Differentially methylated sequences in pancreatic cancer

Goggins, Michael G. ;   et al.

Patent Application Summary

U.S. patent application number 10/889898 was filed with the patent office on 2005-03-31 for differentially methylated sequences in pancreatic cancer. This patent application is currently assigned to THE JOHNS HOPKINS UNIVERSITY SCHOOL OF MEDICINE. Invention is credited to Goggins, Michael G., Ueki, Takashi.

Application Number20050069924 10/889898
Document ID /
Family ID23034863
Filed Date2005-03-31

United States Patent Application 20050069924
Kind Code A1
Goggins, Michael G. ;   et al. March 31, 2005

Differentially methylated sequences in pancreatic cancer

Abstract

The present invention provides a method for detecting a cellular proliferative disorder in a subject. The method includes contacting a nucleic acid-containing specimen from the subject with an agent that provides a determination of the methylation state of at least one gene or associated regulatory region of the gene and identifying aberrant methylation of regions of the gene or regulatory region, wherein aberrant methylation is identified as being different when compared to the same regions of the gene or associated regulatory region in a subject not having said cellular proliferative, thereby detecting a cellular proliferative disorder in the subject.


Inventors: Goggins, Michael G.; (Baltimore, MD) ; Ueki, Takashi; (Sparks, MD)
Correspondence Address:
    Lisa A. Haile, J.D., Ph.D.
    GRAY CARY WARE & FREIDENRICH LLP
    Suite 1100
    4365 Executive Drive
    San Diego
    CA
    92121-2133
    US
Assignee: THE JOHNS HOPKINS UNIVERSITY SCHOOL OF MEDICINE

Family ID: 23034863
Appl. No.: 10/889898
Filed: July 8, 2004

Related U.S. Patent Documents

Application Number Filing Date Patent Number
10889898 Jul 8, 2004
10084555 Feb 25, 2002
60271268 Feb 23, 2001

Current U.S. Class: 435/6.12 ; 435/199; 435/320.1; 435/325; 435/69.1; 536/23.2
Current CPC Class: C12Q 2600/156 20130101; C12Q 1/6886 20130101
Class at Publication: 435/006 ; 435/069.1; 435/199; 435/320.1; 435/325; 536/023.2
International Class: C12Q 001/68; C07H 021/04; C07K 014/47; C12N 009/22

Claims



What is claimed is:

1. An isolated nucleic acid molecule selected from SEQ ID NO:1-42, and regulatory sequences associated therewith.

2. The nucleic acid molecule of claim 1, wherein said associated regulatory sequences contain CpG-rich regions.

3. The nucleic acid molecule of claim 2, wherein the state of methylation of the CpG-rich regions is determinative of the presence of a cellular proliferative disorder in a subject from which the nucleic acid molecule is isolated.

4. The nucleic acid molecule of claim 2, wherein hypermethylation of said CpG islands is indicative of the presence of a cellular proliferative disorder in a subject from which said nucleic acid is isolated.

5. The nucleic acid molecule of claim 1, wherein the molecule is selected from SEQ ID NO:39-42.

6. A substantially purified polypeptide encoded by a polynucleotide selected from SEQ ID NO:39-42.

7. A method for detecting a cellular proliferative disorder in a subject comprising: a) contacting a nucleic acid-containing specimen from the subject with an agent that provides a determination of the methylation state of at least one gene or associated regulatory region of the gene selected from MICP 1-42 of Table 1 and combinations thereof; and b) identifying aberrant methylation of regions of the gene or regulatory region, wherein aberrant methylation is identified as being different when compared to the same regions of the gene or associated regulatory region in a subject not having said cellular proliferative, thereby detecting a cellular proliferative disorder in the subject.

8. The method of claim 7, wherein the regions of said gene are contained within CpG rich regions.

9. The method of claim 7, wherein the gene is SEQ ID NO:39, 40, 41 or 42.

10. The method of claim 7, wherein aberrant methylation comprises hypermethylation when compared to the same regions of the gene or associated regulatory regions in a subject not having the cellular proliferative disorder.

11. The method of claim 10, wherein the regions comprise regulatory regions of the gene.

12. The method of claim 7, wherein the agent is a pair of primers that hybridize with a target sequence in the gene or associated regulatory region of the gene.

13. The method of claim 7, wherein the nucleic acid-containing specimen comprises a tissue selected from the group consisting of brain, colon, urogenital, lung, renal, prostate, pancreas, liver, esophagus, stomach, hematopoietic, breast, thymus, testis, ovarian, and uterine.

14. The method of claim 7, wherein the nucleic acid-containing specimen is selected from the group consisting of serum, urine, saliva, blood, duodenal fluid, pancreatic fluid, cerebrospinal fluid, pleural fluid, ascites fluid, sputum, stool, and biopsy sample.

15. The method of claim 11, wherein said cellular proliferative disorder is selected from the group consisting of low grade astrocytoma, anaplastic astrocytoma, glioblastoma, medulloblastoma, gastric cancer, colorectal cancer, colorectal adenoma, acute myelogenous leukemia, lung cancer, renal cancer, leukemia, breast cancer, prostate cancer, endometrial cancer and neuroblastoma.

16. A kit useful for the detection of a cellular proliferative disorder in a subject comprising: a) carrier means compartmentalized to receive a sample therein; b) one or more containers comprising a first container containing a reagent which modifies unmethylated cytosine and a second container containing primers for amplification of a CpG-containing nucleic acid, wherein the primer hybridizes with a target polynucleotide sequence having the sequence selected from SEQ ID NO:1-42.

17. The kit of claim 16, further comprising a third container containing a methylation sensitive restriction endonuclease.

18. The kit of claim 16, wherein said modifying reagent is bisulfite.

19. The kit of claim 16, wherein the primer hybridizes with a target polynucleotide sequence having a sequence as set forth in SEQ ID NO:39, 40, 41 or 42.

20. Isolated oligonucleotide primer(s) for detection of a methylated CpG-containing nucleic acid wherein the primer hybridizes with a target polynucleotide sequence having the sequence selected from the group consisting of SEQ ID NO:1-42.

21. Isolated oligonucleotide primer pairs selected from SEQ ID NO:43-105.
Description



RELATED APPLICATIONS

[0001] This application claims priority under 35 U.S.C. .sctn.119(e) to U.S. Provisional Application Ser. No. 60/271,268, filed Feb. 23, 2001, which is herein incorporated by reference in its entirety.

FIELD OF THE INVENTION

[0002] The present invention relates generally to the regulation of gene expression and more specifically to a method of determining the DNA methylation status of CpG sites in a given locus and correlating the methylation status with the presence of a cell proliferative disorder.

BACKGROUND OF THE INVENTION

[0003] DNA methylases transfer methyl groups from the universal methyl donor S-adenosyl methionine to specific sites on the DNA. Several biological functions have been attributed to the methylated bases in DNA. The most established biological function for methylated DNA is the protection of DNA from digestion by cognate restriction enzymes. The restriction modification phenomenon has, so far, been observed only in bacteria. Mammalian cells, however, possess a different methylase that exclusively methylates cytosine residues that are 5' neighbors of guanine (CpG). This modification of cytosine residues has important regulatory effects on gene expression, especially when involving CpG rich areas, known as CpG islands, located in the promoter regions of many genes.

[0004] Methylation has been shown by several lines of evidence to play a role in gene activity, cell differentiation, tumorigenesis, X-chromosome inactivation, genomic imprinting and other major biological processes (Razin, A., H., and Riggs, R. D. eds. in DNA Methylation Biochemistry and Biological Significance, Springer-Verlag, New York, 1984). In eukaryotic cells, methylation of cytosine residues that are immediately 5' to a guanosine, occurs predominantly in CG poor regions (Bird, A., Nature, 321:209, 1986). In contrast, CpG islands remain unmethylated in normal cells, except during X-chromosome inactivation (Migeon, et al., supra) and parental specific imprinting (Li, et al., Nature, 366:362, 1993) where methylation of 5' regulatory regions can lead to transcriptional repression. De novo methylation of the Rb gene has been demonstrated in a small fraction of retinoblastomas (Sakai, et al., Am. J. Hum. Genet., 48:880, 1991), and recently, a more detailed analysis of the VHL gene showed aberrant methylation in a subset of sporadic renal cell carcinomas (Herman, et al., Proc. Natl. Acad. Sci., U.S.A., 91:9700, 1994). Expression of a tumor suppressor gene can also be abolished by de novo DNA methylation of a normally unmethylated CpG island (Issa, et al., Nature Genet., 7:536, 1994; Herman, et al., supra; Merlo, et al., Nature Med., 1:686, 1995; Herman, et al., Cancer Res., 56:722, 1996; Graff, et al., Cancer Res., 55:5195, 1995; Herman, et al., Cancer Res., 55:4525, 1995).

[0005] Human cancer cells typically contain somatically altered nucleic acid, characterized by mutation, amplification, or deletion of critical genes. In addition, the nucleic acid from human cancer cells often displays somatic changes in DNA methylation (E. R. Fearon, et al., Cell, 61:759, 1990; P. A. Jones, et al., Cancer Res., 46:461, 1986; R. Holliday, Science, 238:163, 1987; A. De Bustros, et al., Proc. Natl. Acad. Sci., USA, 85:5693, 1988); P. A. Jones, et al., Adv. Cancer Res., 54:1, 1990; S. B. Baylin, et al., Cancer Cells, 3:383, 1991; M. Makos, et al., Proc. Natl. Acad. Sci., USA, 89:1929, 1992; N. Ohtani-Fujita, et al., Oncogene, 8:1063, 1993). However, the precise role of abnormal DNA methylation in human tumorigenesis has not been established. Aberrant methylation of normally unmethylated CpG islands has been described as a frequent event in immortalized and transformed cells, and has been associated with transcriptional inactivation of defined tumor suppressor genes in human cancers. In the development of colorectal cancers (CRC), a series of tumor suppressor genes (TSG) such as APC, p53, DCC and DPC4 are inactivated by mutations and chromosomal deletions. Some of these alterations result from a chromosomal instability phenotype described in a subset of CRC. Recently, an additional pathway has been shown to be involved in a familial form of CRC, hereditary non-polyposis colorectal cancer. The cancers from these patients show a characteristic mutator phenotype which causes microsatellite instability (MI), and mutations at other gene loci such as TGF-.beta.-RII (Markowitz et al., Science, 268(5215): 1336-8, 1995) and BAX. This phenotype usually results from mutations in the mismatch repair (MMR) genes hMSH2 and hMLH1. A subset of sporadic CRC also show MI, but mutations in MMR genes appear to be less frequent in these tumors.

[0006] Another molecular defect described in CRC is CpG island (CGI) methylation. CGIs are short sequences rich in the CpG dinucleotide and can be found in the 5' region of about half of all human genes. Methylation of cytosine within 5' CGIs is associated with loss of gene expression and has been seen in physiological conditions such as X chromosome inactivation and genomic imprinting. Aberrant methylation of CGIs has been detected in genetic diseases such as the fragile-X syndrome, in aging cells and in neoplasia. About half of the tumor suppressor genes which have been shown to be mutated in the germline of patients with familial cancer syndromes have also been shown to be aberrantly methylated in some proportion of sporadic cancers, including Rb, VHL, p16, hMLH1, and BRCA1. TSG methylation in cancer is usually associated with (Antequera, et al., Proc. Natl. Acad. Sci. USA, 90:11995-11999, 1993) lack of gene transcription and (Baylin, et al., Adv. Cancer Res., 72:141-196, 1998) absence of coding region mutation. Thus it has been proposed that CGI methylation serves as an alternative mechanism of gene inactivation in cancer.

[0007] The causes and global patterns of CGI methylation in human cancers remain poorly defined. Aging could play a factor in this process because methylation of several CGIs could be detected in an age-related manner in normal colon mucosa as well as in CRC. In addition, aberrant methylation of CGIs has been associated with the MI phenotype in CRC as well as specific carcinogen exposures. However, an understanding of aberrant methylation in CRC has been somewhat limited by the small number of CGIs analyzed to date. In fact, previous studies have suggested that large numbers of CGIs are methylated in immortalized cell lines, and it is not well understood whether this global aberrant methylation is caused by the cell culture conditions or whether they are an integral part of the pathogenesis of cancer.

[0008] Most of the methods developed to date for detection of methylated cytosine depend upon cleavage of the phosphodiester bond alongside cytosine residues, using either methylation-sensitive restriction enzymes or reactive chemicals such as hydrazine which differentiate between cytosine and its 5-methyl derivative. Genomic sequencing protocols which identify a 5-MeC residue in genomic DNA as a site that is not cleaved by any of the Maxam Gilbert sequencing reactions have also been used, but still suffer disadvantages such as the requirement for large amount of genomic DNA and the difficulty in detecting a gap in a sequencing ladder which may contain bands of varying intensity.

[0009] Mapping of methylated regions in DNA has relied primarily on Southern hybridization approaches, based on the inability of methylation-sensitive restriction enzymes to cleave sequences which contain one or more methylated CpG sites. This method provides an assessment of the overall methylation status of CpG islands, including some quantitative analysis, but is relatively insensitive and requires large amounts of high molecular weight DNA.

[0010] Another method utilizes bisulfite treatment of DNA to convert all unmethylated cytosines to uracil. The altered DNA is amplified and sequenced to show the methylation status of all CpG sites. However, this method is technically difficult, labor intensive and without cloning amplified products, it is less sensitive than Southern analysis, requiring approximately 10% of the alleles to be methylated for detection.

[0011] Identification of the earliest genetic changes in tumorigenesis is a major focus in molecular cancer research. Diagnostic approaches based on identification of these changes are likely to allow implementation of early detection strategies and novel therapeutic approaches targeting these early changes might lead to more effective cancer treatment.

[0012] About half of all human genes have 5'CpG islands and these islands are usually associated with the 5' regulatory regions of genes (Antequera, et al., Proc. Natl. Acad. Sci. USA 90:11995-11999, 1993). The 5' CpG islands of most nonimprinted genes are thought to remain unmethylated in normal cells but may become methylated during aging or tumorigenesis. Through interactions between methyl CpG binding proteins, histones and histone deacetylase, 5' CpG island methylation can contribute to changes in chromatin that cause transcriptional silencing (Baylin, et al., Adv. Cancer Res. 72:141-196, 1998). Promoter methylation is implicated in the transcriptional silencing of tumor suppressor and mismatch repair genes (e.g. p16, Rb, VHL, hMLH1) in many cancers. Although 13 hypermethylated genes and clones in pancreatic cancers were previously identified (Ueki, et al., Cancer Res. 60:1835-1839, 2000), there almost certainly are others. Costello et al. have estimated that .about.400 genes are aberrantly methylated in cancers and have found evidence for tumor-specific pattern of methylation (Costello, et al., Nat. Genet. 24:132-138, 2000). A better description of the pattern of DNA methylation abnormalities in cancer may improve an understanding of the role of DNA methylation in tumorigenesis and identification of differentially methylated CpG islands in cancer may lead to the discovery of novel genes with tumor suppressor properties. Finally, identified genes or loci could be utilized as cancer-specific markers for the early detection of cancer (Belinsky, et al., Proc. Natl. Acad. Sci. USA 95:11891-11896, 1998).

[0013] Pancreatic cancer is the fourth leading cause of cancer death in men and in women and each year .about.28,000 Americans die of the disease (Greenlee, et al., CA Cancer J. Clin. 50:7-33, 2000). Frequent genetic changes such as mutational activation of the K-ras oncogene and inactivation of the p16, DPC4, p53, MKK4, STK11, TGFBR2, and TGFBR1 tumor suppressor genes have been described in pancreatic cancer (Goggins, et al., Ann. Oncol. 10:4-8, 1999, Rozenblum, et al., Cancer Res. 57:1731-1734, 1997). Although multiple tumor suppressor pathways have been shown to play a role in pancreatic carcinogenesis, little is known about the contribution of DNA methylation to inactivation of genes in these pathways. Recently, a novel technique, methylated CpG island amplification (MCA), was developed to enrich for methylated CpG rich sequences. MCA coupled with RDA (MCA/RDA) can recover CpG islands differentially methylated in cancer cells (Toyota, et al., Cancer Res. 59:2307-2312, 1997).

SUMMARY OF THE INVENTION

[0014] The present invention is based on the finding that several genes are newly identified as being differentially methylated in cancer. This seminal discovery is useful for cancer screening, risk-assessment, prognosis, minimal-residual disease identification, staging and identification of therapeutic targets. The identification of new genes that are methylated in cancer, aging or diseases associated with aging increases the likelihood of finding genes methylated in a particular cancer; increases the sensitivity and specificity of methylation detection; allows methylation profiling using multiple genes; and allows identification of new targets for therapeutic intervention. The invention also provides a newly identified gene that is a target for hypermethylation in human tumors.

[0015] In one embodiment, there are provided methods for detecting a cellular proliferative disorder in a subject. The subject may have or be at risk of having a cellular proliferative disorder. The method of the invention is useful for diagnostic as well as prognostic analyses. One method for detecting a cellular proliferative disorder in a subject includes contacting a nucleic acid-containing specimen from the subject with an agent that provides a determination of the methylation state of at least one gene or associated regulatory region of the gene; and identifying aberrant methylation of regions of the gene or regulatory region, wherein aberrant methylation is identified as being different when compared to the same regions of the gene or associated regulatory region in a subject not having the cellular proliferative, thereby detecting a cellular proliferative disorder in the subject. The method includes multiplexing by utilizing a combination of primers for more than one loci, thereby providing a methylation "profile" for more than one gene or regulatory region.

[0016] For the first time, the invention provides methylated forms of genes and/or their associated regulatory sequences referred to herein as MICP1-42. MICP39-42 have no homology to known human sequences. Eleven clones matched human genes (MICP1-11); 10 clones matched human ESTs (MICP12-21); 5 clones matched human CpG islands (MICP22-26); and 12 clones matched human genome sequences (MICP27-38). (see Table 1 as reference).

[0017] Invention methods include determining, in a nucleic acid-containing specimen taken from a subject, the methylation state of a gene or regulatory sequences associated therewith, wherein the expression or non-expression of the gene is associated with the presence of the cellular proliferative disorder, and identifying as having a cellular proliferative disorder a subject that has aberrant methylation of regions of the gene or associated regulatory sequences when compared to the same regions of the gene in a subject not having the cellular proliferative disorder. In one aspect of this embodiment, the methylated regions of the gene and associated regulatory sequences are contained within CpG islands (i.e., CpG rich regions). In particular, the aberrant methylation typically includes hypermethylation as compared with the same regions of the gene or regulatory sequences in a subject not having the cellular proliferative disorder.

[0018] Determining the methylation state of the gene includes contacting the nucleic acid-containing specimen with an agent that modifies unmethylated cytosine, amplifying a CpG-containing nucleic acid in the specimen by means of CpG-specific oligonucleotide primers, wherein the oligonucleotide primers distinguish between modified methylated and nonmethylated nucleic acid, and detecting the methylated nucleic acid based on the presence-or absence of amplification products produced in said amplifying step. The method includes optionally contacting the amplification products with a methylation sensitive restriction endonuclease. Other methods for determining methylation status of a gene and/or regulatory sequences are well known in the art and are described more fully herein.

[0019] In another embodiment of the present invention there is provided a kit useful for the detection of a cellular proliferative disorder in a subject having or at risk for having a cellular proliferative disorder. Invention kits include a carrier means compartmentalized to receive a sample, one or more containers comprising a first container containing a reagent which modifies unmethylated cytosine and a second container containing primers for amplification of a CpG-containing nucleic acid, wherein the primers distinguish between modified methylated and nonmethylated nucleic acid, and optionally, a third container containing a methylation sensitive restriction endonuclease. In a preferred embodiment, the cell proliferative disorder is pancreatic carcinoma.

[0020] To identify CpG islands differentially methylated in pancreatic adenocarcinoma, methylated CpG island amplification (MCA) was used, coupled with representational difference analysis (MCA/RDA). Clones differentially methylated (termed MICP, methylated in carcinoma of the pancreas) were isolated in a panel of 8 pancreatic cancer cell lines compared to normal pancreas. 95% of these clones were CpG islands and among these clones were 5' CpG islands of several known genes, including Cyclin G and Preproenkephalin (ppENK, encoding [Met5]-enkephalin). Seven of the clones (Cyclin G, ppENK, MICP20, 23, 33, 35 and 36) were not methylated in 14 normal pancreata by MSP while 15 primary pancreatic adenocarcinomas were methylated in 7%, 87%, 13%, 53%, 33%, 40% and 0% of cases, respectively. Two of the 5 chronic pancreatitis specimens harbored methylation of three (Cyclin G, ppENK and MICP23) and one (ppENK) clones, respectively. There was no identification of methylation of MICP33 and 35 in any normal gastrointestinal tissues tested. Three clones (ppENK, MICP20 and 23) were variably methylated in normal gastric, duodenal and colonic mucosae. Aberrant methylation of Cyclin G and ppENK in methylated pancreatic cancer cell lines was associated with transcriptional silencing that was reversible with 5-aza-2'-deoxycytidine treatment.

[0021] These data indicate that multiple CpG islands undergo de novo methylation during pancreatic carcinogenesis. Methylation of some CpG islands is cancer-specific while others show tissue-specific patterns of methylation.

BRIEF DESCRIPTION OF THE FIGURES

[0022] FIG. 1 shows the representative results of MCIPs isolated by MCA/RDA. Dot blot analysis. 1A, An example of kinetic enrichment of methylated sequences by RDA. MCA products from the driver and the tester (PL8) and the PCR products from first (1st SA), second (2nd SA) and third (3rd SA) competitive hybridization/selective amplification were blotted onto the membrane and hybridized with a labeled Cyclin G probe. 1B, Dot blot-analysis using MICPs isolated from MCA/RDA as probes. First three MICPs were hybridized only to the tester (either PL3 or PL8), whereas the next three MICPs were weakly hybridized to the driver as well as the tester.

[0023] FIG. 2 shows a CpG plot across the 5' CpG islands of Cyclin G and ppENK showing the relation between isolated clones and their corresponding genes. The positions of MSP primers are also indicated. The black boxes represent exons.

[0024] FIG. 3 shows a summary of the average level of methylation of selected MCA/RDA MICPs by bisulfite-modified genomic sequencing. The specimen number and the age of the patients are in the left column. Empty oval, 0-10% methylation; white oval with black dots, 11-30% methylation; black oval with white dots, 31-70% methylation; black oval, 71-100% methylation; .cndot., not determined. NP, normal pancrease; PCa, primary pancreatic adenocarcinonma.

[0025] FIG. 4 shows MSP analyses of MICP36 (A) and MICP25 (B) in primary pancreatic adenocarcinomas and normal tissues. The PCR products in the lanes U and M indicate the presence of unmethylated and methylated templates, respectively. PCa, primary pancreatic carcinoma, NP, normal pancreas, NC, normal colonic mucosa. Expression of Cyclin G (C) and ppENK (D) in pancreatic cancer cell lines and normal pancreata by RT-PCR analysis. Cyclin G and ppENK were coamplified with GAPDH to ensure the RNA integrity. Cyclin G was expressed at low level in the methylated cell line PL8 compared with unmethylated cell lines (PL3, CAPAN2, MiaPaca2, and Panc1) and four normal pancreata. 5 Aza-dC treatment increased Cyclin G expression in PL8. All methylated cell lines examined (PL3, CAPAN2, MiaPaca2 and Panc1) lacked expression of ppENK, whereas all three normal pancreata expressed ppENK. Treatment of four cell lines with 5 Aza-dC restored the ppENK expression. The expected sizes of the PCR products are 306 bp for GAPDH, 207 bp for Cyclin G, and 179 bp for ppENK.

[0026] FIG. 5 shows aberrant methylation of MICPs and clinicopathological variables. Multivariate linear regression analysis was used to determine the relationship between the number of aberrantly methylated MICPs in a pancreatic adenocarcinoma and the variables patient age (left plot) and tumor diameter (right plot). For each decade increase in age, the average number of methylated loci increases by 0.28, regardless of the tumor diameter. For each increase in tumor diameter (in cm), the number of mehtylated loci increases by 0.156 regardless of patient age.

[0027] FIGS. 6a-6i show the nucleic acid sequence for SEQ ID NO:1-42.

DETAILED DESCRIPTION OF THE INVENTION

[0028] It has been determined that an aberrant methylation state of nucleic acids in certain genes, particularly regulatory sequences, is diagnostic for the presence or potential development of a cellular proliferative disorder in subjects bearing the aberrantly methylated nucleic acids. More particularly, the hypermethylation of certain nucleotides localized in CpG islands has been shown to affect the expression of genes associated with the CpG islands; typically such hypermethylated genes have reduced or abolished expression, primarily due to down-regulated transcription. Using a recently developed PCR-based technique called methylated CpG island amplification (MCA), several nucleic acid molecules aberrantly methylated in pancreatic cancer cells line were identified.

[0029] Of the 42 unique clones isolated using MCA/RDA, seven (Cyclin G, ppENK, MICP20, 23, 33, 35 and 36) were CpG islands aberrantly methylated in pancreatic carcinoma compared to normal pancreas. Indeed, none of these 7 clones, were methylated in a panel of normal pancreata using the highly sensitive method of MSP. Two of these seven clones corresponded to the 5' CpG island of the genes ppENK and Cyclin G. ppENK encodes opioid growth factor, also known as [Met5]-enkephalin. This opioid peptide induces apoptosis in lung cancer cell lines (Maneckjee & Minna, Cell Growth Differ. 5:1033-1040, 1994) and several studies have demonstrated that this peptide has a negative growth regulatory effect on various kinds of cancers, including pancreatic cancer (Zagon, et al., Int. J. Oncol. 14:577-584, 1999). Aberrant methylation of the 5' CpG island of ppENK was found in 87% of primary pancreatic carcinomas. Hypermethylation was associated with transcriptional silencing of this gene in pancreatic cancer cell lines. Although a low level of methylation in other normal mucosae by MSP was also observed, these results indicate that de novo methylation of 5' CpG islands and transcriptional repression of ppENK might contribute to pancreatic carcinogenesis.

[0030] Cyclin G is a target for transcriptional activation by p53 (Okamoto, et al., Embo. J. 13:48616-4822, 1994). Conflicting functions for Cyclin G have been reported. When overexpressed Cyclin G is associated with cell growth in vitro (Smith, et al., Exp. Cell. Res. 230:61-68, 1997), while increased expression of Cyclin G also augments apoptosis of multiple cancer cell lines in response to different stimuli (Okamoto & Prives, Oncogene 18:4606-4615, 1999). The data demonstrate the association between hypermethylation of the 5'CpG island of Cyclin G and its transcriptional repression in the pancreatic cancer cell line PL8 (FIG. 4C), suggesting that Cyclin G could have been selected for silencing because of tumor suppressive functions. These data extend previous findings demonstrating aberrant methylation of multiple cancer-related genes including p16 and hMLH1 in pancreatic cancer (Ueki, et al., Cancer Res. 60:1835-1839, 2000).

[0031] Methylation of several clones in pancreata with chronic pancreatitis was also observed. Two of the 5 pancreata with chronic pancreatitis harbored aberrant methylation and one of these two pancreata contained a PanIN lesion and this latter sample displayed methylation of 3 clones. Previous studies demonstrated that chronic pancreatitis is a significant risk factor of development of pancreatic cancer (Lowenfels, et al., N. Engl. J. Med. 328:1433-1437, 1993) and duct lesions (PanIN) often found in chronic pancreatitis are considered as the precursors of infiltrating pancreatic carcinoma (Hruban, et al., Clin. Cancer Res. 6:2969-2972, 2000). The presence of aberrant methylation in DNA from chronic pancreatitis suggests that de novo methylation of CpG islands may be an early event in pancreatic cancer development in this setting. Additional studies are needed to determine the role and the timing of de novo methylation of CpG islands in progression model of pancreatic cancer (Hruban, et al., Clin. Cancer Res. 6:2969-2972, 2000). The absence of methylated templates of MICP33 and 35 in any normal tissue examined raises the possibility that MSP could be used to detect aberrant methylation of these clones in clinical samples such as stool, blood, or pancreatic fluid, for the early detection of pancreatic cancers.

[0032] Three additional known genes containing methylated 5'CpG islands in pancreatic cancers were identified (GAD1, ECEL1 and PAX5). These genes were isolated by MCA/RDA because relatively fewer DNA templates were methylated in normal pancreas. Some genes that are methylated in cancer and in only a small percentage of normal cells may appear to undergo selection during carcinogenesis (Salem, et al., Cancer Res. 60:2473-2476, 2000), but are merely unselected epigenetic markers of stem cells that have evolved to cancers by other clonal selection events. This phenomenon is important to be aware of when studying methylation in human cancer and makes it much more difficult to assign causality to methylation phenomena in cancers compared to genetic events such as homozygous deletion. For normally unmethylated genes whose function is well characterized such as hMLH1, or for genes that are methylated as a second hit for a tumor suppressor gene (e.g. VHL, E-cadherin), or for tumor suppressor genes alternatively targeted by genetic and epigenetic inactivation (e.g. p16 and RB) (Baylin, et al., Adv. Cancer Res. 72:141-196, 1998), the biological significance of "aberrant methylation" is well accepted. As additional genes are identified that are methylated in pancreatic cancer, it will be important to rule out low-level methylation using sensitive techniques such as MSP (Herman, et al., Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996) before such genes are accepted as having undergone selection through methylation.

[0033] Three CpG islands that were differentially methylated in pancreatic cancer but were not located within 5' regions of their corresponding genes were also identified, CSX, MCT3 and ICAM5. The role of hypermethylation of non-5' CpG islands need to be defined, but recent studies have shown that aberrant methylation in cancer is not confined to the 5' region but can occur in internal exons and 3' regions of genes (Costello, et al., Nat. Genet. 24:132-138, 2000, Liang, et al., Genomics 53:260-268, 1998). It is probable that several of the unknown clones identified are genes whose expression is repressed by aberrant methylation. For example, GRAIL and GENSCAN programs suggest putative coding exons downstream of MICP23 and MICP33.

[0034] In addition to identifying CpG islands with cancer-related methylation, CpG islands that were methylated in both normal pancreata and neoplastic tissues were isolated. Methylation of non-neoplastic colorectal (Ahuja, et al., Cancer Res. 58:5489-5494, 1998, Toyota, et al., Proc. Natl. Acad. Sci. USA 96:8681-8686, 1999), bladder and prostate tissues (Liang, et al., Genomics 53:260-268, 1998) has been observed for several genes and CpG islands. Methylation in normal tissues that is not the result of imprinting is frequently "age-related." This has been best shown in the colonic mucosa for genes such as ER (Ahuja, et al., Cancer Res. 58:5489-5494, 1998, Toyota, et al., Proc. Natl. Acad. Sci. USA 96:8681-8686, 1999). However, normal pancreata used in this study were obtained from 14 patients (mean age of 62) and for at least 5 of these clones there was no evidence of age-related methylation (FIG. 3). This suggests that the aberrant methylation observed in the pancreatic cancers in this study is not simply a function of age. Some of the clones listed in Table 1 could undergo age-related methylation, but to demonstrate this would require many more normal pancreata. Methylation of ppENK, MICP20 and 23 in a significant percentage of the DNA templates within normal gastric, duodenal and colonic mucosae highlights the tissue specific nature of DNA methylation. This low level methylation of different normal tissues might also explain some of the tumor-specific methylation patterns observed by others (Costello, et al., Nat. Genet. 24:132-138, 2000). The data indicate that there are tissue-specific methylation patterns in normal tissues (Liang, et al., Genomics 53:260-268, 1998) and age-related methylation changes are probably restricted to certain genes and/or tissues (Ahuja, et al., Cancer Res. 58:5489-5494, 1998).

[0035] In this study, the MCA/RDA technique was modified and these modifications may have improved the efficiency of the MCA/RDA technique. Betaine was included in the PCR reaction and amplified the methylated templates under a higher annealing temperature (77.degree. C.). The combination of betaine and DMSO can uniformly amplify a mixture of DNA with different GC content (Baskaran, et al., Genome Res. 6:633-638, 1996). These modifications might have enhanced the amplification of distinct clones instead of Alu repetitive sequences that accounted for 60% of the recovered clones using the original protocol (Toyota, et al., Cancer Res. 59:2307-2312, 1997). The subtractive and kinetic enrichment of differentially methylated sequence by RDA as shown in this study (FIG. 1A) may have advantages over other techniques to isolate differentially methylated sequences between normal tissue and cancer (Costello, et al., Nat. Genet. 24:132-138, 2000, Liang, et al., Genomics 53:260-268, 1998). MCA/RDA, however, has limitations for identifying differentially methylated sequences. First, MCA only detects differentially methylated sequences with two restriction enzyme sites (SmaI in this study). Second, MCA/RDA not only identifies absolute differences in methylation between the tester and the driver, it also will identify methylated sequences both in the tester and the driver if for example, there is low level methylation in the driver (normal pancreas). Third, some clones that appeared to be unmethylated by dot-blot hybridization were indeed methylated by bisulfite sequencing in normal pancreata (MICP15, FIG. 1B and FIG. 4C). Finally, even if bisulfite sequencing suggests that there is no methylation of a clone in normal pancreata, it is important to rule out low-level methylation in normal tissues using a sensitive technique, such as MSP.

[0036] The results indicate that aberrant methylation of CpG islands is a common event in pancreatic carcinogenesis. Some de novo methylated CpG islands could serve as cancer-specific markers while others reflect tissue specific pattern of DNA methylation.

[0037] Methylated nucleic acid sequences are also provided. For the first time, the present invention provides methylated chemical structures for MICP 1-42 (see Table 1). One of skill in the art can now readily locate the CpG-rich sequences associated with these genes and identify such methylated forms of the genes/regulatory sequences by methods described herein (The gene sequences can be identified in a gene database found at http://www.ncbi.nim.nih.gov/UniGene/index.html). The invention provides CpG-rich regions from the above genes as set forth in SEQ ID Nos 1-42, equivalent to MICP 1-42, respectively.

[0038] The term "polynucleotide" or "nucleic acid sequence" refers to a polymeric form of nucleotides at least 10 bases in length. An "isolated polynucleotide" is a polynucleotide that is not immediately contiguous with both of the coding sequences with which it is immediately contiguous (one on the 5' end and one on the 3' end) in the naturally occurring genome of the organism from which it is derived. Thus, an isolated polynucleotide may include a coding region with its associated regulatory sequences. The term therefore includes, for example, a recombinant DNA which is incorporated into a vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., a cDNA) independent of other sequences. The nucleotides of the invention can be ribonucleotides, deoxyribonucleotides, or modified forms of either nucleotide. Specifically, methylated forms of nucleotides in a polynucleotide sequence are also included. The term includes single and double forms of DNA.

[0039] As will be understood by those of skill in the art, when the sequence is RNA, the deoxynucleotides A, G, C, and T of SEQ ID NO:1-42, are replaced by ribonucleotides A, G, C, and U, respectively. Also included in the invention are fragments of the above-described nucleic acid sequences that are at least 15 bases in length, which is sufficient to permit the fragment to selectively hybridize to DNA that encodes polypeptides encoded by SEQ ID NO:1-42. The term "selectively hybridize" refers to hybridization under moderately or highly stringent conditions (See, Sambrook, as cited herein) which excludes non-related nucleotide sequences.

[0040] The nucleic acid sequence includes the disclosed sequence and sequences that encode conservative variations of the polypeptides encoded by polynucleotides provided herein. The term "conservative variation" as used herein denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like. The term "conservative variation" also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide.

[0041] Nucleic acid sequences of the invention can be expressed in vitro by DNA transfer into a suitable host cell. "Host cells" are cells in which a vector can be propagated and its DNA expressed. The cell may be prokaryotic or eukaryotic. The term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term "host cells" is used. Methods of stable transfer, meaning that the foreign DNA is continuously maintained in the host, are known in the art.

[0042] In one aspect, the nucleic acid sequences may be inserted into an expression vector. The term "expression vector" refers to a plasmid, virus or other vehicle known in the art that has been manipulated by insertion or incorporation of the sequence of interest genetic sequences. Polynucleotide sequence which encode sequence of interest can be operatively linked to expression control sequences. "Operatively linked" refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. An expression control sequence operatively linked to a coding sequence is ligated such that expression of the coding sequence is achieved under conditions compatible with the regulatory or expression control sequences. As used herein, the terms "regulatory sequences" and "expression control sequences" refers to nucleic acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively linked. Expression control sequences are operatively linked to a nucleic acid sequence when the expression control sequences control and regulate the transcription and, as appropriate, translation of the nucleic acid sequence. Thus expression control sequences can include appropriate promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein-encoding gene, splicing signal for introns, maintenance of the correct reading frame of that gene to permit proper translation of mRNA, and stop codons. The terms "regulatory sequences" and "expression control sequences" are intended to included, at a minimum, components whose presence can influence expression, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. An example of an expression control sequence includes a promoter.

[0043] A "promoter" is a minimal sequence sufficient to direct transcription. Also included in the invention are those promoter elements which are sufficient to render promoter-dependent gene expression controllable for cell-type specific, tissue-specific, or inducible by external signals or agents; such elements may be located in the 5' or 3' regions of the gene. Both constitutive and inducible promoters, are included in the invention (see, e.g., Bitter et al., Methods in Enzymology 153:516-544, 1987). For example, when cloning in bacterial systems, inducible promoters such as pL of bacteriophage plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used. When cloning in mammalian cell systems, promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the retrovirus long terminal repeat; the adenovirus late promoter; the vaccinia virus 7.5K promoter) may be used. Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the nucleic acid sequences of the invention.

[0044] In the present invention, the polynucleotide sequences may be inserted into an expression vector which contains a promoter sequence which facilitates the efficient transcription of the inserted genetic sequence of the host. The expression vector typically contains an origin of replication, a promoter, as well as specific genes which allow phenotypic selection of the transformed cells. Vectors suitable for use in the present invention include, but are not limited to the T7-based expression vector for expression in bacteria (Rosenberg et al., Gene 56:125, 1987), the pMSXND expression vector for expression in mammalian cells (Lee and Nathans, J. Biol. Chem. 263:3521, 1988) and baculovirus-derived vectors for expression in insect cells. The DNA segment can be present in the vector operably linked to regulatory elements, for example, a promoter (e.g., T7, metallothionein I, or polyhedron promoters).

[0045] Polynucleotide sequences of the invention can be expressed in either prokaryotes or eukaryotes. Hosts can include microbial, yeast, insect and mammalian organisms. Methods of expressing DNA sequences having eukaryotic or viral sequences in prokaryotes are well known in the art. Biologically functional viral and plasmid DNA vectors capable of expression and replication in a host are known in the art. Such vectors are used to incorporate DNA sequences of the invention.

[0046] "Transformation" means a genetic change induced in a cell following incorporation of new DNA (i.e., DNA exogenous to the cell). Where the cell is a mammalian cell, the genetic change is generally achieved by introduction of the DNA into the genome of the cell (i.e., stable).

[0047] Thus, a "transformed cell" is a cell into which (or into an ancestor of which) has been introduced, by means of recombinant DNA techniques, a DNA molecule encoding sequence of interest. Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as E. coli, competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCl2 method using procedures well known in the art. Alternatively, MgCl2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell if desired.

[0048] When the host is a eukaryote, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransformed with DNA sequences encoding the sequence of interest, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein (see for example, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).

[0049] Isolation and purification of microbial expressed polypeptide, or fragments thereof, provided by the invention, may be carried out by conventional means including preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies.

[0050] In one embodiment, the invention provides substantially purified polypeptide encoded by polynucleotide sequences SEQ ID NO:1-42. The term "substantially purified" as used herein refers to a polypeptide which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. One skilled in the art can purify a polypeptide sequence using standard techniques for protein purification. The substantially pure polypeptide will yield a single major band on a non-reducing polyacrylamide gel. The purity of the polypeptide can also be determined by amino-terminal amino acid sequence analysis.

[0051] Minor modifications of the primary amino acid sequences may result in proteins which have substantially equivalent activity as compared to the unmodified counterpart polypeptide described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous. All of the polypeptides produced by these modifications are included herein as long as the biological activity still exists.

[0052] The polypeptides of the invention also include dominant negative forms of the invention polypeptide which do not have the biological activity of invention polypeptide sequence. A "dominant negative form" of invention is a polypeptide that is structurally similar to invention polypeptide but does not have wild-type invention function. For example, a dominant-negative invention polypeptide may interfere with wild-type invention function by binding to, or otherwise sequestering, regulating agents, such as upstream or downstream components, that normally interact functionally with the invention polypeptide.

[0053] To identify genes differentially methylated in colorectal cancer, methylated CpG island amplification was used followed by representational difference analysis (Razin and Cedar, Cell 17: 473-476, 1994, herein incorporated by reference). One of the clones recovered (MINT31, see U.S. patent application Ser. No. 09/309,175, incorporated by reference herein in its entirety) mapped to human chromosome 17q21 using a radiation hybrid panel. A Blast search revealed this fragment to be completely identical to part of a BAC clone (Genbank: AC004590) sequenced by high throughput genomic sequencing. The region surrounding MINT31 fulfills the criteria of a CpG island: GC content 0.67, CpG/GpC ratio 0.78 and a total of 305 CpG sites in a 4 kb region. Using this CpG island and 10 kb of flanking sequences in a Blast analysis, several regions highly homologous to the rat T-type calcium channel gene, CACNA1G, were identified (Perez-Reyes et al., Nature 391: 896-900. 1998, herein incorporated by reference). Several ESTs were also identified in this region. Using Genscan, 2 putative coding sequences (G1, and G2) were identified. Blastp analysis revealed that G1 has a high homology to the EH-domain-binding protein, epsin, while G2 is homologous to a C. elegans hypothetical protein (accession No. 2496828).

[0054] Due to the clear correlation between methylation of CpG islands and cellular proliferative disorders, in another embodiment of the present invention, there are provided methods for detecting a cellular proliferative disorder in a subject having or at risk for said cellular proliferative disorder. The method includes assaying, in nucleic acid-containing specimen taken from said subject, the methylation state of a gene or its associated regulatory regions, wherein the expression state of the gene or its associated regulatory regions is associated with the presence of the cellular proliferative disorder, and identifying as having a cellular proliferative disorder a subject that has aberrant methylation of regions of said gene. The method provides for detecting a cellular proliferative disorder in a subject having or at risk for said cellular proliferative disorder by identifying aberrantly methylation of regions of a gene when compared to the same regions of the gene in a subject not having said cellular proliferative disorder.

[0055] The aberrant methylation comprises hypermethylated CpG rich regions (i.e., islands). In one aspect of the present invention, the CpG rich regions are associated with the invention genes gene, and affect the expression thereof in a methylation state-dependent manner.

[0056] A "cell proliferative disorder" or "cellular proliferative disorder" is any disorder in which the proliferative capabilities of the affected cells is different from the normal proliferative capabilities of unaffected cells. An example of a cell proliferative disorder is neoplasia. Malignant cells (i.e., cancer) develop as a result of a multistep process. Specific, non-limiting examples of cell proliferative disorders associated with increased methylation of CpG-islands are low grade astrocytoma, anaplastic astrocytoma, glioblastoma, medulloblastoma, gastric cancer, colorectal cancer, colorectal adenoma, acute myelogenous leukemia, lung cancer, renal cancer, pancreatic cancer, leukemia, breast cancer, prostate cancer, endometrial cancer and neuroblastoma.

[0057] A cell proliferative disorder as described herein may be a neoplasm. Such neoplasms are either benign or malignant. The term "neoplasm" refers to a new, abnormal growth of cells or a growth of abnormal cells that reproduce faster than normal. A neoplasm creates an unstructured mass (a tumor) which can be either benign or malignant. For example, the neoplasm may be a head, neck, lung, esophageal, stomach, small bowel, colon, bladder, kidney, or cervical neoplasm. The term "benign" refers to a tumor that is noncancerous, e.g. its cells do not proliferate or invade surrounding tissues. The term "malignant" refers to a tumor that is metastastic or no longer under normal cellular growth control.

[0058] A cell proliferative disorder may be an age-associated disorder. Examples of age-associated disorders which are cell proliferative disorders include colon cancer, lung cancer, breast cancer, prostate cancer, and melanoma, amongst others.

[0059] A "nucleic acid containing specimen" includes any type of material containing a nucleic acid to be subject to invention methods. The nucleic acid may be contained in a biological sample. Such samples include but are not limited to any bodily fluid, such as a serum, urine, saliva, blood, cerebrospinal fluid, pleural fluid, ascites fluid, sputum, stool, or a biopsy sample.

[0060] Samples or specimens include any CpG-rich DNA sequence, whatever the origin, as long as the sequence is detectably present in a sample. While routine diagnostic tests may not be able to identify cancer cells in these samples, the method of the present invention identifies neoplastic cells derived from the primary tumor or from a metastases. The method includes non-invasive sampling (e.g., bodily fluid) as well as invasive sampling (e.g., biopsy). The sample of DNA of the subject may be serum, plasma, lymphocytes, urine, sputum, bile, stool, cervical tissue, saliva, tears, pancreatic juice, duodenal juice, cerebral spinal fluid, regional lymph node, histopathologic margins, and any bodily fluid that drains a body cavity or organ. Therefore, the method provides for the non-invasive detection of various tumor types including head and neck cancer, lung cancer, esophageal cancer, stomach cancer, small bowel cancer, colon cancer, bladder cancer, kidney cancers, cervical cancer and any other organ type that has a draining fluid accessible to analysis. For example, neoplasia of regional lymph nodes associated with a primary mammary tumor can be detected using the method of the invention. Regional lymph nodes for head and neck carcinomas include cervical lymph nodes, prelaryngeal lymph nodes, pulmonary juxta-esophageal lymph nodes and submandibular lymph nodes. Regional lymph nodes for mammary tissue carcinomas include the axillary and intercostal nodes. Samples also include urine DNA for bladder cancer or plasma or saliva DNA for head and neck cancer patients.

[0061] Any nucleic acid sample, in purified or nonpurified form, can be utilized as the starting nucleic acid or acids in accordance with the present invention, provided it contains, or is suspected of containing, a nucleic acid sequence containing a target locus (e.g., CpG-containing nucleic acid). In general, the CpG-containing nucleic acid is DNA. However, invention methods may employ, for example, samples that contain DNA, or DNA and RNA, including messenger RNA, wherein DNA or RNA may be single stranded or double stranded, or a DNA-RNA hybrid may be included in the sample. A mixture of nucleic acids may also be employed. The specific nucleic acid sequence to be detected may be a fraction of a larger molecule or can be present initially as a discrete molecule, so that the specific sequence constitutes the entire nucleic acid. It is not necessary that the sequence to be studied be present initially in a pure form; the nucleic acid may be a minor fraction of a complex mixture, such as contained in whole human DNA. The nucleic acid-containing sample used for detection of methylated CpG may be from any source including, but not limited to, brain, colon, urogenital, lung, renal, pancreas, liver, esophagus, stomach, hematopoietic, breast, thymus, testis, ovarian, and uterine tissue, and may be extracted by a variety of techniques such as that described by Maniatis, et al. (Molecular Cloning: a Laboratory Manual, Cold Spring Harbor, N.Y., pp 280, 281, 1982).

[0062] The nucleic acid of interest can be any nucleic acid where it is desirable to detect the presence of a differentially methylated CpG island. The CpG island comprises a CpG island located in a gene or regulatory region for a gene. A "CpG island" is a CpG rich region of a nucleic acid sequence. The nucleic acid sequence may include, for example, MICP 1-42. However, any gene or nucleic acid sequence of interest containing a CpG sequence can provide diagnostic information (i.e., via its methylation state) using invention methods.

[0063] Moreover, these markers can also be multiplexed in a single amplification reaction to generate a low cost, reliable cancer screening test for many cancers simultaneously.

[0064] A combination of DNA markers for CpG-rich regions of nucleic acid may be amplified in a single amplification reaction. The markers are multiplexed in a single amplification reaction, for example, by combining primers for more than one locus. For example, DNA from a urine sample can be amplified with three different randomly labeled primer sets, such as those used for the amplification of the MICP38-42 loci, in the same amplification reaction. The reaction products are separated on a denaturing polyacrylamide gel, for example, and then exposed to film for visualization and analysis. By analyzing a panel of markers, there is a greater probability of producing a more useful methylation profile for a subject.

[0065] If the sample is impure (e.g., plasma, serum, stool, ejaculate, sputum, saliva, cerebrospinal fluid, or blood or a sample embedded in paraffin), it may be treated before amplification with a reagent effective for lysing the cells contained in the fluids, tissues, or animal cell membranes of the sample, and for exposing the nucleic acid(s) contained therein. Methods for purifying or partially purifying nucleic acid from a sample are well known in the art (e.g., Sambrook et al., Molecular Cloning: a Laboratory Manual, Cold Spring Harbor Press, 1989, herein incorporated by reference).

[0066] In order to detect a differential methylation state for a gene or CpG-containing region of interest, invention methods include any means known in the art for detecting such differential methylation. For example, detecting the differential methylation may include contacting the nucleic acid-containing specimen with an agent that modifies unmethylated cytosine, amplifying a CpG-containing nucleic acid in the specimen by means of CpG-specific oligonucleotide primers, wherein the oligonucleotide primers distinguish between modified methylated and nonmethylated nucleic acid, and detecting the methylated nucleic acid based on the presence or absence of amplification products produced in said amplifying step. This embodiment includes the PCR-based methods described in U.S. Pat. No. 5,786,146, incorporated herein in its entirety.

[0067] For the first time, the methylation state of a number of genes has been correlated with cell proliferative disorders. Examples of such genesand their NCBI accession numbers, including the location of the clone, are set out in Table 1.

[0068] In another embodiment, detection of differential methylation is accomplished by contacting a nucleic acid sample suspected of comprising a CpG-containing nucleic acid with a methylation sensitive restriction endonuclease that cleaves only unmethylated CpG sites under conditions and for a time to allow cleavage of unmethylated nucleic acid. The sample is further contacted with an isoschizomer of the methylation sensitive restriction endonuclease, that cleaves both methylated and unmethylated CpG-sites, under conditions and for a time to allow cleavage of methylated nucleic acid. Oligonucleotides are added to the nucleic acid sample under conditions and for a time to allow ligation of the oligonucleotides to nucleic acid cleaved by the restriction endonuclease, and the digested nucleic acid is amplified by conventional methods such as PCR wherein primers complementary to the oligonucleotides are employed. Following identification, the methylated CpG-containing nucleic acid can be cloned, using method well known to one of skill in the art (see Sambrook et al., Molecular Cloning: a Laboratory Manual, Cold Spring Harbor Press, 1989).

[0069] As used herein, a "methylation sensitive restriction endonuclease" is a restriction endonuclease that includes CG as part of its recognition site and has altered activity when the C is methylated as compared to when the C is not methylated. Preferably, the methylation sensitive restriction endonuclease has inhibited activity when the C is methylated (e.g., SmaI). Specific non-limiting examples of a methylation sensitive restriction endonucleases include Sma I, BssHII, or HpaII. Such enzymes can be used alone or in combination. Other methylation sensitive restriction endonucleases will be known to those of skill in the art and include, but are not limited to SacII, EagI, and BstUI, for example. An "isoschizomer" of a methylation sensitive restriction endonuclease is a restriction endonuclease which recognizes the same recognition site as a methylation sensitive restriction endonuclease but which cleaves both methylated and unmethylated CGs. One of skill in the art can readily determine appropriate conditions for a restriction endonuclease to cleave a nucleic acid (see Sambrook et al., Molecular Cloning: a Laboratory Manual, Cold Spring Harbor Press, 1989). Without being bound by theory, actively transcribed genes generally contain fewer methylated CGs than in other genes.

[0070] In one embodiment of the invention, a nucleic acid of interest is cleaved with a methylation sensitive endonuclease. In one aspect, cleavage with the methylation sensitive endonuclease creates a sufficient overhang on the nucleic acid of interest. Following cleavage with the isoschizomer, the cleavage product can still have a sufficient overhang. An "overhang" refers to nucleic acid having two strands wherein the strands end in such a manner that a few bases of one strand are not base paired to the other strand. A "sufficient overhang" refers to an overhang of sufficient length to allow specific hybridization of an oligonucleotide of interest. In one embodiment, a sufficient overhang is at least two bases in length. In another embodiment, the sufficient overhang is four or more bases in length. An overhang of a specific sequence on the nucleic acid of interest may be desired in order for an oligonucleotide of interest to hybridize. In this case, the isoschizomer can be used to create the overhang having the desired sequence on the nucleic acid of interest.

[0071] In another aspect of this embodiment, the cleavage with a methylation sensitive endonuclease results in a reaction product of the nucleic acid of interest that has a blunt end or an insufficient overhang. In this embodiment, an isoschizomer of the methylation sensitive restriction endonuclease can create a sufficient overhang on the nucleic acid of interest. "Blunt ends" refers to a flush ending of two stands, the sense stand and the antisense strand, of a nucleic acid.

[0072] Once a sufficient overhang is created on the nucleic acid of interest, an oligonucleotide is ligated to the nucleic acid cleaved of interest which has been cleaved by the methylation specific restriction endonuclease. "Ligation" is the attachment of two nucleic acid sequences by base pairing of substantially complementary sequences and/or by the formation of covalent bonds between two nucleic acid sequences. In one aspect of the present invention, an "oligonucleotide" is a nucleic acid sequence of about 2 up to about 40 bases in length. It is presently preferred that the oligonucleotide is from about 15 to 35 bases in length.

[0073] In one embodiment, an adaptor is utilized to create DNA ends of desired sequence and overhang. An "adaptor" is a double-stranded nucleic acid sequence with one end that has a sufficient single-stranded overhang at one or both ends such that the adaptor can be ligated by base-pairing to a sufficient overhang on a nucleic acid of interest that has been cleaved by a methylation sensitive restriction enzyme or an isoschizomer of a methylation sensitive restriction enzyme. Adaptors can be obtained commercially, or two oligonucleotides can be utilized to form an adaptor. Thus, in one embodiment, two oligonucleotides are used to form an adaptor; these oligonucleotides are substantially complementary over their entire sequence except for the region(s) at the 5' and/or 3' ends that will form a single stranded overhang. The single stranded overhang is complementary to an overhang on the nucleic acid cleaved by a methylation sensitive restriction enzyme or an isoschizomer of a methylation sensitive restriction enzyme, such that the overhang on the nucleic acid of interest will base pair with the 3' or 5' single stranded end of the adaptor under appropriate conditions. The conditions will vary depending on the sequence composition (GC vs AT), the length, and the type of nucleic acid (see Sambrook et al., Molecular Cloning: a Laboratory Manual, 2nd Ed.; Cold Spring Harbor Laboratory Press, Plainview, N.Y., 1998).

[0074] Following the ligation of the oligonucleotide, the nucleic acid of interest is amplified using a primer complementary to the oligonucleotide. Specifically, the term "primer" as used herein refers to a sequence comprising two or more deoxyribo-nucleotides or ribonucleotides, preferably more than three, and more preferably more than eight, wherein the sequence is capable of initiating synthesis of a primer extension product, which is substantially complementary to a nucleic acid such as an adaptor or a ligated oligonucleotide. Environmental conditions conducive to synthesis include the presence of nucleoside triphosphates and an agent for polymerization, such as DNA polymerase, and a suitable temperature and pH. The primer is preferably single stranded for maximum efficiency in amplification, but may be double stranded. If double stranded, the primer is first treated to separate its strands before being used to prepare extension products. In one embodiment, the primer is an oligodeoxyribo-nucleotide. The primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing agent for polymerization. The exact length of primer will depend on many factors, including temperature, buffer, and nucleotide composition. The oligonucleotide primer typically contains 12-20 or more nucleotides, although it may contain fewer nucleotides.

[0075] Primers of the invention are designed to be "substantially" complementary to each strand of the oligonucleotide to be amplified and include the appropriate G or C nucleotides as discussed above. This means that the primers must be sufficiently complementary to hybridize with their respective strands under conditions which allow the agent for polymerization to perform. In other words, the primers should have sufficient complementarity with a 5' and 3' oligonucleotide to hybridize therewith and permit amplification of CpG containing nucleic acid sequence.

[0076] Primers of the invention are employed in the amplification process which is an enzymatic chain reaction that produces exponential quantities of target locus relative to the number of reaction steps involved (e.g., polymerase chain reaction or PCR). Typically, one primer is complementary to the negative (-) strand of the locus and the other is complementary to the positive (+) strand. Annealing the primers to denatured nucleic acid followed by extension with an enzyme, such as the large fragment of DNA Polymerase I (Klenow) and nucleotides, results in newly synthesized + and - strands containing the target locus sequence. Because these newly synthesized sequences are also templates, repeated cycles of denaturing, primer annealing, and extension results in exponential production of the region (i.e., the target locus sequence) defined by the primer. The product of the chain reaction is a discrete nucleic acid duplex with termini corresponding to the ends of the specific primers employed.

[0077] The oligonucleotide primers of the invention may be prepared using any suitable method, such as conventional phosphotriester and phosphodiester methods or automated embodiments thereof. In one such automated embodiment, diethylphos-phoramidites are used as starting materials and may be synthesized as described by Beaucage, et al. (Tetrahedron Letters, 22:1859-1862, 1981). One method for synthesizing oligonucleotides on a modified solid support is described in U.S. Pat. No. 4,458,066.

[0078] Where the CpG-containing nucleic acid sequence of interest contains two strands, it is necessary to separate the strands of the nucleic acid before it can be used as a template for the amplification process. Strand separation can be effected either as a separate step or simultaneously with the synthesis of the primer extension products. This strand separation can be accomplished using various suitable denaturing conditions, including physical, chemical, or enzymatic means, the word "denaturing" includes all such means. One physical method of separating nucleic acid strands involves heating the nucleic acid until it is denatured. Typical heat denaturation may involve temperatures ranging from about 80.degree. to 105.degree. C. for times ranging from about 1 to 10 minutes. Strand separation may also be induced by an enzyme from the class of enzymes known as helicases or by the enzyme RecA, which has helicase activity, and in the presence of riboATP, is known to denature DNA. The reaction conditions suitable for strand separation of nucleic acids with helicases are described by Kuhn Hoffmann-Berling (CSH-Quantitative Biology, 43:63, 1978) and techniques for using RecA are reviewed in C. Radding (Ann. Rev. Genetics, 16:405-437, 1982).

[0079] When complementary strands of nucleic acid or acids are separated, regardless of whether the nucleic acid was originally double or single stranded, the separated strands are ready to be used as a template for the synthesis of additional nucleic acid strands. This synthesis is performed under conditions allowing hybridization of primers to templates to occur. Generally synthesis occurs in a buffered aqueous solution, generally at a pH of about 7-9. Preferably, a molar excess (for genomic nucleic acid, usually about 108:1 primer:template) of the two oligonucleotide primers is added to the buffer containing the separated template strands. It is understood, however, that the amount of complementary strand may not be known if the process of the invention is used for diagnostic applications, so that the amount of primer relative to the amount of complementary strand cannot be determined with certainty. As a practical matter, however, the amount of primer added will generally be in molar excess over the amount of complementary strand (template) when the sequence to be amplified is contained in a mixture of complicated long-chain nucleic acid strands. a large molar excess is preferred to improve the efficiency of the process.

[0080] The deoxyribonucleoside triphosphates dATP, dCTP, dGTP, and dTTP are added to the synthesis mixture, either separately or together with the primers, in adequate amounts and the resulting solution is heated to about 90.degree.-100.degree. C. from about 1 to 10 minutes, preferably from 1 to 4 minutes. After this heating period, the solution is allowed to cool to approximately room temperature, which is preferable for the primer hybridization. To the cooled mixture is added an appropriate agent for effecting the primer extension reaction (called herein "agent for polymerization"), and the reaction is allowed to occur under conditions known in the art. The agent for polymerization may also be added together with the other reagents if it is heat stable. This synthesis (or amplification) reaction may occur at room temperature up to a temperature above which the agent for polymerization no longer functions. Thus, for example, if DNA polymerase is used as the agent, the temperature is generally no greater than about 40.degree. C. Most conveniently the reaction occurs at room temperature.

[0081] The agent for polymerization may be any compound or system which will function to accomplish the synthesis of primer extension products, including enzymes. Suitable enzymes for this purpose include, for example, E. coli DNA polymerase I, Klenow fragment of E. coli DNA polymerase I, T4 DNA polymerase, other available DNA polymerases, polymerase muteins, reverse transcriptase, and other enzymes, including heat-stable enzymes (i.e., those enzymes which perform primer extension after being subjected to temperatures sufficiently elevated to cause denaturation such as Taq DNA polymerase, and the like). Suitable enzymes will facilitate combination of the nucleotides in the proper manner to form the primer extension products which are complementary to each locus nucleic acid strand. Generally, the synthesis will be initiated at the 3' end of each primer and proceed in the 5' direction along the template strand, until synthesis terminates, producing molecules of different lengths. There may be agents for polymerization, however, which initiate synthesis at the 5' end and proceed in the other direction, using the same process as described above.

[0082] Preferably, the method of amplifying is by PCR, as described herein and as is commonly used by those of ordinary skill in the art. However, alternative methods of amplification have been described and can also be employed. PCR techniques and many variations of PCR are known. Basic PCR techniques are described by Saiki et al. (1988 Science 239:487-491) and by U.S. Pat. Nos. 4,683,195, 4,683,202 and 4,800,159, which are incorporated herein by reference.

[0083] The conditions generally required for PCR include temperature, salt, cation, pH and related conditions needed for efficient copying of the master-cut fragment. PCR conditions include repeated cycles of heat denaturation (i.e. heating to at least about 95.degree. C.) and incubation at a temperature permitting primer: adaptor hybridization and copying of the master-cut DNA fragment by the amplification enzyme. Heat stable amplification enzymes like the pwo, Thermus aquaticus or Thermococcus litoralis DNA polymerases are commercially available which eliminate the need to add enzyme after each denaturation cycle. The salt, cation, pH and related factors needed for enzymatic amplification activity are available from commercial manufacturers of amplification enzymes.

[0084] As provided herein an amplification enzyme is any enzyme which can be used for in vitro nucleic acid amplification, e.g. by the above-described procedures. Such amplification enzymes include pwo, Escherichia coli DNA polymerase I, Klenow fragment of E. coli DNA polymerase I, T4 DNA polymerase, T7 DNA polymerase, Thermus aquaticus (Taq) DNA polymerase, Thermococcus litoralis DNA polymerase, SP6 RNA polymerase, T7 RNA polymerase, T3 RNA polymerase, T4 polynucleotide kinase, Avian Myeloblastosis Virus reverse transcriptase, Moloney Murine Leukemia Virus reverse transcriptase, T4 DNA ligase, E. coli DNA ligase or Q.beta.replicase. Preferred amplification enzymes are the pwo and Taq polymerases. The pwo enzyme is especially preferred because of its fidelity in replicating DNA.

[0085] Once amplified, the nucleic acid can be attached to a solid support, such as a membrane, and can be hybridized with any probe of interest, to detect any nucleic acid sequence. Several membranes are known to one of skill in the art for the adhesion of nucleic acid sequences. Specific non-limiting examples of these membranes include nitrocellulose (NITROPURE) or other membranes used in for detection of gene expression such as polyvinylchloride, diazotized paper and other commercially available membranes such as GENESCREEN, ZETAPROBE (Biorad), and NYTRAN Methods for attaching nucleic acids to these membranes are well known to one of skill in the art. Alternatively, screening can be done in a liquid phase.

[0086] In nucleic acid hybridization reactions, the conditions used to achieve a particular level of stringency will vary, depending on the nature of the nucleic acids being hybridized. For example, the length, degree of complementarity, nucleotide sequence composition (e.g., GC v. AT content), and nucleic acid type (e.g., RNA v. DNA) of the hybridizing regions of the nucleic acids can be considered in selecting hybridization conditions. An additional consideration is whether one of the nucleic acids is immobilized, for example, on a filter.

[0087] An example of progressively higher stringency conditions is as follows: 2.times.SSC/0.1% SDS at about room temperature (hybridization conditions); 0.2.times.SSC/0.1% SDS at about room temperature (low stringency conditions); 0.2.times.SSC/0.1% SDS at about 42.degree. C. (moderate stringency conditions); and 0.1.times.SSC at about 68.degree. C. (high stringency conditions). Washing can be carried out using only one of these conditions, e.g., high stringency conditions, or each of the conditions can be used, e.g., for 10-15 minutes each, in the order listed above, repeating any or all of the steps listed. However, as mentioned above, optimal conditions will vary, depending on the particular hybridization reaction involved, and can be determined empirically. In general, conditions of high stringency are used for the hybridization of the probe of interest.

[0088] The probe of interest can be detectably labeled, for example, with a radioisotope, a fluorescent compound, a bioluminescent compound, a chemiluminescent compound, a metal chelator, or an enzyme. Those of ordinary skill in the art will know of other suitable labels for binding to the probe, or will be able to ascertain such, using routine experimentation.

[0089] In one embodiment, representational difference analysis (RDA, see Lisitsyn et al., Science 259:946-951, 1993, herein incorporated by reference) can be performed on CpG-containing nucleic acid following MCA. MCA utilizes kinetic and subtractive enrichment to purify restriction endonuclease fragments present in one population of nucleic acid fragments but not in another. Thus, RDA enables the identification of small differences between the sequences of two nucleic acid populations. RDA uses nucleic acid from one population as a "tester" and nucleic acid from a second population as a "driver" in order to clone probes for single copy sequences present in (or absent from) one of the two populations. In one embodiment, nucleic acid from a "normal" individual or sample, not having a disorder such as a cell-proliferative disorder is used as a "driver," and nucleic acid from an "affected" individual or sample, having the disorder such as a cell proliferative disorder is used as a "tester." In one embodiment, the nucleic acid used as a "tester" is isolated from an individual having a cell proliferative disorder such as low grade astrocytoma, anaplastic astrocytoma, glioblastoma, medulloblastoma, gastric cancer, colorectal cancer, colorectal adenoma, acute myelogenous leukemia, leukemia, lung cancer, renal cancer, breast cancer, prostate cancer, endometrial cancer and neuroblastoma. The nucleic acid used as a "driver" is thus normal astrocytes, normal glial cells, normal brain cells, normal gastric cells, normal colorectal cells, normal leukocytes, normal lung cells, normal kidney cells, normal breast cells, normal prostate cells, normal uterine cells, and normal neurons, respectively. In an additional embodiment, the nucleic acid used as a "driver" is isolated from an individual having a cell proliferative disorder such as low grade astrocytoma, anaplastic astrocytoma, glioblastoma, medulloblastoma, gastric cancer, colorectal cancer, colorectal adenoma, acute myelogenous leukemia, leukemia, lung cancer, renal cancer, breast cancer, prostate cancer, endometrial cancer and neuroblastoma. The nucleic acid used as a "tester" is thus normal astrocytes, normal glial cells, normal brain cells, normal gastric cells, normal colorectal cells, normal leukocytes, normal lung cells, normal kidney cells, normal breast cells, normal prostate cells, normal uterine cells, and normal neurons, respectively. One of skill in the art will readily be able to identify the "tester" nucleic acid useful with to identify methylated nucleic acid sequences in given "driver" population.

[0090] The materials for use in the assay of the invention are ideally suited for the preparation of a kit. Therefore, in accordance with another embodiment of the present invention, there is provided a kit it useful for the detection of a cellular proliferative disorder in a subject having or at risk for said cellular proliferative disorder. Invention kits include a carrier means compartmentalized to receive a sample in close confinement therein, one or more containers comprising a first container containing a reagent which modifies unmethylated cytosine and a second container containing primers for amplification of a CpG-containing nucleic acid, wherein the primers distinguish between modified methylated and nonmethylated nucleic acid, and optionally, a third container containing a methylation sensitive restriction endonuclease. Primers contemplated for use in accordance with the invention include those that would amplify sequences or fragments thereof as set forth in SEQ ID NOs: 1-42.

[0091] Carrier means are suited for containing one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method. In view of the description provided herein of invention methods, those of skill in the art can readily determine the apportionment of the necessary reagents among the containiner means. For example, one of the container means can comprise a container containing an oligonucleotide for ligation to nucleic acid cleaved by a methylation sensitive restriction endonuclease. One or more container means can also be included comprising a primer complementary to the oligonucleotide. In addition, one or more container means can also be included which comprise a methylation sensitive restriction endonuclease. One or more container means can also be included containing an isoschizomer of said methylation sensitive restriction enzyme.

[0092] It should be noted that as used herein and in the appended claims, the singular forms "a," "and," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a cell" includes a plurality of such cells and reference to "the restriction enzyme" includes reference to one or more restriction enzymes and equivalents thereof known to those skilled in the art, and so forth.

[0093] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described.

[0094] All publications mentioned herein are incorporated herein by reference in full for the purpose of describing and disclosing the methodologies which are described in the publications which might be used in connection with the presently described invention. The publications discussed above and throughout the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention.

[0095] The following example is intended to illustrate but not to limit the invention in any manner, shape, or form, either explicitly or implicitly. While they are typical of those that might be used, other procedures, methodologies, or techniques known to those skilled in the art may alternatively be used.

EXAMPLE 1

Collection and Preparation of Pancreatic Cell Lines

[0096] The following pancreatic adenocarcinoma cell lines were used: PL3 and PL8 (both CIMP-cell lines (CpG island methylator phenotype) were from Dr. Elizabeth Jaffe, John Hopkins University); CAPAN1, CAPAN2, Panc1, Hs766T, and MiaPaca2 (from the American Type Culture Collection, Rockville, Md.) and Colo357 (from the European Collection of Animal Cell Cultures, Salisbury, United Kingdom). In addition, seventeen pancreatic cancer xenografts were selected at random from a total of 90 xenografts, which were established from the primary carcinomas as described in Ueki, et al., Cancer Res., 60:1835-1839, 2000. Forty-seven primary pancreatic adenocarcinomas, 15 normal pancreata, 5 pancreata from patients with chronic pancreatitis, and a panel of normal tissues were obtained from the resected surgical specimens at The Johns Hopkins Medical Institutions, Baltimore, Md. Frozen tissues or paraffin-embedded tissues were microdissected to obtain >40% neoplastic cellularity in the primary pancreatic adenocarcinomas, and 3 of the 15 frozen normal pancreatic tissues were also microdissected to enrich the normal ductal epithelium. DNA was extracted from microdissected primary pancreatic adenocarcinomas and normal tissues as well as from lymphocytes of four cancer-free individuals using standard methods.

EXAMPLE 2

Methylated CpG Island Amplification/Representational Difference Analysis (MCA/RDA)

[0097] MCA/RDA was performed as described by Toyota et al., Cancer Res., 59:2307-2312, 1999, modifying such procedure to increase the efficiency by digesting 5 .mu.g of DNA with SmaI and XmaI (New England Biolabs). The restriction fragments were then ligated to RMCA adapter and amplified by PCR in 10 mM Tris-HCl (pH 8.3), 1.5 mM MgCl2, 50 mM KCl, 0.5 M betaine, 2% DMSO, 200 .mu.M each deoxynucleotide triphosphate, 100 .mu.mol of RMCA 24mer primer and 15 units of Taq polymerase (Life Technologies, Inc.) in a final reaction volume of 100 .mu.l. The reaction mixture was then incubated at 72.degree. C. for 5 min and at 95.degree. C. for 3 min, and then subjected to 25 cycles of 1 min at 95.degree. C. and 3 min at 77.degree. C. followed by a final extension of 10 min at 77.degree. C. Betaine was included in the PCR reaction to help amplify the methylated templates at a higher annealing temperature (77.degree. C.). The combination of betaine and DMSO can uniformly amplify a mixture of DNA with different GC content (Baskaran, et al., Genome Res. 6:633-638, 1996). These modifications might have enhanced the amplification of distinct MICPs (methylated in carcinoma of the pancreas) instead of Alu repetitive sequences that accounted for 60% of the recovered clones using the original protocol (Toyota et al., Cancer Res. 59:2307-2312, 1999). The MCA amplicon from either the pancreatic cancer cell line PL3 or PL8 was used as the tester for RDA, and a MCA amplicon generated from a mixture of DNA from the normal pancreata of six different patients was used as the driver. RDA was performed on these MCA amplicons using different adapters, JMCA and NMCA. Sequences of adapters used for MCA/RDA are listed in Table 3A and 3B and are available at http://pathology2.jhu.edu/pancreas/prim0425.htm, which website is incorporated herein by reference. After the third round of competitive hybridization and selective amplification, the RDA difference products of second and third round amplifications were cloned into pbluescript II plasmid vector (Stratagene).

EXAMPLE 3

DNA Sequencing of Clones and Dot Blot Hybridization

[0098] The clones recovered from each cell line after MCA/RDA were amplified with T3 and T7 primers and then sequenced using KS primer as recommended by the manufacturer (Sequitherm Excel; Epicentre Technologies). To determine the methylation status of MCA/RDA MICPs in pancreatic cancer and normal pancreas, MICPs were screened by hybridizing them to a dot blot of MCA products of pancreatic cancers and normal pancreata. Plasmid DNA containing each independent clone was prepared and digested with SmaI. DNA fragments were recovered from agarose gel and used as a probe for dot blot hybridization. Aliquots (1 .mu.l) of the mixture of 10.times.SSC and MCA products from the driver and from the tester (PL3 and PL8) both before and after each of the three rounds of RDA competitive hybridization/selective amplification were blotted onto nylon membranes in duplicate. Similarly, MCA products from six pancreatic cell lines (CAPAN1, CAPAN2, Panc1, Hs766T, MiaPaca2, and Colo357) and from eight other normal pancreata were also blotted onto the membranes. The membranes were hybridized with 32P-labeled probes overnight, washed, and exposed to a Kodak X-ray film.

EXAMPLE 4

Bisulfite Modification, Bisulfite-Modified Genomic Sequencing, and Methylation-Specific PCR (MSP)

[0099] The bisulfite treatment was carried out for 16 h at 50.degree. C. using 1 .mu.g of genomic DNA, as described in Ueki, et al., Cancer Res., 60:1835-1839, 2000. Genomic sequencing was performed on bisulfite-treated DNA to examine the methylation status of 10-20 CpG dinucleotides located in and/or around SmaI sites of each clone in 22 pancreatic tissues (8 cancer cell lines, 6 primary adenocarcinomas, and 8 normal pancreata; Ueki, et al., Cancer Res. 60:1835-1839, 2000). Genomic sequencing of the coding sequence of cyclin G was also performed in PL8. The level of methylation of each clone was determined by quantifying the level of methylation of each CpG site by comparing the intensity of unconverted cytosine with that of cytosine plus thymidine. Generally, in pancreatic cancer cell lines, the level of methylation observed at each CpG dinucleotide was consistent throughout the CpG island. Therefore, the average level of methylation of each sequence was graded and placed into one of 4 grades: 0-10%, 11-30%, 31-70%, and 71-100%. MSP was performed as described in Herman, et al., Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996, and to acquire optimal specificity, each primer pair contained four to six CpG sites, and high specific annealing temperatures were used. The primers and the specific annealing temperatures for each clone are listed in Table 3 and are available at http://pathology2jhu.edu/pancreas/prim042- 5.htm, which website is incorporated herein by reference. If validated MSP primers sets specific for methylated and unmethylated templates revealed that there was only amplification of methylated templates, the samples are presumed to be 100% methylated. Methylated and unmethylated templates were identified by bisulfite-modified sequencing. In describing MSP results performed on CpG islands that were normally unmethylated in non-neoplastic pancreas, a cancer sample was termed "methylated" if MSP yielded any methylated templates.

EXAMPLE 5

Reverse Transcription-PCR (RT-PCR) and 5-aza-2'-deoxycytidine (5Aza-dC) Treatment

[0100] Five pancreatic cancer cell lines (PL3, PL8, CAPAN2, Panc1, and MiaPaca2) and four normal pancreata were used for RT-PCR analysis. The cell lines were treated with demethylating agent 5Aza-dC (Sigma Chemical Co.) at a final concentration of 1 .mu.M for 5 days. Total RNA was prepared using TRIzol (Life Technologies, Inc.), reverse-transcribed and amplified. As a control for cDNA integrity, glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) was also amplified. Primer sequences for RT-PCR are listed in Table 3 and are available at http://pathology2 jhu.edu/pancreas/prim0425.htm, which website is incorporated herein by reference.

EXAMPLE 6

Statistics

[0101] The primary outcome variable was the observed number of 7 MICP loci found to be methylated in 64 pancreatic cancers. Wilcoxon's rank-sum test compared the observed number of methylated loci by tumor differentiation (poorly versus well or moderately differentiated), lymph node status (0 or 1 versus>1 node positive), and prior CIMP classification (CIMP positive versus CIMP negative). Simple linear regression assessed the relationship between the observed number of methylated loci and these covariates: age, age squared, and tumor diameter (in cm). Multivariate linear regression assessed the simultaneous contribution of the clinicopathological and demographic variables to the observed number of methylated loci. All of the tests were two-sided. A P of <0.05 signified statistical significance.

EXAMPLE 7

Patient Population and Tissue Samples

[0102] Normal and tumor specimens were obtained from pancreatic adenocarcinomas resected at The Johns Hopkins Hospital. Pancreatic cancer xenografts were established from the primary carcinomas as previously described in Caldas, et al., Nature Genet. 8:27-31, 1994, and carcinoma and normal tissues were stored at -70.degree. C. Thirty-two xenografts were selected at random. Three MSI carcinoma xenografts were added, as reported previously, as well as another MSI primary carcinoma (Goggins, et al., Am. J. of Pathology 152:1501-1507, 1998). Genomic DNA was prepared from 35 xenografts and 9 pancreatic adenocarcinoma cell lines (BxPc3, Capan1, Capan2, Panc1, CFPAC1, MiaPaca2, Hs766T (all from ATCC, Rockville, Md.), Co10357 (from ECACC, Salisbury, UK) and PL45, a low passage cell line established in the Goggin Laboratory at John Hopkins University School of Medicine). Where available, DNA was obtained from primary pancreatic cancer tissue frozen at -80 C since Whipple resection. Frozen tissues were microdissected to obtain DNA from normal pancreatic tissue and the pancreatic carcinoma.

[0103] Patient records were reviewed to determine a history of cigarette smoking, diabetes mellitus, and prognosis, and a family history of pancreatic cancer. These data were reviewed in the context of the DNA methylation data.

EXAMPLE 8

Bisulfite Modification and Genomic Sequencing

[0104] The bisulfite treatment was carried out for 16 h at 56.degree. C. on 1 .mu.g of genomic DNA, according to the procedure of Herman et al, Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996. Modified DNA was purified and eluted into 50 .mu.l of LoTE buffer. The primers used for genomic sequencing of bisulfite-treated DNA are listed in Table 3 and are available at http://pathology2 jhu.edu/pancreas/prim0425.htm, which website is incorporated herein by reference. PCR was performed on 1-2 .mu.l of bisulfite-treated DNA, and prior to sequencing, PCR reactions were incubated with exonuclease I and shrimp alkaline phosphatase (Amersham, according to the manufacturer recommendations). Sequencing of PCR products was performed in microtiter plates as recommended by the manufacturer (Sequitherm Excel, Epicentre Technologies, Madison, Wis.

EXAMPLE 9

Methylation-Specific PCR (MSP) Assay

[0105] The methylation status of each gene was determined by MSP as described by Herman et al, Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996, in which the sequence difference of bisulfite-treated DNA was detected by PCR using primers specific for either the methylated or for the unmethylated DNA. Primer sequences and the specific annealing temperatures for 13 genes are listed in Table 3 and are available at http://pathology2 jhu.edu/pancreas/prim0425.htm, which website is incorporated herein by reference. MSP was performed on 1 .mu.l of bisulfite-treated DNA under the conditions as follows: 95.degree. C. for 3 min; then 35-40 cycles of 95.degree. C. for 30 s, the specific annealing temperature for 30 s, and 72.degree. C. for 30 s; and a final extension of 4 min at 72.degree. C. Five .mu.l of each PCR product were directly loaded onto 3% agarose gels or 10% acrylamide gels, stained with ethidium bromide and visualized under UV illumination. All PCR reactions were performed with positive controls for both unmethylated and methylated alleles, and no DNA control. Finally, 3-6 CpG sites were included in each primer pair and the specific annealing temperatures were used for each gene to obtain optimal specificity.

EXAMPLE 10

Identification of Differentially Methylated Sequences

[0106] The strategy of MCA/RDA has been previously reported (Toyota, et al., Cancer Res. 59:2307-2312, 1997). Ninety-six randomly selected clones recovered from each cell line were subjected to DNA sequencing and 66 clones were revealed to be independent. Only 3 clones contained Alu-repetitive sequences. The subsequent probing of labeled clones to MCA products of tester and driver by dot-blot hybridization revealed that 43 of 66 clones (MICP1-43) were differentially methylated in the tester compared to the driver (FIG. 1). These 43 clones were also variably methylated in the other 6 pancreatic cancer cell lines examined. A description of these 43 differentially methylated clones is shown in Table 1. All of the 43 clones had a GC content of greater than 50% and 41 (95%) had a sequence uniqueness sustaining the criteria of CpG island (Gardiner-Garden & Frommer, J. Mol. biol. 196:261-282, 1987). The DNA homology search of each clone with the BLAST program (National Center for Biotechnology Information) demonstrated that 84% (36) of the 43 clones had significant homologies to known human sequences, including 8 clones matched to human gene sequences and 9 clones matched to human ESTs. Five clones were also matched or contained a part of CpG islands isolated previously (Toyota, et al., Cancer Res. 59:2307-2312, 1997, Cross, et al., Nat. Genet., 6:236-244, 1994) and 14 clones had significant homology to High-Throughput Genome Sequences in the three International Nucleotide Sequence databases: DDBJ, EMBL and GenBank. The remaining 7 had no significant homology to known sequences. MICP1 corresponded to the 3' noncoding region of the human homeobox gene CSX, MICP2 corresponded to exon 1 and intron 1 of the human Cyclin G, MICP3 corresponded to 5' region of the human endothelin-converting enzyme-like I gene ECEL1, MICP4 corresponded to intron 1 of the human glutamate decarboxylase 1 gene GAD I, MICP5 corresponded to exon 7 to exon 9 of the human ICAM5, MICP6 corresponded to intron 2 to intron 3 of the human monocarboxylate transporter 3 gene MCT3, MICP7 corresponded to the 5' region of the human B-cell specific transcriptional factor gene PAX5, MICP8 corresponded to intron 1 and exon 2 of the human Preproenkephalin gene (ppENK) (FIG. 2). Interestingly, 3 clones (MICP1, 17 and 22) matched to CpG islands originally recovered from colorectal cancer cell line using the same technique (named MINT 23, 20 and 32, respectively) (Toyota, et al., Cancer Res. 59:2307-2312, 1997).

EXAMPLE 11

Characterization of the Sequences

[0107] For 30 of the 43 clones, methylation was detected in 2 or more out of 8 normal pancreata by dot blot analysis, suggesting that these clones could be frequently methylated in normal pancreas. Therefore, only the remaining 13 clones were further analyzed by bisulfite sequencing. For seven of the clones (Cyclin G, ppENK, MICP20, 23, 33, 35 and 36), methylation was restricted to pancreatic cancers (FIGS. 4A and 4B). In the case of 5 clones (MCT3, PAX5, MICP15, 16 and 38), methylation was detectable in DNA from normal pancreata as well as DNA from cancer tissues (FIG. 3C). In the thirteenth clone (MICP42), cytosines at CpG sites were similarly methylated in both pancreatic cancers and normal pancreata. The sequence uniqueness of this clone also did not satisfy the criteria for CpG islands. A summary of the level of methylation of each clone is shown in FIG. 3D. There was little individual variation in the level of methylation of these-6 clones (MCT3, PAX5, MICP15, 16, 38 and 42) in normal pancreata from 8 patients at the age from 34 to 84 years old (FIG. 3 and data not shown). Interestingly, the methylation of these CpG islands in normal pancreata was sometimes heterogeneous. For example, although the SmaI site of MICP 15 was not methylated in normal pancreata, 2 CpG sites near the SmaI site were methylated (FIG. 3C). A summary of the results of MCA/RDA and bisulfite sequencing is also provided in FIG. 4.

[0108] In order to identify low level of methylation of identified clones in pancreatic tissues, MSP primers were designed for the 7 clones (Cyclin G, ppENK, MICP20, 23, 33, 35 and 36) differentially methylated in pancreatic cancers by bisulfite sequencing. The methylation status of these clones was examined in 15 primary pancreatic adenocarcinomas, 5 DNA samples from pancreata with chronic pancreatitis as well as 14 normal pancreata including three specimens enriched in normal ductal epithelium. For MICP36, MSP detected methylation in DNA of three pancreatic cancer cell lines but not in any other specimens examined. Aberrant methylation of the remaining 6 clones was detected in 7% to 87% of primary pancreatic adenocarcinomas, while MSP confirmed a lack of methylation of these 6 clones in DNA from 14 normal pancreata (FIG. 5 and Table 2A and 2B). DNA from one chronic pancreatitis tissue containing pancreatic intraductal neoplasia (PanIN) (Hruban, et al., Clin. Cancer Res. 6:2969-2972, 2000) harbored methylated templates of Cyclin G, ppENK and MICP20 and DNA from another chronic pancreatitis sample also had methylated templates of ppENK.

[0109] Because tissue-specific methylation differences can be found in normal tissues (Liang, et al., Genomics 53:260-268, 1998), the clones found that were aberrantly methylated in pancreatic cancer, as compared to normal pancreas, were also analyzed to see if they are methylated in other normal tissues. By MSP, MICPs 33 and 35 were not methylated in any normal gastrointestinal tissues, while ppENK, MICP20 and 23 were methylated in DNA samples from normal gastric, duodenal and colonic mucosae (FIG. 5 and Table 2A and 2B). Amplification of methylated templates of these clones was always weak in normal mucosae compared to the primary pancreatic adenocarcinomas (FIG. 3), suggesting that there were few methylated DNA templates in these mucosae.

EXAMPLE 12

Expression of Cyclin G and ppENK in Pancreatic Cancer

[0110] To determine whether methylation of these clones resulted in loss or decrease of gene expression, Cyclin G and ppENK were further examined using RT-PCR in 5 and 4 pancreatic cell lines, respectively. Partial methylation (-50%) of the 5' CpG island of Cyclin G in PL8 (FIG. 4C) was associated with decreased expression of Cyclin G by RT-PCR. The 5' CpG island of Cyclin G was not methylated in a panel of normal pancreata. Cyclin G was expressed in 4 normal pancreata by RT-PCR. Treatment with 5Aza-dC restored the expression of Cyclin G in PL8 (FIG. 4C). Using RT-PCR, it was found that ppENK was expressed in normal pancreata but not in any of the 4 pancreatic cancer cell lines examined in which 5'CpG island of this gene was methylated. 5Aza-dC treatment restored ppENK expression in all 4 cell lines (FIG. 4D). Thus, hypermethylation of the 5' CpG islands of Cyclin G and ppENK is coincides with decreased expression in pancreatic cancers.

EXAMPLE 13

Determination of Methylated ppENK by Methylation Specific PCR of Pancreatic Fluid as a Specific Marker of Pancreatic Cancer

[0111] Pancreatic fluid obtained from patients undergoing pancreatic surgical resection from 38 patients with pancreatic cancer, 7 with chronic pancreatitis, 4 with miscellaneous tumors (islet cell, serous cyst adenoma, and lymphoma), 9 with ampullary cancers, 9 with IPMNs (intraductal papillary mucinous neoplasms) and 3 from bile duct cancers was analyzed. Methylated ppENK was detected in 55% of pancreatic cancer fluids, none in the chronic pancreatitis or other miscellaneous tumor group, 2 of 9 with IPMN, 2 of 3 with bile duct cancer, and 4 of 9 with ampullary cancer, suggesting that the specificity of methylated ppENK in pancreatic fluid is useful as a diagnostic marker.

[0112] Although the invention has been described with reference to the presently preferred embodiment, it should be understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims.

1TABLE 1A The 42 MICPs identified by MCA/RDA in pancreatic cancers* CpG MICP Blast homology Size (bp) Islands** Chromosome 1 CSX 414 yes 5q35 2 Cyclin G 392 yes 5q32-34 3 FLJ00083 479 no not known 4 GADI 309 yes 2 5 HLH 179 yes 20 6 ICAM5 630 yes 19p13.2 7 MCT3 308 yes 22q12.3-q31.2 8 PAX5 548 yes 9p13 9 ppENK 255 yes 8q23-q24 10 SMO 229 yes 7q34-q36 11 ZBP 399 yes 5q22 12 Human EST 431 yes Xp22 13 Human EST 600 yes 3p 14 Human EST 510 yes 5 15 Human EST 359 yes not known 16 Human EST 370 yes 12q 17 Human EST 324 yes 6q 18 Human EST 415 yes 11q 19 Human EST 392 yes not known 20 Human EST 359 yes 11q14 21 Human EST 545 yes 19q13.3 22 Human CpG island 386 yes 7q34-q36 23 Human CpG island 464 yes 20q13 24 Human CpG island 526 yes 15 25 Human CpG island 280 yes 6p22.1-23 26 Human CpG island 422 yes 1 27 Human genomic DNA 257 yes 17 28 Human genomic DNA 350 yes 7q34-q36 29 Human genomic DNA 336 yes 10 30 Human genomic DNA 293 yes 2q36-q37 31 Human genomic DNA 427 yes 13 32 Human genomic DNA 470 no 21q22.3 33 Human genomic DNA 264 yes Xq22.1-23 34 Human genomic DNA 371 yes 20p11.21- 11.23 35 Human genomic DNA 445 yes 4 36 Human genomic DNA 357 yes 19q13.1 37 Human genomic DNA 312 yes 7p14-15 38 Human genomic DNA 372 yes 5 39 No homology 307 yes not known 40 No homology 331 yes not known 41 No homology 304 yes not known 42 No homology 307 yes not known *7 of these 42 MICPs were methylated in pancreatic cancer and not normal pancreas (see text) **minimal length, 200 Bp; GC content, >50%; CpG/GpC, >0.5(11)

[0113]

2TABLE 1B MICP* Size % GC CpG/GpC CpG Islands? NCBI Acc. #** Location of clone*** 1 414 64.3 0.811 yes AF135523 1-414 2 392 62.8 0.613 yes D86077 35-426 3 479 63.9 0.444 no AK024484 1645-2107 4 309 64.4 0.727 yes AC007405 72554-72862 5 179 68.3 1.330 yes AL121673 66270-66448 6 630 65.6 0.625 yes AF082802 5502-6131 7 308 66.6 0.704 yes AF132611 1082-1389 8 548 60.4 0.683 yes AL161781 129660-130226 9 255 67.8 0.867 yes V00509 622-876 10 229 58.2 0.927 yes AC083866 37615-37843 11 399 59.1 0.800 yes AC016663 106325-106723 12 431 68.4 0.909 yes AC002365 159360-159790 13 600 65.5 0.661 yes AC024167 62307-62906 14 510 63.7 0.771 yes AF135520 1-510 15 359 65.5 0.972 yes AW028607 259-376 16 370 64.9 1.030 yes AC026336 11182-11568 17 324 67.6 0.816 yes AC068247 66045-66347 18 415 61.9 0.767 yes AP002781 190243-190537 19 392 55.1 1.214 yes AI557774 193-392 20 359 55.6 0.731 yes AP002354 58648-59006 21 545 65.0 1.023 yes AC007785 13354-13898 22 386 66.3 0.605 yes AC005998 48200-48585 23 464 65.7 0.975 yes AF135532 1-464 24 526 64.2 0.645 yes AC074201 59279-59804 25 280 68.1 0.929 yes AL136305 115130-115409 26 422 63.0 0.767 yes AL137797.1 38754-39186 27 257 65.8 0.852 yes AC004706 88868-89124 28 350 64.9 0.900 yes AC004897 60876-61125 29 336 64.4 0.606 yes AC005661 43755-44090 30 293 67.0 0.760 yes AC012033 113181-113473 31 427 66.7 0.884 yes AC013721 31973-32339 32 470 59.6 0.292 no AJ239326 83971-84440 33 264 63.3 0.580 yes AL035088 102596-102859 34 371 63.0 0.722 yes AL080312 56645-57015 35 445 64.0 0.884 yes AC023868 130032-030491 36 357 68.9 0.800 yes AC002133 31977-32333 37 312 67.5 0.885 yes AC005826 151694-152005 38 372 65.6 0.778 yes AC011376 56983-57313 39 307 68.7 0.844 yes no homology 40 331 70.7 0.707 yes no homology 41 304 68.8 0.794 yes no homology 42 307 68.7 0.875 yes no homology *Total 4 clones with no homology to known human sequences (MICP38-42) and 38 clones matched $$ 11 clones matched to the human genes (MICP1-11) 10 clones matched to human ESTs (MICP12-21)

[0114]

3TABLE 2A Tumor no. CpG Island Sample age Diam.(cm) nodes methylated ppENK MICP27 MICP38 MICP25 MICP36 CyclInG zbp AsPC1 3 1 2 3 BxPC3 3 4 5 6 CAPAN1 3 7 8 9 CAPAN2 3 10 11 12 CFPAC 2 13 14 Colo357 5 15 16 17 18 19 20 Hs766T 3 21 22 23 MlaPaca2 4 24 25 26 27 28 Panc1 2 29 30 PL3 5 31 32 33 34 35 PL8 4 36 37 38 39 40 41 Px17 69 4.4 + 2 42 43 44 Px29 66 2.5 + 3 45 46 47 48 Px30 57 3.0 + 0 49 Px64 76 2.5 + 1 50 Px65 63 2.2 + 1 51 Px74 83 3.5 + 2 52 53 Px75 63 5.0 0 2 54 55 56 Px76 64 2.2 + 3 57 58 59 Px102 47 3.0 0 1 60 Px108 76 3.0 + 2 61 62 Px120 70 5.0 + 4 63 64 65 66 Px132 61 3.6 + 3 67 68 69 Px143 54 2.0 + 1 70 Px184 59 4.0 + 4 71 72 73 74 Px186 55 4.0 + 2 75 76 Px195 67 4.0 + 0 Px282 79 3.0 0 2 77 78 1 64 4.7 0 1 79 2 67 3.0 + 2 80 81 4 54 2.7 + 2 82 83 6 69 4.5 + 2 84 85 9 85 8.0 + 4 86 87 88 89 11 65 2.5 + 0 12 81 10.0 + 2 90 91 13 53 + 1 92 14 57 3.0 + 2 93 94 95 196 84 6.0 + 4 96 97 98 99 198 76 4.0 + 3 100 101 102 240 50 3.5 0 0 248 67 2.0 + 1 103 281 36 6.0 + 1 104 287 72 2.5 0 1 105 666 34 8.0 + 4 106 107 108 109 5350 44 4.0 + 0 11632 66 6.5 + 2 110 111 12616 58 8x 1 112 13671 58 6.0 + 1 113 13653 63 14.5 + 3 114 115 116 17496 73 3.5 + 3 117 118 119 20852 78 5.0 + 2 120 121 21190 82 3.5 + 3 122 123 124 21889 60 4.0 + 3 125 126 127 22384 74 2.0 + 3 128 129 130 24494 78 3.5 + 1 131 24721 71 2.5 + 2 132 133 25392 57 3.0 + 1 134 26183 56 1.5 0 2 135 136 30011 65 4.0 + 3 137 138 139 30845 76 2.6 + 3 140 141 142 31742 66 1.5 0 1 143 32006 61 0 2 144 145 35083 59 1.5 + 1 146 35583 75 3.0 + 1 147 36378 74 5.2 + 2 148 149 37680 43 4.0 0 1 150 42946 71 3.5 + 2 151 152 45885 53 8x 3 153 154 155 46391 79 2.3 + 2 156 157 49126 49 4.0 + 1 158 49390 79 2.5 + 3 159 160 161 52150 72 7.0 0 3 162 163 164 165 52225 58 3.5 + 3 166 167 168 53413 85 3.5 + 2 169 170 55090 71 4.5 + 1 171

[0115]

4TABLE 2B Results of MSP CLONE 2 CLONE 8 n* Cyclin G PPENK CLONE 20 CLONE 23 CLONE 33 CLONE 35 CLONE 36 Primary pancreatic carcinoma 15 7% 87% 13% 53% 33% 40% 0% Normal pancreata 14 0% 0% 0% 0% 0% 0% 0% Chronic pancreatitis 5 20% 40% 20% 0% 0% 0% ND Normal gastirc mucosae 6 ND ND 50% 17% 0% 0% ND Normal duodenal mucoase 8 ND 100%** 50% 75% 0% 0% ND Normal colonic mucosae 6 ND 100%*** 33% 100% 0% 0% ND Normal lymphocytes 4 0% 0% 0% 0% 0% 0% ND *number examined **only 4 samples examined ***only 3 samples examined

[0116]

5TABLE 3A PRIMER SEQUENCE FOR BISULFITE-SEQUENCE Annealing Clone Orientation Sequence Temperature SEQ ID NO: RAR.beta. Forward 5'-GAGTTGGTGATGTTAGATTAG-3' 56 43 Reverse 5'-TTCCCAAAAAAATCCCAAATTC-3' 44 Sequence 5'-CTCCTTCCAAATAAATACTTAC-3' 45 THBS1 Forward 5'-AGAGAGGAGTTTAGATTGG-3' 54 46 Reverse 5'-CAAAAAAACTAAAACCTCAAC-- 3' 47 Sequence Forward primer CACNA1G Forward 5'- 55, 53, 51, 48 TGGATAAAGGATGTTTGGGGTTTG- 49* 3' Reverse 5'-CCCTCCCCTTACCCCTAAATCC-3' 49 Sequence 5'-ACTCCCCCTCACTTTATTC-- 3' 50 hMLH1 Forward 5'- 58 51 ATTATTTTAGTAGAGGTATATAAG- 3' Reverse 5'-CCAACCCCACCCTTCAAC-3' 52 Sequence Forward primer MINT1 Forward 5'-AAGAGAGGGTTGGAGAGTAG-3' 62 53 Reverse 5'- 54 CCCCTAAAAAAAAAATCAAAAATC- 3' Sequence 5'-GGGTTGGAGAGTAGGGGAGTT-3' 55 MINT2 Forward 5'- 60, 58, 56, 56 YGTTATGATTTTTTTGTTTAGTTAAT 54** -3' Reverse 5'-TACACCAACTACCCAACTACCTC- 3' Sequence 5'-ACTTCCATTAAAAACAACTAC-3' 57 MINT31 Forward 5'- 58 58 TTTATTTATATAATTTTGTGTATGG- 3' Reverse 5'-CACCCCTCACTTTACTAAAAC-3' 59 Sequence Reverse primer MINT32 Forward 5'-TTTGGGAGGTAAATTYGTTGATT- 58, 56, 54, 60 3' 52*** Reverse 5'- 61 ACCRAACAAAAAACCTAAAAAAAC- 3' Sequence Forward primer *55 (5 cycles), 53 (5 cycles), 51 (5 cycles), 49 (26 cycles) **60 (3 cycles), 58 (4 cycles), 56 (5 cycles), 54 (26 cycles) ***58 (3 cycles), 56 (4 cycles), 54 (5 cycles), 52 (26 cycles)

[0117]

6TABLE 3B PRIMER SEQUENCES FOR MSP SEQ Orientation & Annealing ID Clone Methylation Sequence Temperature NO: P16 Unmethylated F 5'-TTATTAGAGGGTGGGGTGGA- TTGT-3' 60 62 R 5'-CAACCCCAAACCCACAACCATAA-3' Methylated F 5'-TTATTAGAGGGTGGGGCGGATCGC-3' 65 63 R 5'-GACCCCCGAACCGCGACCCT- AA-3' RAR.beta. Unmethylated F 5'-AGGATTGGGATGTTGAGAATG-3' 58 64 R 5'-TTACAAAAAACCTTCCAAATACA-3' Methylated F 5'-GGATTGGGATGTCGAGAAC-3' 64 65 R 5'-TACAAAAAACCTTCCGAATACG-3' CACNA1G Unmethylated F 5'-GTTTTTTTTTGGATTTTTGTTTTTTG-3' 60 66 R 5'-TTTATTCCAACTTCTTCACTTCA-3' Methylated F 5'-GTTTTTTCGGGGCGGTTTC-3' 62 67 R 5'-TTCCGACTTCTTCGCTTCG-3" TIMP-3 Unmethylated F 5'- 59 68 TTTTGTTTTGTTATTTTTTGTTTTTGGTT- TT-3 R 5'-CCCCCCAAAAACCCCACCTCA-3' Methylated F 5'- 59 69 CGTTTCGTTATTTTTTGTTTTCGGTTTTC- 3' R 5'-CCGAAAACCCCGCCTCG-3' THBS1 Unmethylated F 5'-GTTTGGTTGTTGTTTATTGGTTG-3' 62 70 R 5'-CCTAAACTCACAAACCAACTCA-- 3' 71 Methylated F 5'-TGCGAGCGTTTTTTTAAATGC-3' 62 72 R 5'-TAAACTCGCAAACCAACTCG-3' 73 HMLH1 Unmethylated F 5'-TTAATAGGAAGAGTGGATAGTG-3' 56 74 R 5'-TCTATAAATTACTAAATCTCTTCA- -3' 75 Methylated F 5'-TTAATAGGAAGAGCGGATAGC-3' 58 76 R 3'-CTATAAATTACTAAATCTCTTCG-3' 77 E-Cad Unmethylated F 5'-TAATTTTAGGTTAGAGGGTTATTGT-3' 53 78 R 5'-CACAACCAATCAACAACACA-- 3' 79 Methylated F 5'-TTAGGTTAGAGGGTTATCGCGT-3' 57 80 R 5'-TAACTAAAAATTCACCTACCGAC-3' 81 DAPK Unmethylated F 5'-GGAGGATAGTTGGATTGAGTTAATGTT- 60 82 3' R 5'-CAAATCCCTCCCAAACACCAA-3' 83 Methylated F 5'-GGATAGTCGGATCGAGTTAACGTC-3' 60 84 R 5'-CCCTCCCAAACGCCGA-3' 85 MGMT Unmethylated F 5'- 59 86 TTTGTGTTTTGATGTTTGTAGGT- TTTTGT- 3' R 5'- 87 AACTCCACACTCTTCCAAAAACAA- AACA- 3' Methylated F 5'-TTTCGACGTTCGTACCTTTTCGC-3' 59 88 R 5'-GCACTCTTCCGAAAACGAAACG-3' 89 MINT1 Unmethylated F 5'-GGGGTTGAGGTTTTTTGTTAGT-3' 64 90 R 5'-TTCACAACCTCAAATCTACTTC- A-3' 91 Methylated F 5'-GGGTTGAGGTTTTTTGTTAGC-3' 64 92 R 5'-CTACTTCGCCTAACCTAACG-3' 93 MINT2 Unmethylated F 5'-GGTGTTGTTAAATGTAAATAATTTG-3' 58 94 R 5'-AAAAAAAAACACCTAAAACTC- A-3' 95 Methylated F 5'-AATCGAATTTGTCGTCGTTTC-3' 60 96 R 5'-AAATAAATAAATAAAAAAAAACGCG-3' 97 MINT31 Unmethylated F 5'-GAATTGAGATGATTTTAATTTTTTGT-3' 64 98 R 5'-CTAAAACCATCACCCCTAAA- CA-3' 99 Methylated F 5'-TTGAGACGATTTTAATTTTTTGC-3' 62 100 R 5'-AAAACCATCACCCCTAAACG-3' 101 MINT32 Unmethylated F 5'-GAGTGGTTAGAGGAATTTAGGT-3' 62 102 R 5'-CTAAAAAAACAAACAAAACATCC- A-3' 103 Methylated F 5'-GTGGTTAGAGGAATTTAGGC-3' 64 104 R 5'-AAAACGAACGAAACGTCCG-3' 105

[0118]

Sequence CWU 1

1

118 1 414 DNA Homo sapiens 1 cccgggcgcc cggccctggc tcgcggaatg ggcggccaga tctcaggccc tgcgtgcccg 60 agctcagtcc cagttccaac cgggggtgcc catggactct cggagggcac tcctgggggg 120 acagctaaga caccaggctg caggatcact cattgcacgc tgcataatcg ccgccacaaa 180 ctctcccgtg cgcaagaaca aacgcgcgtg ggacagaaaa agttcctagg tctccgcagg 240 agtgaatgca aaatccaggg gactcagggt catgttggga gccccttctc cccccgagag 300 tcagggagct gttgaggtgg gatcggtgag ggtcgcgcca cgcgggtccc ttccctacca 360 ggctcggata ccatgcagcg tggacactcc cgagttgctc tgcggaatcc cggg 414 2 392 DNA Homo sapiens 2 cccggggagg gtggttaccg ctgaggagct gcagtctctg tcaaggtgag tgggactgcg 60 cgagagttga ccgccagtgc gggtggggag ctgggttggg ggcgcggggc gaggagtagg 120 tctggcccgc gcccttttcc acactaaact ctaccgctgt tgtgagcaca agcccaggct 180 agtccgaggc tggaggggcg gagcggatcc ggcctcctga ggtgcctttc gtgtctgccg 240 acccagtccc agggactagc ctggggagga agaatggaac ccctgcagtt agaggttcct 300 cacatgacta gctctgaaga cctcctgcct tcctgtcttt agttggtgtg ggagggacct 360 tccatgtatc cagggcttag cttgtgcccg gg 392 3 298 DNA Homo sapiens 3 cccggggaga gctgatcccg gacaggggtt ccgaggaggt aaaggaagct ccctggtgaa 60 ggtgagctct gcgcaggcgc tgtgcgggca gcccaggcac tccgtcagcg ccctctcaat 120 ccttaggagg agtcctccct gggtggtgct ccagggctgt gggctggacc atgacggccc 180 ccagctgcgt agctccagct cctttccaac cggttggtgg gcggttccga gtggggacac 240 gggatcggag gggactcctg ccgggtgtac ccgctaccct cactagtccc gggatgcg 298 4 309 DNA Homo sapiens 4 cccgggacag agagattgcg ggctaatctg ggtagatcga ggaccccaca gagaagggcc 60 caccggccat cgcctccaca ccctccctcc gacctcactc ctagccccgc gcgcgcagtc 120 gcacagcaac tcgggcagcg ggtcgactac ggcccctgga aaagtaacag gttaccgttc 180 ctagtagcgc cttcggctgc tgctgcccag gcgccctttg gagggacagg cgctcgcagc 240 atggaagctc aagggaaaat cgctcttgcc ccacttcaac tagagcctca gcctccgtgc 300 ttccccggg 309 5 630 DNA Homo sapiens 5 cccgggagca tgcgggcact taccgctgcg aagccaccaa ccctcggggc tctgcggcca 60 aaaatgtggc cgtcacggtg gaatgtgagt aggggcaccg cggagttagg caggatctgt 120 gggacaaccc cggctggact tcctggcccc cgtgtgagcc cctgcaatcc tgtttcccag 180 atggccccag gtttgaggag ccgagctgcc ccagcaattg gacatgggtg gaaggatctg 240 ggcgcctgtt ttcctgtgag gtcgatggga agccacagcc aagcgtgaag tgcgtgggct 300 ccgggggcac cactgagggg gtgctgctgc cgctggcacc cccagaccct agtcccagag 360 ctcccagaat ccctagagtc ctggcacccg gtatctacgt ctgcaacgcc accaaccgcc 420 acggctccgt ggccaaaaca gtcgtcgtga gcgcggagtg tgagcgaggc ccaggcgggt 480 agggagcagg gtgccccacg gtccaggcac tccctgacat cccccatggc tgttttgcag 540 cgccaccgga gatggatgaa tctacctgcc caagtcacca gacgtggctg gaaggggctg 600 aggcttccgc gctggcctgc gccgcccggg 630 6 308 DNA Homo sapiens 6 cccgggtggt gaccctgccc tgcccacagg ccccgtgtcc agcatcctcg tgacccgctt 60 tggctgtcgc ccggtgatgc tggcgggtgg gctgctggct tccgcgggca tgatcctagc 120 ttcctttgcc acgcgccttc tggagctcta cctgaccgct ggggtgctca caggtgaggg 180 ccccctggtc tcctctccgc tgggttgggg gtcgggggtt cttgctgcaa gatctgtcct 240 cggtttccct atgagggaca gtcttcgaag tccctcggct gggttcccgg atctgctggg 300 ttcccggg 308 7 548 DNA Homo sapiens 7 cccgggaatc cacagccaca tttccgattg tggcgaaatc tgctcagtgg ctctgcgatg 60 tccagttgcc gccgggggcc cccctttcct cgagctctag gctcttcccc ctaggttgcg 120 acccagcctc gtgaccaccc cctccaaaaa aacaaacaac actcttgctg aggacgattc 180 actctccaaa actgcattgt ccggcgggcc aggagtctct acggacgcgt cccgcctgag 240 acttccctcg agccacccgc tgcaggccca cggcttcagt ggctaggccc agcagctgaa 300 ccaactcaag gctggggggg aacaggaggg aggcttgagt ctggcccgaa gagagagggc 360 tggacatgcc acacctctgc tcggtctctg tggatctgat ttcctctctg gaatcaagtc 420 ctggggctct gggactccac aacgtctcag ggctcgaggg caatgcgatt ccacttacgg 480 gccggggtaa ggtgtctgga actcccgcca atctccagaa actactgaga tgttctgctc 540 tgcccggg 548 8 255 DNA Homo sapiens 8 cccgggaacc gcgaggcgat ctgagtcgcc tccacgtcta cctaaaagct gtcggccggg 60 agggcggggc cccagaaagg agcattcctg cgggcttttg ctcgacgatc ccctgctgag 120 gctgtcgcgg cgagggtcct gccgagggac cccgttctgc gcccaggcag gctcgaagca 180 cgcgtccctc tctcctcgca gtccatggcg cggttcctga cactttgcac ttggctgctg 240 ttgctcggcc ccggg 255 9 431 DNA Homo sapiens 9 cccgggcaga gctcggagcg cctccatccc cggaaccagg ggactccctg gagtgctccg 60 gtccaggcta cgatcgaggc gcccccatcc cttgggccca gggagaggat cggagacacc 120 aggaggccct cggggctggg tgaagatctt tggttccggg ggtccgggag aggatccacc 180 ctcccaatac cccgactccc agggctctga ccaagaatgg aggtgcccct tctccaggcc 240 tcgagccctc tgagcgccga ggccggccgc ctacaggtcc cccgccgctg ggcggaccct 300 ctcattcggt tccctcacgt cacccgctgt ccggcgcctg ggaactgggc tcctggaatt 360 tcctctcctg gggctgacag atggccctct tttcctttct ctgcggcagc ctcgcccatc 420 ccggtcccgg g 431 10 600 DNA Homo sapiens 10 cccggggacg gggagggagg agggctgccg ggatgtgaac cggggaaggc agctggggct 60 ggagagcagc gcggaaaggg ggcccaggga gctggaaaga gggccaagag gagggcaagg 120 aaggtggcgg gcgacgggga gaggaaagaa aaagggtgtc ttggcggtgg ccttggtaag 180 agaaaggggc aaggggtata attgacaagg cactgaaagt attgaagtca gagccttggg 240 aaggatctac cgaactctcg gcggtccacg cggggacaga cctcagcccg tgagccttga 300 gctccacgcg gggacagacc tcagcccgtg agccttgagc tccacgcggg gacagacctc 360 agcccgtgag ccttgagctc cacgcgggga cagacctcag cccgtgagcc ttgagctcca 420 cgcggggaca gacctcagcc cgtgagcctt gagctccacg cggggacaga cctcagcccg 480 tgagccttga gcccagaagg agtggcagcc tcaggacgtt tgccaggtgg cctggaatgt 540 gagggaagcc tcagccccgc caggaacaga gctggcgctg agttcccggc tcggcccggg 600 11 359 DNA Homo sapiens 11 cccggggcgc acgggcgtga gggtggggtc tcatcgcagg ggcgccggga gcctccccgc 60 tccgctagct caaccaagga ccgctcagag gggctctcac cctgaacctc ggcttttcta 120 aaggagcgag accagattcc cttctcttct cgacgtcgtt tggtgttttc tcgttctttt 180 cgactgagca cggcaatgcg cagacgtcga cgtctctcac tgctcatccc gatctgtaac 240 ctgaggtgag cccgaagacc gctgccgccg gcggccaccc cagcgcgggt ccgctgagga 300 tggaaacagc aagtgcgcgc cggccaggcc gccacctctc cctcctccaa cagcccggg 359 12 370 DNA Homo sapiens 12 cccgggaggt agcgggagat tgcacacgcg atcctgcgag tttccgaact ttggaagatc 60 gtgacccgga gagacccttg ggaggagagg gccggccacc tcctaggggt gctgtttttt 120 taagggtcaa cccaggacgc tacgggaagc acctggcgca tccttggaac agtgggcttg 180 gtggcccacc gcaacgcctg gcgggaagga atggcggggc atcgtgtgcc taatggaccc 240 cgtcacagac ccgccccaat ccgaggggcg gatgagctca gaggacctgc ccaggacgct 300 ccttctccac tttccaggaa aaccgacggc gtgcgcgcct ccgtgtcctc gcggagctgg 360 ggtccccggg 370 13 324 DNA Homo sapiens 13 cccgggcctg ggctgtgcca gcccggcctg ccagtctcgg cccccattct cgtacggagg 60 gatgcggcgc ctggggcctc gaagctccgg gcggttttgg agaagttgaa gctcagccgc 120 gatgatatct ccacggcggc ggggatggtg aaaggggttg tggaccacct gctgctcaga 180 ctgaagtgcg actccgcgtt cagaggcgtc gggctgctga acaccgggag ctactacgag 240 cacgtgaagg tgagctgctt ggcgccctcc cgccgagccc cgctgctcgg ccttccgcaa 300 tccgcagtcc ctaccttccc cggg 324 14 392 DNA Homo sapiens 14 cccgggaaag acctcgagag accttcttca acacgtctcc aggccagatt cccctaccat 60 ggctcgggag caatgacgcg ctccccctcg ccactcgcat ggagacccga cttccctggc 120 gccccacgag aggctcactg acctcgccgt cgtacctcgt gagagaccgc acctgggccg 180 cgtcgagaca cccgagattc cccgtcaccg agagcttgag gccttcgtct cctgcatggc 240 ctagagacca atctcgcgac ctgtctccaa acgccctcag gaggcttgac tcccttgagt 300 ccgcccagtg agctccaaga gatacccgtc gcgattcgag agcagagcgg ggttctttgc 360 ttccactcga gatgaatgcc tgtctccccg gg 392 15 358 DNA Homo sapiens 15 cccgggagag gcgacagcct catccgttta tttcctccct tgaccatttg ttcagcgact 60 ctcccctccg ttcagcatcc aggttcctta cggctacagt gccccagccc cgcctcacca 120 gcgcgacatt ctgccctgcc tacccactca gacacagtgc ccttttcggt tcttcaaact 180 tgctaagcgt tttcctatcg atatctgcag gtaacagatg gcacgctctc aaatagggta 240 atcggaggag ggtctaataa aggaactatt ttcaacagcg gagtaggcgt tagggactcc 300 agtaggagta gggctgtgtc ccaggatact aacagcaggg cgccttggcc gccccggg 358 16 545 DNA Homo sapiens 16 cccgggctgt gcggagatgc gcaggcttgg ggcggcgttc aggagggact gggaaaggga 60 tcgtgcccta gggtctcctg gtgcgaaagg gtgcggcgca gcaggtggga tcagggtgag 120 gtccgctggc atttatgggg tgggtggtgg aaaattggaa agagtttccg gggagttgtg 180 gaagactccg gaaagaaggg tctgttgaag gcggtgtgtt gaaaggatgt aggggaatga 240 cagaggggtg ttttaggggt ccaattgggt gaggtctggg ggggaagata tcgagagggt 300 catgggccca ggagtgcacg tctaggagtt gatggggtag gcctgagggt tcggagaagg 360 tgcggtcggg gaggagtctc gcgattgagt ttgtcggggc gggcgggaag ctgacagctg 420 cctctgtggt ctcaggaggc ggactccgcc ggtgcagccg cccccgacgc gggaggaccg 480 ggcctcatcg tcccgggagc gctccgcgtc gcgaggccgc ggcgccgcgc gctcctcatc 540 ccggg 545 17 510 DNA Homo sapiens 17 cccggggggc gctgaggttg ccttctcagg cggaggaggc gaaccctgta gcccgacaac 60 gccgggcttc gattttgagg agcttcttcc gggatgtcgc tatactggcc ggagacgggc 120 cttgcgtgtc cattggggga tggcgatggg gaccagtttc cgggtagaga aggagataac 180 tcgatacagg aacgcacagg cagcctgaaa gcagcccagg atctcgacag gggaagggaa 240 gaccctgctc cctttgccca aatcctcccg ccctgtcctt gccttctgtc ccagatccca 300 agcccctcgt acctgcactc gggactcggt cagcccgata cgcagtgcca gctcctcacg 360 cataaagatg tcggggtagt gagtcttggc gaagctcctc tccaactcgt tgagctgtgc 420 gggggtgaag cgcgtccggt ggcgcttctg cttctgttgg ccctgctgct ggccggcttg 480 gctggggttc gggccgccct ggggcccggg 510 18 386 DNA Homo sapiens 18 cccgggaaac agttcaggac gctcaagacc agaagcggga gcaaacccaa aaggagctcc 60 aaggaggtgt gtgtggggag agccaggggg acgcaggact aggctctttc ctgcgcaagg 120 ggtggggaaa cccgcgaaag ccagggagtc gcgcgcactc acgccctcgc gccaccaggc 180 agagccaccg ctgcaaggag cccacgggtg cgcgctcgct ccagggcgga tctttccaca 240 cccccctcac cctcaaaagc tcaggctgga gcggtcatca gtgcggactc cggcacccca 300 cccacccagc aggggttaag gagggactgg cgcccactct tgcctacagc tcctgcgcag 360 ggctccagcc gccaaatctt cccggg 386 19 514 DNA Homo sapiens misc_feature (415)..(415) n is any nucleotide 19 cccgggctga atggtagacg ttctggcgcc gggcagcggc caccggctgg ttcccacttc 60 cgcgcgcacc ccttaaactg tgttctagag gccccagcct cgccttgcag cgcctcacta 120 gctcctgagg actagggact cggcggtgag gcgggttggc ggctcgaacg agctgggcgt 180 cttcgttctc tctcgctgcc tggctggctc agctggccct ccacagctcg gagcaaggcc 240 atagcaggga gtggaggtat attgggctgt cacctccttg ctggccggag ttatttgtag 300 actacagact cggaagaaca gacgcgccac gctctcgctt ggcattgctt cggatcgcag 360 ctcctccttg gggtgcccca gcttggcgtt tatttgcctg cgccaggctc tggcnacggt 420 caccgggcca gcggggaggg acggacggca ggtgaccagc ctctgctgtg aagaaattcc 480 tgcgcgcccg gagctgtccc taatgcatcc cggg 514 20 279 DNA Homo sapiens misc_feature (27)..(27) n is any nucleotide 20 ccggggatcg ggagcttgga gtgaggngct cggcgtgacc cgtgaggagc cccgcgggta 60 gagcggcgct gccccttcgc tccggggtga actgaaactt tgctagggga gagggtcggc 120 gccagcctcg cggggttcgg agaagaccca gcgctgtgcg aggtcggggc cgggcagggc 180 agagcagggg tgaaaggaga gacctgtaat gacggcggga tttggggtgc ggagggttgc 240 gagggagggg ccgcaaccct gaacaattgc attcccggg 279 21 422 DNA Homo sapiens 21 cccgggtctg gcttggtctc gcccgcgcag atcccgttca aactcagctg ccaccaagtg 60 cgccttttct ctctggattg cgattctgca cgaattttcc agttgagggt ggttcggcgc 120 tcagccagcc tctgccctcg aagacgcggc cttggtctag gaaccttcag gtgggtgttt 180 gggcgcagtg gccctagttc cctagaattc gttttgcctc ccgcggcctc agccgcgtgg 240 tcagcgagct cgcgagaacg cagctgggca gtcccggacg gctctcgggc gcttctaggg 300 agcagtcaaa gggttgcgag gactgccagg gtccttcctc cctaggcttc ccacactgga 360 tggccgtaac tcagtcgttg acggcgacag ccagggccct gatggactgt gcaggtcccg 420 gg 422 22 464 DNA Homo sapiens 22 cccgggtacc tgcacagctc gctccctccc atccttcggg tcttcgctcg aacgtccgct 60 cctcggtgag gccttccctg gacaacgcat ttgaaacgta accccaaggc aagaagccac 120 cttccaggcg cgcagccgaa gcccagtgcc aaggaggccg gagactcggg tgcccgcgca 180 tcccgaaaac agcctctgag gggtcctctg agcatccttc cagcgtgttt gggaggcaaa 240 ctcgttgact agctcttgag aggagtggct agaggaatcc aggcggggaa ggggacggtg 300 gactccagga gagtgtaatt tacaaaggcg gggggcgggg acgcccaggt ccgagtccca 360 ggactctgcg ccggacgctt cgcccgccct ttcaggtccc ctgcccggtc ctcgtacccg 420 cgcgggtccg gagaacctct gagcaccggc ccccagcccc cggg 464 23 257 DNA Homo sapiens 23 cccgggatcg gtgcccgtta gtgggcacag gacgccgggt gtccgaaggg ctgcctggag 60 atgcaactga ccttggcggg catcagaatg acgagtggca gcagcaggag cggggtgacg 120 aacaagatca cgaaggactt gaacttggag acatagctca gcgccgaggc catcgcgcgg 180 gagggagact ggcgggcgag acgagtgagg ggcagctaga ggcgccgcgg gcttaagaag 240 gggccacagt ccccggg 257 24 350 DNA Homo sapiens 24 cccggggatg gcgcgccagg taaggcaggc ggacgcgcgg acccctcggc gcagtgcggc 60 cccgaaggcg gtagggcgag aaagagctgg gacccccccg caacttcggg atcggctggg 120 tgaggctggg caggcctgat gctcaggggt tcgtgccggg aagttcaggt cctcggacaa 180 cctctctcca gctctccgcc gccggtaccc acggcccagt ctccacctgg ggaaaccccc 240 ttggcgtggc ttgtttcgtt acaagttatc ctggtagagt gggcatgaag gcctcggagg 300 cagtgagtaa atctcatact tctgttcttg gtggaatcct ggatcccggg 350 25 337 DNA Homo sapiens 25 ccccgggctg ccggctggag cccggcagat tcgctgcgca gcactgcccc ctggtatcca 60 gcgccgaaag tgcccccctc cgcagctgca aggctcctcc ctgggctgcg cgggacagat 120 tttttctcct ttcctggcta cacgcctaac agagaagcta tcccgaggga cctcaagaag 180 tccccccaag ccgtactcaa aagcctttct ccctcctcct caagcgctca cttccccaaa 240 gaggacccgg acccctgact gcctgagcca ggtccccagc atggtccgca acccttctcg 300 actccggcat ccacctccag gctgacgtct acccggg 337 26 229 DNA Homo sapiens 26 cccgggggaa agagatgagt ggaaatcgtg ggccggacct gcaaggagag actcggcggg 60 caccttgctt ggtctgaggt cgtctgcagg aagcggactt ttctcctggc tcaggatggg 120 aaagacaggg gatgcctgaa gtcaacgggg acttctgttc catctctgcc ccgttctcca 180 ggcccgccag tttttcctgc tttggttaga ttttccaacg tatcccggg 229 27 427 DNA Homo sapiens 27 cccggggctg ctgaccgagc cccagggcag cccgcccttc cccctcctga agcccgacgt 60 cttcttcatg gctccgtcta ccggcttcgg agacccgctg gacttttcgc cgcctccgga 120 accctatatg aggaagcaaa tcgcgtccgc cacagcctcc aactaggaaa ctccgcgact 180 ctcagcccct cagaagagaa acggagacgc gccaagcaaa gccgttacac ggactgtgca 240 cgcgcctccg gtgtccctgc gcgtgacaca aatttggccc cgagggagct ccatgtgcct 300 gagtcccagg agccctagat gccagcgaca gcttgtcacc aggcctgcga cgccaatggg 360 cgggagtcgg cggagctcag gacactgacg acgggcctgg gggaaagcgg tccccacaca 420 gcccggg 427 28 470 DNA Homo sapiens 28 cccggggaca gggggacccc cagatgctgc acggctgaca ggccaacgtg gcagaagctc 60 cagcttcaca ggaagccagt gaccatgaga gtctgtagct gtaacgaagc cacagagctg 120 tggctttctt tccccttcag ctctaggaaa ggttatctgc cctgcacaga tctccggagg 180 cctggctggg ctctgagagc atcagactga ttatcgtaag aaaataatct ctgcagacac 240 attccttgct agaagcaggg gacaaagccc agcttcaaag acaattccac acacgccctc 300 cctgccctgc acagctgcct gccgggtggg agcagagccc ttgcagccgg gctcaggggc 360 ctgggcaggg acagcgtgtg gcaggggcac agctgagaca ggagcctcaa agcgacacca 420 acccgacgtg aagctacagt tgaggagaca cagctgcccc cattcccggg 470 29 264 DNA Homo sapiens 29 cccgggcggc ggccggattg cgggtgggtg agaggcagca gacgccgtgt ttacagctct 60 ctcgctagtt cgccacctca gccgcggctc tagggctgag ccagtcgcct ccttctttaa 120 gattctggtc acagcagggg ctgggtttct aaggcaggtt tctaaggtgt cttcctacag 180 acaccgctgc tgctaccttg ctaccttcag cgctgggcac agccaggggc agcgcgagag 240 ggaggcaacg agagggttcc cggg 264 30 371 DNA Homo sapiens 30 cccgggaccc caatgccagg gaggggcctg caggacccca gcggtgggcg agttgtgtcc 60 tgggtcacct tgtgtttcgc agcgtggcgg tggcaggagc ccagcgcggg aggacatttt 120 catagcctcc tacagtgaga aacgcccccc acccgacgct gcgctcatct gtgtccccgc 180 tgttgccggg gctctggtat ccacttgcgc gccctatgtg gtggggatcc acccagagcc 240 cagcgtcaag ttatacgggc gcttcactca gcgtcagcca agaccaggga agcgcttctt 300 gccgtttagg agacgtctgc aagagataaa aagctagccc acgatccacc cacaatcctc 360 gtgtccccgg g 371 31 179 DNA Homo sapiens 31 cccgggaact cgcggcaccc actgggtatt gtcgggaccc agcaagtcta ggaacggggg 60 tgggtagagc atccttcggg cactgccgtt cgtccccaaa agaagaccac cgcggggtcc 120 cagggccacg gcgaggacgg gcactggtca gattccggac aggcggtcct ggccccggg 179 32 445 DNA Homo sapiens 32 cccgggctgt gaccagcgaa ttcgggcccc gcaggtgcag ctgataggag aaatccggct 60 ccgggagcga acccagcggc ggaaaggcgg gctccgcgcc caggcgggcc ttggactgca 120 agaaggcgag gatgcgcgcg tacttcgtgt ccttggtctc atcgtcacgt gtgagtatcg 180 accaggtcat catcgcacgt ggtaccatag tggaagtagt tggcaaactc gctagagtct 240 gctggaggaa cgagcccgcc gtaggacgga cacacctgag tgcccctccc acgcgagccc 300 aaagcgggtg cagggcacct cccaccacat ttctggccaa agttcccatt tgaggcccgc 360 cctctcctct gcgcagtctt agagactggc gaggcacgcg caaacgccct cttccctgag 420 acctgacccc acccacccac ccggg 445 33 357 DNA Homo sapiens 33 cccgggggag ccccggaccc cgcatccccc agggcgcgga aactggcgag gccccaggag 60 ctcccattta tagctcagtt tccactgagc gcagtccctc taggacctgg gctgagcaag 120 tttcttccac tctctccctt ccctcctcct caccccttgc ctgcccctca accccggcag 180 ggcgcaggtg tccaacccag ccgggacccc ctccctcctc gaacccaggt gttccggctc 240 ccagacccca attgagctgg gggcgcccac ccgccggggg atcccgccct gcgtccccca 300 ttcatccgcg tctcagccgc gggagtttct caacgggaag agggcggagc tcccggg 357 34 312 DNA Homo sapiens 34 cccgggagga gagttggggc ttgggggacg ccgtgaactc catggtcccc agcacgcggt 60 cctggccagg gacggggtcg tccgaactgc cgtccagatt ccccaaggga gacaaagacc 120 cgaaacacag ctcaaagttt ccgagagcag tcacagcggg gccagggact ccagaagtgt 180 cagctccaac gactccagag ctgcacactg gcctctattc cccaccgcaa agccccagag 240 ccgcagagac ttcgaaggca gccggagagg agagggccca ccgagcacta cggcgggtgc 300 gcacgccccg gg

312 35 372 DNA Homo sapiens 35 cccgggagcc ggctcgctgg cggcgccagg ccacgctctc tcattaacat cccgctcccg 60 gtggcgcagg ggagccggcc aaagttcctc gcaaagtggc gagcgaagga gcgctgagca 120 ctgacgtctg ggctggggag gagcgggtcc gagcgaggac ggagagggga cagagggaaa 180 gggaggcggg tgtcttcctc aggaatttga gctggggatc tgcatcctgg ccattgcagt 240 cctttagcat cctcgccgcg ccctgagcgc gctggaggct cgcaggctgc gccctcccag 300 ggctgatgcc gcgtcctgct ccgccgttct gggacgtcgg ggacaaaagt ggaggagacg 360 ggagagcccg gg 372 36 399 DNA Homo sapiens 36 cccgggctgc gagcgcggct cctggattcc agcctcccgc ccttcccagg cgctggaatg 60 gacacggacg cccacagtgg cgggccaggt agtgccggag tcgggggccc aggccgcggc 120 gccccgcgcc tcatcactta ccttgccttt agctatcaat tccatgatgt agccaaattc 180 actcatctcc ccagactccg acatgtttac accccttcac aaactctgga ggaccgacgc 240 gggtgtatcg aatttgtcct ttcttttctc tttttctgtt tttagtctga gttttgccga 300 gctccccgcc cataagctgt taaccaggaa aagaggggaa gcgccgggga aagcaagaag 360 cgggcttggg tgaaatgaag gccatcgagg gctcccggg 399 37 307 DNA Homo sapiens 37 cccgggcccg gtgcgcaccg gtgccgactt ggcagccgcc ctgtgcgctc gacgaaaggg 60 tgagaaggag gcaggagtgc aggcggaagg agtgggcaat cagcggcggg gacgagagtg 120 tgtcttcggg gaaaccaagt ctgagtgagc gctgaagggg agtgtgcgga gcgtgccgtg 180 caccccgagc cccccgcctc attgcctctc gcctctctcc acctgcccca tgatctgcgc 240 cagggaccgg tcctctcccg tccgcaggct gtctaggtgg ccgttctggt ttgctgggac 300 ccccggg 307 38 331 DNA Homo sapiens 38 cccggggcca cctctgaggc atgaacccag agacgcgcgc cctggtctgg gaaagcagga 60 ccgctgcgcc cagcgcctca ggggtagagg cgggaacagg cccgcggtcg ctttgctggc 120 ggcggggaag ggcgatctga cgatcaggga gttgcgcccc tctctctggg cctcgtgaag 180 gaacaagagc aattacagcg ctgggccggc cacgtagtcc tggggctagg tgggcccaat 240 gctccgggcc gcggggctgg agcgcggagg ctggagaggg aggaggaccc tccgcggctc 300 caggtctccc agctggaggc tcacgcccgg g 331 39 304 DNA Homo sapiens 39 ccgggcccgg tgcgcaccgg tgccgacttg gcagccgccc tgtgcgctcg acgaaagggt 60 gagaaggagg caggagtgca ggcggaagga gtgggcaatc agcggcggga cgagagtgtg 120 tcttcgggga aaccaagtct gagtgagcgc tgaaggggag tgtgcggagc cgtgccgtgc 180 accccgagcc ccccgcctca ttgcctctcg cctctctcca cctgccccat gatctgcgcc 240 agggagccgg tcctctcccg tccgcagctg tctaggtggc cgttctggtt tgctgggccc 300 cggg 304 40 307 DNA Homo sapiens 40 ccgggggtcc cagcaaacca gaacggccac ctagacagcc tgcggacggg agaggaccgg 60 ttccctggcg cagatcatgg ggcaggtgga gagaggcgag aggcaatgag gcgggggggc 120 tcggggtgca cggcacgctc cgcacactcc cctccagcgc tcactcagac ttggtttccc 180 cgaagacaca ctctcgtccc cgccgcgtga ttgcccactc cttccgcctg cactccagcc 240 tccttctcac cctttcgtcg agcgcacagg cggctgccaa gtcggcaccg gtgcgcaccg 300 gcccggg 307 41 304 DNA Homo sapiens 41 ccgggcccgg tgcgcaccgg tgccgacttg gcagccgccc tgtgcgctcg acgaaagggt 60 gagaaggagg caggagtgca ggcggaagga gtgggcaatc agcggcggga cgagagtgtg 120 tcttcgggga aaccaagtct gagtgagcgc tgaaggggag tgtgcggagc cgtgccgtgc 180 accccgagcc ccccgcctca ttgcctctcg cctctctcca cctgccccat gatctgcgcc 240 agggagccgg tcctctcccg tccgcagctg tctaggtggc cgttctggtt tgctgggccc 300 cggg 304 42 479 DNA Homo sapiens misc_feature (1)..(479) n is any nucleotide 42 cccgggttcc tggcttgaac cctgtttctc cctgttctgc caggcatgct ggtccggaag 60 gtgtgtgtng ctgtnggctt taggtgggtg cagcccgtcc cacgtcacgg cgagctctgt 120 ttcctgggct ggggacagtg aggtcatcgc tgcccatcct ggagctctgg ctcctttcgg 180 gtacctgttc cctctcccag agagaccccc agctgcatgc aggcctagtg ggctccacgg 240 cggagctggt tcccaggcta cctgggttgc cacctctgtg ggtcccggct gccctctcgc 300 agccgccgct acttcctcac cctcttggcc ctgcatttcc acgtctcatg gagccaacga 360 gagcaggggg tttgagccct tgtggaaatc tggggaggca ctgcttctcc ctccatgtga 420 gcagcttcac ccagcctggg gtcagtgctt acgctccacg cggcctggcc ttccccggg 479 43 21 DNA Artificial sequence PCR primer 43 gagttggtga tgttagatta g 21 44 22 DNA Artificial sequence PCR primer 44 ttcccaaaaa aatcccaaat tc 22 45 22 DNA Artificial sequence PCR primer 45 ctccttccaa ataaatactt ac 22 46 19 DNA Artificial sequence PCR primer 46 agagaggagt ttagattgg 19 47 21 DNA Artificial sequence PCR primer 47 caaaaaaact aaaacctcaa c 21 48 24 DNA Artificial sequence PCR primer 48 tggataaagg atgtttgggg tttg 24 49 22 DNA Artificial sequence PCR primer 49 ccctcccctt acccctaaat cc 22 50 19 DNA Artificial sequence PCR primer 50 actccccctc actttattc 19 51 24 DNA Artificial sequence PCR primer 51 attattttag tagaggtata taag 24 52 18 DNA Artificial sequence PCR primer 52 ccaaccccac ccttcaac 18 53 20 DNA Artificial sequence PCR primer 53 aagagagggt tggagagtag 20 54 24 DNA Artificial sequence PCR primer 54 cccctaaaaa aaaaatcaaa aatc 24 55 21 DNA Artificial sequence PCR primer 55 gggttggaga gtaggggagt t 21 56 26 DNA Artificial sequence PCR primer 56 ygttatgatt tttttgttta gttaat 26 57 23 DNA Artificial sequence PCR primer 57 tacaccaact acccaactac ctc 23 58 25 DNA Artificial sequence PCR primer 58 tttatttata taattttgtg tatgg 25 59 21 DNA Artificial sequence PCR primer 59 cacccctcac tttactaaaa c 21 60 23 DNA Artificial sequence PCR primer 60 tttgggaggt aaattygttg att 23 61 24 DNA Artificial sequence PCR primer 61 accraacaaa aaacctaaaa aaac 24 62 24 DNA Artificial sequence PCR primer 62 ttattagagg gtggggtgga ttgt 24 63 24 DNA Artificial sequence PCR primer 63 ttattagagg gtggggcgga tcgc 24 64 21 DNA Artificial sequence PCR primer 64 aggattggga tgttgagaat g 21 65 19 DNA Artificial sequence PCR primer 65 ggattgggat gtcgagaac 19 66 26 DNA Artificial sequence PCR primer 66 gttttttttt ggatttttgt tttttg 26 67 19 DNA Artificial sequence PCR primer 67 gttttttcgg ggcggtttc 19 68 31 DNA Artificial sequence PCR primer 68 ttttgttttg ttattttttg tttttggttt t 31 69 29 DNA Artificial sequence PCR primer 69 cgtttcgtta ttttttgttt tcggttttc 29 70 23 DNA Artificial Sequence PCR primer 70 gtttggttgt tgtttattgg ttg 23 71 22 DNA Artificial sequence PCR primer 71 cctaaactca caaaccaact ca 22 72 21 DNA Artificial sequence PCR primer 72 tgcgagcgtt tttttaaatg c 21 73 20 DNA Artificial sequence PCR primer 73 taaactcgca aaccaactcg 20 74 22 DNA Artificial sequence PCR primer 74 ttaataggaa gagtggatag tg 22 75 24 DNA Artificial sequence PCR primer 75 tctataaatt actaaatctc ttca 24 76 21 DNA Artificial sequence PCR primer 76 ttaataggaa gagcggatag c 21 77 23 DNA Artificial sequence PCR primer 77 ctataaatta ctaaatctct tcg 23 78 25 DNA Artificial sequence PCR primer 78 taattttagg ttagagggtt attgt 25 79 20 DNA Artificial sequence PCR primer 79 cacaaccaat caacaacaca 20 80 22 DNA Artificial sequence PCR primer 80 ttaggttaga gggttatcgc gt 22 81 23 DNA Artificial sequence PCR primer 81 taactaaaaa ttcacctacc gac 23 82 27 DNA Artificial sequence PCR primer 82 ggaggatagt tggattgagt taatgtt 27 83 21 DNA Artificial sequence PCR primer 83 caaatccctc ccaaacacca a 21 84 24 DNA Artificial sequence PCR primer 84 ggatagtcgg atcgagttaa cgtc 24 85 16 DNA Artificial sequence PCR primer 85 ccctcccaaa cgccga 16 86 29 DNA Artificial sequence PCR primer 86 tttgtgtttt gatgtttgta ggtttttgt 29 87 28 DNA Artificial sequence PCR primer 87 aactccacac tcttccaaaa acaaaaca 28 88 23 DNA Artificial sequence PCR primer 88 tttcgacgtt cgtacctttt cgc 23 89 22 DNA Artificial sequence PCR primer 89 gcactcttcc gaaaacgaaa cg 22 90 22 DNA Artificial sequence PCR primer 90 ggggttgagg ttttttgtta gt 22 91 23 DNA Artificial sequence PCR primer 91 ttcacaacct caaatctact tca 23 92 21 DNA Artificial sequence PCR primer 92 gggttgaggt tttttgttag c 21 93 20 DNA Artificial sequence PCR primer 93 ctacttcgcc taacctaacg 20 94 25 DNA Artificial sequence PCR primer 94 ggtgttgtta aatgtaaata atttg 25 95 22 DNA Artificial sequence PCR primer 95 aaaaaaaaac acctaaaact ca 22 96 21 DNA Artificial sequence PCR primer 96 aatcgaattt gtcgtcgttt c 21 97 25 DNA Artificial sequence PCR primer 97 aaataaataa ataaaaaaaa acgcg 25 98 26 DNA Artificial sequence PCR primer 98 gaattgagat gattttaatt ttttgt 26 99 22 DNA Artificial sequence PCR primer 99 ctaaaaccat cacccctaaa ca 22 100 23 DNA Artificial sequence PCR primer 100 ttgagacgat tttaattttt tgc 23 101 20 DNA Artificial sequence PCR primer 101 aaaaccatca cccctaaacg 20 102 22 DNA Artificial sequence PCR primer 102 gagtggttag aggaatttag gt 22 103 24 DNA Artificial sequence PCR primer 103 ctaaaaaaac aaacaaaaca tcca 24 104 20 DNA Artificial sequence PCR primer 104 gtggttagag gaatttaggc 20 105 19 DNA Artificial sequence PCR primer 105 aaaacgaacg aaacgtccg 19 106 21 DNA Artificial sequence PCR primer 106 acttccatta aaaacaacta c 21 107 23 DNA Artificial sequence PCR primer 107 caaccccaaa cccacaacca taa 23 108 22 DNA Artificial sequence PCR primer 108 gacccccgaa ccgcgaccct aa 22 109 23 DNA Artificial sequence PCR primer 109 ttacaaaaaa ccttccaaat aca 23 110 22 DNA Artificial sequence PCR primer 110 tacaaaaaac cttccgaata cg 22 111 23 DNA Artificial sequence PCR primer 111 tttattccaa cttcttcact tca 23 112 19 DNA Artificial sequence PCR primer 112 ttccgacttc ttcgcttcg 19 113 21 DNA Artificial sequence PCR primer 113 ccccccaaaa accccacctc a 21 114 17 DNA Artificial sequence PCR primer 114 ccgaaaaccc cgcctcg 17 115 21 DNA Artificial sequence Amplification primer 115 ttgtgtgggg agttattgag t 21 116 23 DNA Artificial sequence Amplification primer 116 caccttcaca aaaaaaatca atc 23 117 18 DNA Artificial sequence Amplification primer 117 tgtggggagt tatcgagc 18 118 19 DNA Artificial sequence Amplification primer 118 gccttcgcga aaaaaatcg 19

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed