precursorD50495_atHs.224397NM_003195; transcriptionelongation factor A(SII), 2D63135_atHs.27935NM_032646; tweetyhomolog 2D64053_atHs.198288NM_002849; proteintyrosine phosphatase,receptor type, R isoform1 precursorNM_130846; proteintyrosine phosphatase,receptor type, R isoform 2D83920_atHs.440898NM_002003; ficolin 1precursorD85131_s_atHs.433881NM_002383; MYC-associated zinc fingerproteinD86062_s_atHs.413482NM_004649; chromosome21 open readingframe 33D86479_atHs.439463NM_001129; adipocyteenhancer binding protein1 precursorD86957_atHs.307944D86959_atHs.105751NM_014720; Ste20-related serine/threoninekinaseD86976_atHs.196914D87433_atHs.301989NM_015136; stabilin 1D87443_atHs.409862NM_014758; sortingnexin 19D87682_atHs.134792D89077_atHs.75367NM_006748; Src-like-adaptorD89377_atHs.89404NM_002449; mshhomeo box homolog 2D90279_s_atHs.433695NM_000093; alpha 1type V collagen preproproteinHG1996-HT2044_atHG2090-HT2152_s_atHG2463-HT2559_atHG2994-HT4850_s_atHG3044-HT3742_s_atHG3187-HT3366_s_atHG3342-HT3519_s_atHG371-HT26388_s_atHG4069-HT4339_s_atHG67-HT67_f_atHG907-HT907_atJ02871_s_atHs.436317NM_000779; cytochromeP450, family 4,subfamily B, polypeptide 1J03040_atHs.111779NM_003118; secretedprotein, acidic, cysteine-rich (osteonectin)J03060_atJ03068_atJ03241_s_atHs.2025NM_003239; transforminggrowth factor, beta 3J03278_atHs.307783NM_002609; platelet-derived growth factorreceptor beta precursorJ03909_atJ03925_atHs.172631NM_000632; integrinalpha M precursorJ04056_atHs.88778NM_001757; carbonylreductase 1J04058_atHs.169919NM_000126; electrontransfer flavoprotein,alpha polypeptideJ04093_s_atHs.278896NM_019075; UDPglycosyltransferase 1family, polypeptide A10J04130_s_atHs.75703NM_002984;chemokine (C—C motif)ligand 4 precursorJ04152_rna1_s_atJ04162_atHs.372679NM_000569; Fc fragmentof IgG, low affinityIIIa, receptor for (CD16)J04456_atHs.407909NM_002305; beta-galactosidase bindinglectin precursorJ05032_atHs.32393NM_001349; aspartyl-tRNA synthetaseJ05036_s_atHs.1355NM_001910; cathepsinE isoform a preproproteinNM_148964; cathepsinE isoform bpreproproteinJ05070_atHs.151738NM_004994; matrixmetalloproteinase 9preproproteinJ05448_atHs.79402NM_002694; DNAdirected RNA polymeraseII polypeptide CNM_032940; DNAdirected RNA polymeraseII polypeptide CK01396_atHs.297681NM_000295; serine (orcysteine) proteinaseinhibitor, clade A (alpha-1 antiproteinase,antitrypsin), member 1K03430_atL06797_s_atHs.421986NM_003467;chemokine (C—X—Cmotif) receptor 4L10343_atHs.112341NM_002638; skin-derived protease inhibitor3 preproproteinL11708_atHs.155109NM_002153; hydroxysteroid(17-beta) dehydrogenase 2L13391_atHs.78944NM_002923; regulatorof G-protein signalling2, 24 kDaL13698_atHs.65029NM_002048; growtharrest-specific 1L13720_atHs.437710NM_000820; growtharrest-specific 6L13923_atHs.750NM_000138; fibrillin 1AB000220_atHs.171921NM_006379; semaphorin3CAC002073_cds1_atAF000231_atHs.75618NM_004663; Ras-related protein Rab-11AD10922_s_atHs.99855NM_001462; formylpeptide receptor-like 1D10925_atHs.301921NM_001295;chemokine (C—C motif)receptor 1D11086_atHs.84NM_000206; interleukin2 receptor, gammachain, precursorD11151_atHs.211202NM_001957; endothelinreceptor type AD13435_atHs.426142NM_002643; phosphatidylinositolglycan,class F isoform 1NM_173074; phosphatidylinositolglycan,class F isoform 2D13666_s_atHs.136348NM_006475; osteoblastspecific factor 2 (fasciclinI-like)D14520_atHs.84728NM_001730; Kruppel-like factor 5D21878_atHs.169998NM_004334; bonemarrow stromal cellantigen 1 precursorD26443_atHs.371369NM_004172; solutecarrier family 1 (glialhigh affinity glutamatetransporter), member 3D28589_atHs.17719D42046_atHs.194665D45370_atHs.74120NM_006829; adiposespecific 2D49372_s_atHs.54460NM_002986; smallinducible cytokine A11precursorD50495_atHs.224397NM_003195; transcriptionelongation factor A(SII), 2D63135_atHs.27935NM_032646; tweetyhomolog 2D64053_atHs.198288NM_002849; proteintyrosine phosphatase,receptor type, R isoform1 precursorNM_130846; proteintyrosine phosphatase,receptor type, R isoform 2D83920_atHs.440898NM_002003; ficolin 1precursorD85131_s_atHs.433881NM_002383; MYC-associated zinc fingerproteinD86062_s_atHs.413482NM_004649; chromosome21 open readingframe 33D86479_atHs.439463NM_001129; adipocyteenhancer binding protein1 precursorD86957_atHs.307944D86959_atHs.105751NM_014720; Ste20-related serine/threoninekinaseD86976_atHs.196914D87433_atHs.301989NM_015136; stabilin 1D87443_atHs.409862NM_014758; sortingnexin 19D87682_atHs.134792D89077_atHs.75367NM_006748; Src-like-adaptorD89377_atHs.89404NM_002449; mshhomeo box homolog 2D90279_s_atHs.433695NM_000093; alpha 1type V collagen preproproteinHG1996-HT2044_atHG2090-HT2152_s_atHG2463-HT2559_atHG2994-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, polypeptide 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 sub-




unit, 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, polypeptide 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_ma1_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_ma1_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_ma1_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_ma1_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_ma1_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_ma1_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, polypeptide 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; hemopoletic 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_ma1_s_at


U20158_at
Hs.2488
NM_005565; lymphocyte cytosolic protein 2


U41315_ma1_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 dehydrogenasa 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_ma1_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_ma1_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_ma1_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_ma1_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_ma1_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 16Disease course information of all patients involved.GroupPatientTumour (date)PatternCarcinoma in situTime to recurrenceA968-1Ta gr2Papillaryno27 month A928-1Ta gr2Papillaryno38 month. A934-1Ta gr2 (220798)PapillarynoA709-1Ta gr2 (210798)PapillarynoA930-1Ta gr2 (300698)PapillarynoA524-1Ta gr2 (201095)PapillarynoA455-1Ta gr2 (060695)PapillarynoA370-1Ta gr2 (100195)PapillarynoA810-1Ta gr2 (031097)PapillarynoA1146-1Ta gr2 (231199)PapillarynoA1161-1Ta gr2 (101299)MixednoA1006-1Ta gr2 (231198)PapillarynoA942-1Ta gr2Papillaryno24 month. A1060-1Ta gr2Papillaryno36 month. A1255-1Ta gr2Papillaryno24 month. B441-1Ta gr2Papillaryno6 month.B780-1Ta gr2Papillaryno2 month.B815-2Ta gr2Papillaryno6 month.B829-1Ta gr2Papillaryno4 month.B861-1Ta gr2Papillaryno4 month.B925-1Ta gr2Papillaryno5 month.B1008-1Ta gr2Papillaryno5 month.B1086-1Ta gr2Papillaryno6 month.B1105-1Ta gr2Papillaryno8 month.B1145-1Ta gr2Papillaryno4 month.B1327-1Ta gr2Papillaryno5 month.B1352-1Ta gr2Papillaryno6 month.B1379-1Ta gr2Papillaryno5 month.B533-1Ta gr2Papillaryno4 month.B679-1Ta gr2Papillaryno4 month.B692-1Ta gr2Papillaryno5 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 17Recurrence prediction results of 39 gene cross-validation loops.PredictionGroupPatientTumour (date)PredictionErrorstrengthA968-1Ta gr200.19A928-1Ta gr200.49A934-1Ta gr2 (220798)01.73A709-1Ta gr2 (210798)00.45A930-1Ta gr2 (300698)00.82A524-1Ta gr2 (201095)00.14A455-1Ta gr2 (060695)1*0.68A370-1Ta gr2 (100195)00.32A810-1Ta gr2 (031097)00.45A1146-1Ta gr2 (231199)00.98A1161-1Ta gr2 (101299)00.03A1006-1Ta gr2 (231198)1*1.57A942-1Ta gr200.31A1060-1Ta gr21*0.81A1255-1Ta gr21*0.71B441-1Ta gr211.03B780-1Ta gr210.37B815-2Ta gr210.35B829-1Ta gr210.75B861-1Ta gr20*2.55B925-1Ta gr210.78B1008-1Ta gr20*0.12B1086-1Ta gr20*0.51B1105-1Ta gr210.37B1145-1Ta gr210.44B1327-1Ta gr211.96B1352-1Ta gr20*0.97B1379-1Ta gr210.67B533-1Ta gr210.31B679-1Ta gr210.82B692-1Ta gr210.45
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 18The 26 genes that we find optimal for recurrence prediction.UnigeneFeaturebuild 168DescriptionNumberTest (W-N)AF006041_atHs.336916NM_001350; death-associated protein 6310.054 (161-7)D21337_atHs.408NM_001847; type IV alpha 6 collagen isoform A precursor310.058 (160-6)NM_033641; type IV alpha 6 collagen isoform B precursorD49387_atHs.294584NM_012212; NADP-dependent leukotriene B4 12-310.118 (313-8)hydroxydehydrogenaseD64154_atHs.90107NM_007002; adhesion regulating molecule 1 precursor310.078 (165-9)NM_175573; adhesion regulating molecule 1 precursorD83780_atHs.437991NM_014846; KIAA0196 gene product310.094 (159-4)D87258_atHs.75111NM_002775; protease, serine, 11300.112 (168-11D87437_atHs.43660NM_014837; chromosome 1 open reading frame 16310.058 (160-6)HG1879-HT1919_at310.122 (314-7)HG3076-HT3238_s_at310.080 (309-17HG511-HT511_at310.348 (319-2)L34155_atHs.83450NM_000227; laminin alpha 3 subunit precursor310.122 (314-7)L38928_atHs.118131NM_006441; 5,10-methenyltetrahydrofolate synthetase (5-290.348 (319-2)formyltetrahydrofolate cyclo-ligase)L49169_atHs.75678NM_006732; FBJ murine osteosarcoma viral oncogene310.108 (155-2)homolog BM16938_s_atHs.820NM_004503; homeo box C6 isoform 1 NM_153693; homeo29 0.09 (170-16)box C6 isoform 2M63175_atHs.295137NM_001144; autocrine motility factor receptor isoform a290.098 (308-18NM_138958; autocrine motility factor receptor isoform bM64572_atHs.405666NM_002829; protein tyrosine phosphatase, non-receptor310.064 (305-31type 3M98528_atHs.79404NM_014392; DNA segment on chromosome 4 (unique)310.122 (314-7)234 expressed sequenceU21858_atHs.60679NM_003187; TBP-associated factor 9 NM_016283; adrenal310.122 (314-7)gland protein AD-004U45973_atHs.178347NM_016532; skeletal muscle and kidney enriched inositol310.094 (310-14phosphatase isoform 1 NM_130766; skeletal muscle andkidney enriched inositol phosphatase isoform 2U58516_atHs.3745NM_005928; milk fat globule-EGF factor 8 protein290.100 (175-28U62015_atHs.8867NM_001554; cysteine-rich, angiogenic inducer, 61310.106 (169-13U66702_atHs.74624NM_002847; protein tyrosine phosphatase, receptor type,310.146 (149-1)N polypeptide 2 isoform 1 precursor NM_130842; proteintyrosine phosphatase, receptor type, N polypeptide 2isoform 2 precursor NM_130843; protein tyrosine phosphatase,receptor type, N polypeptide 2 isoform 3 precursorU70439_s_atHs.84264NM_006401; acidic (leucine-rich) nuclear phosphoprotein30 0.08 (309-17)32 family, member BU94855_atHs.381255NM_003754; eukaryotic translation initiation factor 3,300.092 (311-12)subunit 5 epsilon, 47 kDaX63469_atHs.77100NM_002095; general transcription factor IIE, polypeptide310.092 (311-12)2, beta 34 kDaZ23064_atHs.380118NM_002139; RNA binding motif protein, X chromosome300.066 (307-24)


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, Ore.) 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, Ore.) 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 duster 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 transformed 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 resealed. 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/gc2.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 biopsies and 5 biopsies with CIS).

TABLE 19Clinical data on patient disease courses and results of molecular CIS classificationSamplePreviousTumourSubsequentgroupaPatientbtumoursanalysedtumoursCIScCIS classifierd11060-1Ta gr22 TaNoNo CIS11146-1Ta gr2NoNo CIS11216-1Ta gr2NoNo CIS11303-1Ta gr2NoNo CIS1524-1Ta gr2NoNo CIS1692-1Ta gr22 TaNoNo CIS11264-1Ta gr320 TaNoNo CIS11350-1Ta gr31 TaNoNo CIS11354-1Ta gr311 T1NoNo CIS1775-1Ta gr31 TaNoNo CIS11066-1Ta gr31 TaNoNo CIS11276-1Ta gr32 T1NoNo CIS11070-1Ta gr31 TaNoNo CIS1989-1Ta gr3NoNo CIS11482-1Ta gr320 TaNoCIS21345-21 T1Ta gr3Sampling visitCIS21062-2Ta gr31 T1Sampling visitCIS2956-2Ta gr31 TaSampling visitCIS2320-71 Ta, 2 T1Ta gr32 TaSampling visitCIS21330-1Ta gr3Sampling visitCIS2602-85 TaTa gr33 TaSampling visitCIS2763-1Ta gr214 TaSampling visitCIS21024-1T1 gr32 Ta, 1 T1Sampling visitCIS21182-1Ta gr37 TaSubsequent visitCIS21093-1Ta gr34 Ta, 1 T1Subsequent visitCIS2979-1Ta gr3Sampling visitCIS21337-1T1 gr3Sampling visitCIS21625-1Ta gr2Sampling visitCIS31015-1T3b gr4No31337-1T4a gr3Sampling visit31041-1T4b gr3No31044-1T4b gr3ND31055-11 Ta gr2T3a gr3No31109-1T2 gr31 T2-4No31124-1T4a gr32 T2-4No31154-1T3a gr31 Ta, 1 T2-4No31167-11 T2-4T3b gr42 T2-4ND31178-1T4b gr3ND31215-1T4b gr3ND31271-1T3b gr4No31321-11 T1T3b 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 duster 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 20The best 100 markersFeaturePermPermPermUniGene(U133 array)ClassT-test1%5%10%Build 162RefSeq; description221204_s_atno_CIS3.745.124.614.33Hs.326444NM_018058; cartilage acidicprotein 1205927_s_atno_CIS3.674.533.983.73Hs.1355NM_001910; cathepsin E isoforma preproproteinNM_148964; cathepsin E isoformb preproprotein210143_atno_CIS3.354.033.733.45Hs.188401NM_007193; annexin A10204540_atno_CIS3.153.873.513.32Hs.433839NM_001958; eukaryotic translationelongation factor 1 alpha 2214599_atno_CIS3.023.753.373.14Hs.157091NM_005547; involucrin203649_s_atno_CIS2.843.633.203.00Hs.76422NM_000300; phospholipase A2,group IIA (platelets, synovialfluid)203980_atno_CIS2.743.473.122.89Hs.391561NM_001442; fatty acid bindingprotein 4, adipocyte209270_atno_CIS2.393.383.102.85Hs.436983NM_000228; laminin subunitbeta 3 precursor206658_atno_CIS2.353.373.052.78Hs.284211NM_030570; uroplakin 3B isoforma NM_182683; uroplakin3B isoform c NM_182684; uroplakin3B isoform b220779_atno_CIS2.353.332.972.73Hs.149195NM_016233; peptidylargininedeiminase type III216971_s_atno_CIS2.283.292.912.71Hs.79706NM_000445; plectin 1, intermediatefilament binding protein500 kDa206191_atno_CIS2.253.242.862.68Hs.47042NM_001248; ectonucleosidetriphosphate diphosphohydrolase 3218484_atno_CIS2.183.202.812.62Hs.221447NM_020142; NADH: ubiquinoneoxidoreductase MLRQ subunithomolog221854_atno_CIS2.13.192.802.60Hs.313068NM_000299; plakophllin 1203792_x_atno_CIS2.023.162.742.55Hs.371617NM_007144; ring finger protein110207862_atno_CIS2.013.162.722.52Hs.379613NM_006760; uroplakin 2218960_atno_CIS1.933.142.652.47Hs.414005NM_019894; transmembraneprotease, serine 4 isoform 1NM_183247; transmembraneprotease, serine 4 isoform 2203009_atno_CIS1.933.122.622.45Hs.155048NM_005581; Lutheran bloodgroup (Auberger b antigenincluded)204508_s_atno_CIS1.883.102.602.42Hs.279916NM_017689; hypothetical proteinFLJ20151211692_s_atno_CIS1.873.062.582.39Hs.87246NM_014417; BCL2 bindingcomponent 3206465_atno_CIS1.863.042.542.38Hs.277543NM_015162; lipidosin206122_atno_CIS1.852.922.522.36Hs.95582NM_006942; SRY-box 15206393_atno_CIS1.832.892.492.33Hs.83760NM_003282; troponin I, skeletal,fast214639_s_atno_CIS1.792.872.492.30Hs.67397NM_005522; homeobox A1protein isoform a NM_153820;homeobox A1 protein isoform, b214630_atno_CIS1.792.842.442.28Hs.184927NM_000497; cytochrome P450,subfamily XIB (steroid 11-beta-hydroxylase), polypeptide 1precursor204465_s_atno_CIS1.772.812.422.27Hs.76888NM_004692; NM_032727;internexin neuronal intermediatefilament protein, alpha204990_s_atno_CIS1.762.792.412.24Hs.85266NM_000213; integrin, beta 4205453_atno_CIS1.752.772.392.22Hs.290432NM_002145; homeo box B2215812_s_atno_CIS1.742.772.372.20Hs.499113NM_018058; cartilage acidicprotein 1217040_x_atno_CIS1.742.752.362.18Hs.95582NM_001910; cathepsin E isoforma preproproteinNM_148964; cathepsin E isoformb preproprotein203759_atno_CIS1.732.752.342.17Hs.75268NM_007193; annexin A10211002_s_atno_CIS1.732.742.332.17Hs.82237NM_001958; eukaryotic translationelongation factor 1 alpha 2216641_s_atno_CIS1.732.732.312.15Hs.18141NM_005547; involucrin221660_atno_CIS1.712.672.302.13Hs.247831NM_000300; phospholipase A2,group IIA (platelets, synovialfluid)220026_atno_CIS1.712.662.282.13Hs.227059NM_001442; fatty acid bindingprotein 4, adipocyte209591_s_atno_CIS1.692.632.282.11Hs.170195NM_000228; laminin subunitbeta 3 precursor219922_s_atno_CIS1.682.612.262.08Hs.289019NM_030570; uroplakin 3B isoforma NM_182683; uroplakin3B isoform c NM_182684; uroplakin38 isoform b201641_atno_CIS1.672.612.262.07Hs.118110NM_016233; peptidylargininedeiminase type III204952_atno_CIS1.662.592.242.07Hs.377028NM_000445; plectin 1, intermediatefilament binding protein500 kDa204487_s_atno_CIS1.652.592.232.06Hs.367809NM_001248; ectonucleosidetriphosphate diphosphohydrolase 3210761_s_atno_CIS1.642.592.232.05Hs.86859NM_020142; NADH: ubiquinoneoxidoreductase MLRQ subunithomolog217626_atno_CIS1.632.582.212.04Hs.201967NM_000299; plakophilin 1204380_s_atno_CIS1.622.582.192.03Hs.1420NM_007144; ring finger protein110205455_atno_CIS1.612.582.172.02Hs.2942NM_006760; uroplakin 2205073_atno_CIS1.612.582.172.01Hs.152096NM_019894; transmembraneprotease, serine 4 isoform 1NM_183247; transmembraneprotease, serine 4 isoform 2203287_atno_CIS1.612.582.162.00Hs.18141NM_005581; Lutheran bloodgroup (Auberger b antigenincluded)210735_s_atno_CIS1.582.552.151.99Hs.5338NM_017689; hypothetical proteinFLJ20151203842_s_atno_CIS1.572.542.151.97Hs.172740NM_014417; BCL2 bindingcomponent 3206561_s_atno_CIS1.572.532.141.96Hs.116724NM_015162; lipidosin214752_x_atno_CIS1.562.522.131.95Hs.195464NM_006942; SRY-box 15217028_atCIS4.875.174.674.40Hs.421986NM_003282; troponin I, skeletal,fast213975_s_atCIS4.654.434.013.76Hs.234734NM_005522; homeobox A1protein isoform a NM_153620;homeobox A1 protein isoform b201859_atCIS4.594.153.703.45Hs.1908NM_000497; cytochrome P450,subfamily XIB (steroid 11-beta-hydroxylase), polypeptide 1precursor219410_atCIS4.493.983.493.29Hs.104800NM_004692; NM_032727;internexin neuronal intermediatefilament protein, alpha207173_x_atCIS4.373.883.333.11Hs.443435NM_000213; integrin, beta 4214651_s_atCIS4.143.833.222.99Hs.127428NM_002145; homeo box B2201858_s_atCIS4.063.783.092.91Hs.1908NM_018058; cartilage acidicprotein 1211430_s_atCIS4.033.633.052.83Hs.413826NM_001910 cathepsin E isoforma preproproteinNM_148964; cathepsin E isoformb preproprotein213891_s_atCIS3.863.633.022.77Hs.359289NM_007193; annexin A10221872_atCIS3.823.522.892.73Hs.82547NM_001958; eukaryotic translationelongation factor 1 alpha 2212386_atCIS3.773.502.872.69Hs.359289NM_005547; involucrin211161_s_atCIS3.763.422.842.65NM_000300; phospholipase A2,group IIA (platelets, synovialfluid)214669_x_atCIS3.553.362.802.62Hs.377975NM_001442; fatty acid bindingprotein 4, adipocyte217388_s_atCIS3.443.312.792.58Hs.444471NM_000228; laminin subunitbeta 3 precursor203477_atCIS3.363.282.752.56Hs.409034NM_030570; uroplakin 3B isoforma NM_182683; uroplakin3B isoform c NM_182684; uroplakin3B isoform b204688_atCIS3.353.262.742.52Hs.409798NM_016233; peptidylargininedeiminase type III218718_atCIS3.353.222.702.48Hs.43080NM_000445; plectin 1, intermediatefilament binding protein500 kDa215176_x_atCIS3.323.142.672.45Hs.503443NM_001248; ectonucleosidetriphosphate diphosphohydrolase 3201842_s_atCIS3.313.112.652.44Hs.76224NM_020142; NADH: ubiquinoneoxidoreductase MLRQ subunithomolog212667_atCIS3.33.112.632.42Hs.111779NM_000299; plakophilin 1209340_atCIS3.273.102.612.39Hs.21293NM_007144; ring finger protein110215379_x_atCIS3.263.102.592.39Hs.449601NM_006760; uroplakin 2200762_atCIS3.253.052.562.34Hs.173381NM_019894; transmembraneprotease, serine 4 isoform 1NM_183247; transmembraneprotease, serine 4 isoform 2211896_s_atCIS3.213.052.532.32Hs.156316NM_005581; Lutheran bloodgroup (Auberger b antigenincluded)204141_atCIS3.193.052.532.28Hs.300701NM_017689; hypothetical proteinFLJ20151201744_s_atCIS3.183.032.502.27Hs.406475NM_014417; BCL2 bindingcomponent 3209138_x_atCIS3.173.032.472.24Hs.505407NM_015162; lipidosin214677_x_atCIS3.143.022.472.23Hs.449601NM_006942; SRY-box 15212077_atCIS3.112.992.462.21Hs.443811NM_003282; troponin I, skeletal,fast206392_s_atCIS3.112.982.432.20Hs.82547NM_005522; homeobox A1protein isoform a NM_153620;homeobox A1 protein isoform b212998_x_atCIS3.092.942.402.19Hs.375115NM_000497; cytochrome P450,subfamily XIB (steroid 11-beta-hydroxylase), polypeptide 1precursor201616_s_atCIS3.082.932.382.18Hs.443811NM_004692; NM_032727;internexin neuronal intermediatefilament protein, alpha205382_s_atCIS3.072.882.372.15Hs.155597NM_000213; integrin, beta 4212671_s_atCIS3.072.852.352.14Hs.387679NM_002145; homeo box B2215121_x_atCIS3.062.842.342.13Hs.356861NM_018058; cartilage acidicprotein 1200600_atCIS3.052.832.332.11Hs.170328NM_001910; cathepsin E isoforma preproproteinNM_148964; cathepsin E isoformb preproprotein202746_atCIS3.032.802.322.10Hs.17109NM_007193; annexin A10202917_s_atCIS32.792.312.08Hs.416073NM_001958; eukaryotic translationelongation factor 1 alpha 2201560_atCIS32.792.302.08Hs.25035NM_005547; involucrin218918_atCIS2.992.772.292.06Hs.8910NM_000300; phospholipase A2,group IIA (platelets, synovialfluid)218656_s_atCIS2.992.762.272.06Hs.93765NM_001442; fatty acid bindingprotein 4, adipocyte201088_atCIS2.992.762.262.04Hs.159557NM_000228; laminin subunitbeta 3 precursor201291_s_atCIS2.972.752.252.04Hs.156346NM_030570; uroplakin 3B isoforma NM_182683; uroplakin3B isoform c NM_182684; uroplakin3B isoform b215076_s_atCIS2.952.722.242.03Hs.443625NM_016233; peptidylargininedeiminase type III212195_atCIS2.942.712.222.02Hs.71968NM_000445; plectin 1, intermediatefilament binding protein500 kDa209732_atCIS2.942.682.222.00Hs.85201NM_001248; ectonucleosidetriphosphate diphosphohydrolase 3212192_atCIS2.942.672.221.99Hs.109438NM_020142; NADH: ubiquinoneoxidoreductase MLRQ subunithomolog221671_x_atCIS2.922.672.201.98Hs.377975NM_000299; plakophilin 1211671_s_atCIS2.912.662.201.98Hs.126608NM_007144; ring finger protein110214352_s_atCIS2.882.662.191.97Hs.412107NM_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 showed that 13 of these were significant at a 1% confidence level—the remaining three genes were above a 10% confidence level.

TABLE 21The 16 gene molecular classifier of CISFeature(U133aPermPermPermUniGenearray)Classt-test1%5%10%Build 162RefSeq; description213633_atno_CIS1.512.462.041.85Hs.97858NM_018957; SH3-domainbinding protein 1212784_atno_CIS1.362.271.861.70Hs.388236NM_015125; capicuahomolog209241_x_atno_CIS1.131.781.481.33Hs.112028NM_015716; mis-shapen/NIK-related kinaseisoform 1NM_153827; mis-shapen/NIK-related kinaseisoform 3NM_170663; mis-shapen/NIK-related kinaseisoform 2217941_s_atCIS2.31.961.661.47Hs.8117NM_018695; erbb2 interactingprotein201877_s_atCIS2.271.901.621.45Hs.249955NM_002719; gammaisoform of regulatorysubunit B56, proteinphosphatase 2A isoform aNM_178586; gammaisoform, of regulatorysubunit B56, proteinphosphatase 2A isoform bNM_178587; gammaisoform of regulatorysubunit B56, proteinphosphatase 2A isoform cNM_178588; gammaisoform of regulatorysubunit B56, proteinphosphatase 2A isoform d209630_s_atCIS1.971.541.311.15Hs.444354NM_012164; F-box andWD-40 domain protein 2202777_atCIS1.931.511.291.12Hs.104315NM_007373; soc-2 suppressorof clear homolog200958_s_atCIS1.921.491.281.11Hs.164067NM_005625; syndecanbinding protein (syntenin)209579_s_atCIS1.791.361.161.01Hs.35947NM_003925; methyl-CpGbinding domain protein 4209004_s_atCIS1.631.211.000.89Hs.5548NM_012161; F-box andleucine-rich repeat protein5 isoform 1 NM_033535;F-box and leucine-richrepeat protein 5 isoform 2218150_atCIS1.61.180.980.86Hs.342849NM_012097; ADP-ribosylation factor-like 5isoform 1 NM_177985;ADP-ribosylation factor-like 5 isoform 2202076_atCIS1.531.120.920.82Hs.289107NM_001166; baculoviralIAP repeat-containingprotein 2204640_s_atCIS1.451.030.830.75Hs.129951NM_003563; speckle-typePOZ protein201887_atCIS1.320.920.740.66Hs.285115NM_001560; interleukin13 receptor, alpha 1precursor212802_s_atCIS1.310.910.720.65Hs.287266212899_atCIS1.290.890.710.64Hs.129836NM_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 5, 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 group 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 of 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), 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 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), 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.
  • 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. 563 correlating 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, and correlating 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, 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), 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
PA 2002 01685 Nov 2002 DK national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/DK03/00750 11/3/2003 WO 11/16/2005