• Non ci sono risultati.

361 The molecular pathology of inflammatory bowel I disease-associated neoplasia and preneoplasia

N/A
N/A
Protected

Academic year: 2022

Condividi "361 The molecular pathology of inflammatory bowel I disease-associated neoplasia and preneoplasia "

Copied!
8
0
0

Testo completo

(1)

361 The molecular pathology of inflammatory bowel I disease-associated neoplasia and preneoplasia

STEPHEN MELTZER

Introduction

Molecular alterations underlying inflammatory bowel disease-associated neoplasia and preneoplasia (IBDN) have been studied extensively over the past 10 years. Perhaps the most interesting facet of these studies has been the emergence of IBDN as a distinct form of colorectal neoplasia, with both similarities to and differences from sporadic colorectal neoplasia and preneoplasia (SCN). This finding of differences between IBDN and SCN should not surprise IBD clinicians, who are quite familiar with the known biologic differences between these two forms of neoplasia. For example, it is widely held that the interval between dysplasia and frank carcinoma is only a few years in IBDN, contrasted with 10-15 years or longer for SCN. Furthermore, frank carci- noma in IBDN often evolves from flat dysplastic lesions, whereas SCN is commonly believed to evolve from polypoid dysplasias (adenomas). Finally, IBDN develops in a setting of intense mucosal inflammation, often after 20 or more years of non- dysplastic IBD; SCN typically develops on a substratum of non-inflamed colonic mucosa.

Investigations of IBDN, however, have closely paralleled the investigative paradigm estabhshed in SCN by Bert Vogelstein and his coUeagues [1]. Early in the history of these analyses, proto-oncogenes of the ras family were studied [2-5]. Later, loss of heterozygosity (LOH) was evaluated, and evalua- tions of tumor-suppressor genes (TSG) evolved directly from these LOH studies [1, 6-10]. Subse- quently, the unique form of mutation known as microsatellite instability (MSI) was evaluated in IBDN, and the ramifications of this form of mutation were investigated [11-16]. This form of mutation comprises length mutations in oligo- nucleotide repeat sequences, typically in non-coding regions of genomic DNA. The ability of MSI to target specific coding regions was also evaluated in

IBDN [17-19]. MSI is caused by inactivation or defective function of DNA mismatch repair (MMR) genes [20-24]. Finally, gene inactivation by promoter hypermethylation was studied in IBDN [25]. Hyper- methylation appears to be of pivotal importance in IBDN, since it inactivates TSG such as the cyclin- dependent kinase inhibitor p i 6 [26] along with MMR genes [25]. In the future comprehensive new molecularly based t a x o n o m i e s may augment traditional approaches to classifying IBDN. In parti- cular, gene expression 'signatures', or profiles of the expression levels of thousands of genes generated by cDNA microarrays, promise to shed light not only on new taxonomies, but also on novel molecular genetic pathways involved in IBDN [27-35]. Genes involved in these pathways offer the potential to serve as biomarkers of cancer risk and disease progression;

moreover, they present promising targets of future molecular and pharmacologic cancer prevention and treatment strategies.

Abnormal DNA content (aneuploidy)

Much of the groundwork for future investigations of neoplastic progression in ulcerative colitis was laid with studies of DNA content, or ploidy, which can be determined using flow cytometric measurement of disaggregated nuclei [9, 36-43]. This is a very valu- able tool in detecting clonality of tissues or cells. The earliest reports of flow cytometric DNA analysis in ulcerative colitis were published from Stockholm in 1984 [44] and in a later survey by the same group [45].

The prevalence of aneuploidy in this series of 51 patients correlated well with disease duration and the presence of histologic dysplasia or carcinoma.

Similar findings were reported by other investigators [38,46, 47]. A higher prevalence of DNA aneuploidy was found in dysplastic than in nondysplastic tissues,

Stephan R. Targan, Fergus Shanahan andLoren C. Karp (eds.), Inflammatory Bowel Disease: From Bench to Bedside, 2nd Edition, 711-718

© 2003 Kluwer Academic Publishers. Printed in Great Britain

(2)

but this difference did not achieve statistical signifi- cance [45]. A prospective study on aneuploidy in ulcerative colitis was published in 1987 [48]. In this study 53 patients with long-standing universal ulcerative colitis were followed for a mean of 3.5 years. Four of five patients with aneuploidy developed dysplasia; aneuploidy preceded dysplasia by 1 year in one patient and antedated a Dukes A carcinoma by 1 year in another patient.

The precise predictive value of aneuploidy has yet not been consistently substantiated [49, 50]. Other publications on aneuploidy in ulcerative coHtis have included elegant mapping studies, some of which combine ploidy studies with molecular analyses [38, 40, 51, 52]. These systematic investigations demon- strated large areas of colonic mucosa containing identical aneuploid DNA content. This evidence strongly suggests that these large areas of epithelium arose from a single abnormal progenitor cell, in agreement with the clonal theory of cancer proffered above.

In summary, measurement of DNA content is a well-established technique that has been shown to correlate often, if imperfectly, with the clinical progression of premalignant lesions in ulcerative colitis. In this respect it shows potential as a clinical prognostic tool to be used in conjunction with conventional histologic analysis.

Proto-oncogenes in IBD neoplasia

Proto-oncogenes are the normal forms of genes, expressed in normal cells under physiologic circum- stances, which become activated to their oncogenic forms (i.e. into oncogenes) by various molecular mechanisms in tumors [53]. Even though there are two alleles of each proto-oncogene in each cell, only one allele need be activated in order for a carcino- genic eff^ect to be exerted. In this respect proto- oncogene mutations function as dominant muta- tions, and oncogenes are often referred to as 'domi- nant oncogenes'. One group of proto-oncogenes is known as the ras family, comprising three genes:

Kirsten ras (Ki-ra^ or K-ra^), Harvey ras (Ha-ra^ or H-ra^-), and neuroblastoma ras (N-ra^]. These genes are activated in tumors by point mutations of codons 12, 13, and 61. Mutations in ras family genes have been reported in SCN, particularly in ¥^-ras and less frequently in H-ras [54-59]. Mutations in SCN are most frequent in codons 12 and 13 of Y^-ras and H- ras [54-60]. Studies of IBDN lesions identified the

first major contrast between these lesions and SCN:

namely, a paucity of K-ra^ and H-ra.s' mutations [2-5, 43, 61-63]. Reasons for this apparent contrast are unclear, but this finding was the first of many to suggest that molecular pathways underlying the two forms of colonic neoplasia might differ.

Gene amplification in IBD neoplasia

Gene amplification is the process of duplication and reduplication of genomic DNA regions, sometimes encompassing large regions of chromosomes and multiple genes [64]. Certain cellular proto-oncogenes (such as c-myc, N-myc, c-myb, EGF-R, and c-erbB- 2) are activated by DNA amplification in human tumors [59, 61, 65-72]. These extra genomic copies of genes often result in increased levels of mRNA or protein expression of the genes encoded. However, relatively few studies of gene amplification in IBDN have been published. One study found no amplifica- tion of common proto-oncogenes in IBDN [61].

Tumor suppressor genes and loss of heterozygosity in IBD neoplasia

TSG can be considered the opposites of proto- oncogenes. TSG suppress tumor formation by var- ious mechanisms, either by preventing entry into the cell division cycle, promoting programmed cell death (apoptosis), leading to cellular growth arrest, or interrupting proliferative signals to the cellular inter- ior [73]. TSG are present in normal form in all cells of the body, but in contrast to proto-oncogenes, which are activated, they become inactivated in tumors [73]. However, because inactivation of only one copy (allele) still leaves a remaining active copy, both alleles must be inactivated in order for tumorigenesis to proceed. For this reason, TSGs are sometimes referred to as recessive oncogenes or a«r/-oncogenes.

LOH, also known as allelic deletion, is the process of deletion of one allele of genomic DNA, often encompassing large chromosomal regions [7, 59, 74]. Because LOH may remove one functional copy of a TSG, it is a hallmark of TSG inactivation.

However, LOH may also occur non-specifically, in

random fashion, and aff'ect any portion of the

genome [6, 7]. When LOH occurs frequently at a

particular chromosomal locus it is widely believed

to represent a non-random event and to imply the

presence of a potential TSG at that locus [74, 75].

(3)

Generally speaking, LOH frequency must exceed a 'background' frequency in order for non-random- ness to be assumed, but this frequency varies from study to study and among different tumor types [7, 76-87]. For SCN this background LOH rate approx- imates 20% [7]. Thus, LOH rates in SCN significantly higher than 20% imply the existence of a TSG at a given locus.

In IBDN, LOH has been observed frequently at chromosomal arms 5q, 8p, 9p, 13q, 17p, and 18q [8, 15, 16, 88-93]. These loci house the TSGs adenoma- tous polyposis coh (APC) (5q), p i 6 / p i 5 / p i 4 (9p), retinoblastoma (Rb or RBI) (13q), p53 or TP53 (17p), and DPC4 or SMAD4 (18q). 18q also contains the gene deleted in colon cancer (DCC) [94]. LOH in IBDN has been shown to affect large areas of colonic mucosa [9, 88]. This finding suggests that the LOH originated in a single progenitor cell, conferring a growth advantage on it and leading to its clonal expansion.

Point mutational inactivation of tumor suppressor genes in IBD neoplasia

p53

The classic paradigm for TSG inactivation was discovered in sporadic colon neoplasia (SCN) by the Vogelstein group [1]. According to this model, one allele of a TSG is removed by LOH, while the remaining allele is inactivated by point mutation.

The prototypical TSG fitting this paradigm is p53.

p53 is a TSG which encodes a nuclear DNA-binding transcriptional activator with multiple growth-sup- pressive functions, including induction of apoptosis, exit from the cell cycle, and growth arrest [95].

Approximately 50% of all human tumors show inactivation of p53 [96]. In SCN, p53 inactivation constitutes a relatively late event, occurring only in large, severely dysplastic, often villous adenomas [75]. In contrast to SCN, IBDN is characterized by the relatively early occurrence of p53 inactivation [8- 10, 12, 97-99]. p53 mutations have even been described in non-dysplastic IBD mucosa [98]. As with LOH, p53 mutations can populate large regions of colonic mucosa, again suggesting a single event in a parent cell leading to clonal expansion of that cell due to a growth advantage over surrounding non- mutant cells [98]. An analogous finding has been reported for LOH affecting the p53 gene locus on

chromosome 17p, which can occur in non-dysplastic mucosa and occupy large portions of colonic mucosa in IBDN [9].

Adenomatous polyposis coli

APC, which stands for adenomatous polyposis coli, is the TSG responsible for the familial colorectal cancer syndrome known as familial adenomatous polyposis, or FAP [100-102]. When mutant in the germline, APC causes this syndrome. However, APC is also widely regarded as the 'gatekeeper' gene for sporadic colon cancer [103]. 'Gatekeeper' signifies a somatic event that occurs extremely early and frequently in a given tumor type: a gatekeeper event is the sine qua non of a tumor, without which the neoplastic process cannot initiate in that particular organ or cell type [104]. APC mutation, usually accompanied by LOH of its locus on chromosome 5q, occurs in 80% of sporadic colon cancers and adenomatous polyps [103, 105-108]. Mutations in APC tend to cluster in a region comprising the proximal two-thirds of exon 15, known as the muta- tion cluster region or MCR [109]. APC mutations in the MCR occur even in the smallest benign colonic adenomas, less than 0.5 cm in diameter [103]. Herein lies one of the most striking contrasts between SCN and IBDN: in IBDN, APC mutations in the MCR occur in less than 5% of colorectal dysplasias and cancers [63, 110]. 5q-LOH occurs in IBDN, but also at a much lower frequency than in SCN [15, 92].

Moreover, when 5q-LOH or APC mutations do occur in IBDN, they are found late in neoplastic progression [15, 90, 92, 110]. These findings contrast with p53 mutation or 17p-LOH, which represent early events in IBDN and late events in SCN [8-10, 12,97-99].

One interesting aspect of APC is a particular

germline sequence variant, I1307K [111]. This

alteration, occurring in codon 1307, leads to the

replacement of isoleucine by lysine [111]. However,

this amino acid substitution itself does not alter

protein function appreciably. Rather, the underlying

nucleic acid substitution (replacement of a thymidine

by an adenine) creates a microsatellite tract (see

below]. This microsatellite tract itself, along with

surrounding regions within the gene, becomes prone

to secondary mutations, which occur somatically

[111]. Patients with this germline sequence variant

have an increased likelihood of developing colon

cancer within their lifetimes, presumably because

APC is the gatekeeper gene in this organ, and

(4)

somatic APC mutations occurring in colon epithe- lium are more likely than in other organ epithelia to lead to cancer development [111]. This germline mutation is more frequent in Ashkenazi Jews than in other populations [112-114]. However, it is extre- mely rare in Ashkenazi Jews with IBD, occurring in only 1.5% of this group, and it is found in only 0.8%

of all IBD patients [115]. Thus, this germHne muta- tion does not hkely account for a significant propor- tion of neoplasia arising in the setting of chronic IBD.

Microsatellite instability in IBD neoplasia

Microsatellite instability (MSI) comprises length mutations in oligonucleotide repeat portions of the genome [116-118]. This phenomenon is a manifesta- tion of defective DNA mismatch repair, or MMR [119 121]. When DNA MMR genes are mutated in the germline they result in the genetic disorder known as hereditary non-polyposis colorectal cancer (HNPCC) [119]. HNPCC is characterized by a greatly increased risk of developing carcinomas of the endometrium, stomach, right side of the colon, and other anatomic sites [122, 123]. However, DNA MMR gene inactivation and MSI also occur in sporadic tumors affecting the colorectum, endome- trium, stomach, and other organs [124-129]. MSI has been classified into three types: (a) MSI-high or MSI-H, i.e. MSI affecting 40% or more of oligonu- cleotide repeat sites in a particular analysis; (b) MSI- low or MSI-L, connoting MSI affecting less than 40% of these loci; and (c) MSI-negative or micro- satellite-stable (MSS), meaning MSI affecting none ofthe loci analyzed.

MSI-H is widely regarded as 'true' MSI, i.e. reflect- ing an underlying defect in MMR [130, 131]. MSI-L may occur even in the absence of an underlying MMR defect and may represent a random, non- specific event, analogous to the occurrence of LOH at or below its background frequency for a given tumor type [132]. The vast majority of tumors with documented DNA MMR gene defects are MSI-H, rather than MSI-L [132].

IBDN have been studied for MSI. In one study MSI was found in 19% of IBD-associated dysplastic and cancerous lesions, and in 21% of patients with IBDNs [11]. In other published studies the preva- lence of MSI in IBDNs was somewhat higher [13,14, 16].

The most prevalent molecular mechanism under- lying MSI in sporadic human tumors is inactivation of DNA MMR genes. Germline mutations in at least six MMR genes have been described in patients with HNPCC, who have tumors characterized by a high frequency of MSI (MSI-H) [20, 119, 120, 133-141].

However, mutations in MMR genes are rare in sporadic tumors with MSI [24, 142, 143]. Recently, an alternative mode of MMR gene inactivation has been described in these tumors: hypermethylation of the hMLHl mismatch repair gene promoter region [144-148]. Hypermethylation of hMLHl occurs in 80% of SCN with MSI [144, 146, 149]. It has also been described in endometrial and gastric tumors with MSI, where it may occur early in tumorigenesis [145, 147, 148, 150]. Approximately 50% of IBDN with high-frequency MSI (MSI-H neoplasms) show hypermethylation of hMLHl [25]. Thus, this epige- netic alteration constitutes an important example of the contrast in molecular pathways between IBDN and SCN.

References

1. Fearon ER, Vogclstcin B. A genetic model for colorectal tumorigenesis. Cell 1990; 61: 759 67.

2. Mcltzer SJ, Mane SM, Wood PK ct a/.Activation of c-Ki-ras in human gastrointestinal dysplasias determined by direct sequencing of polymerase chain reaction products. Cancer Res 1990; 50: 3627 30.

3. Burmer GC, Levine DS, Kulander BG, Haggitt RC, Rubin CE, Rabinovitch PS. c-Ki-ras mutations in chronic ulcera- tive colitis and sporadic colon carcinoma. Gastroenterology

1990; 99: 416 20.

4. Bell SM, Kelly SA, Hoyle JA ct al. c-Ki-ras gene mutations in dysplasia and carcinomas complicating ulcerative colitis.

Br J Cancer 1991; 64: 174-8.

5. Burmer GC, Rabinovitch PS, Loeb LA. Frequency and spectrum of c-Ki-ras mutations in human sporadic colon carcinoma, carcinomas arising in ulcerative colitis, and pancreatic adenocarcinoma. Environ Health Perspect 1991;

93:27-31.

6. Kern SE, Fearon ER, Tersmette KW et al. Clinical and pathological associations with allelic loss in colorectal carcinoma [Corrected; published erratum appears in J Am Med Assoc 1989; 262: 1952]. J Am Med Assoc 1989; 261:

3099-103.

7. Vogelstein B, Fearon ER, Kern SE et al. Allelotype of colorectal carcinomas. Science 1989; 244: 207-11.

8. Greenwald BD, Harpaz N, Yin J et al. Loss of heterozygos- ity affecting the p53, Rb, and mcc/apc tumor suppressor gene loci in dysplastic and cancerous ulcerative colitis.

Cancer Res 1992; 52: 741-5.

9. Burmer GC, Rabinovitch PS, Haggitt RC et al. Neoplastic progression in ulcerative colitis: histology, DNA content, and loss of a p53 allele. Gastroenterology 1992; 103: 1602- 10.

10. Burmer GC, Crispin DA, Kolli VR et al. Frequent loss of a p53 allele in carcinomas and their precursors in ulcerative colitis. Cancer Commun 1991; 3: 167-72.

(5)

11. Suzuki H, Harpaz N, Tarmin L et al. Microsatellite instabil- ity in ulcerative colitis-associated colorectal dysplasias and cancers. Cancer Res 1994; 54: 4 8 4 1 ^ .

12. Kern SE, Redston M, Seymour AB et al. Molecular genetic profiles of colitis-associated neoplasms. Gastroenterology 1994; 107: 420-8.

13. Brentnall TA, Crispin DA, Bronner MP et al. Microsatellite instability in nonneoplastic mucosa from patients with chronic ulcerative colitis. Cancer Res 1996; 56: 1237-40.

14. Cravo ML, Albuquerque CM, Salazar de Sousa L et al MicrosatelUte instability in non-neoplastic mucosa of pa- tients with ulcerative colitis: effect of folate supplementa- tion. Am J Gastroenterol 1998; 93: 2060-4.

15. Fogt F, Vortmeyer AO, Goldman H, Giordano TJ, Merino MJ, Zhuang Z. Comparison of genetic alterations in colonic adenoma and ulcerative colitis-associated dysplasia and carcinoma. Hum Pathol 1008; 29: 131-6.

16. Fogt F, Urbanski SJ, Sanders ME et al. Distinction between dysplasia-associated lesion or mass (DALM) and adenoma in patients with ulcerative colitis. Hum Pathol 2000; 31:

288-91.

17. Souza RF, Garrigue-Antar L, Lei J et al. Alterations of transforming growth factor-beta 1 receptor type II occur in ulcerative colitis-associated carcinomas, sporadic colorectal neoplasms, and esophageal carcinomas, but not in gastric neoplasms. Hum Cell 1996; 9: 229-36.

18. Souza RF, Lei J, Yin J et al. A transforming growth factor beta 1 receptor type II mutation in ulcerative colitis- associated neoplasms. Gastroenterology 1997; 112: 40-5.

19. Grady W, Rajput A, Myerof L, Markowitz S. What's new with RII? Gastroenterology 1997; 112: 297-302.

20. Leach FS, Nicolaides NC, Papadopoulos N et al. Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell 1993; 75: 1215-25.

21. Fishel R, Kolodner RD. Identification of mismatch repair genes and their role in the development of cancer. Curr Opin Genet Dev 1995; 5: 382-95.

22. Jiricny L Colon cancer and DNA repair: have mismatches met their match? Trends Genet 1994; 10: 164-8.

23. Parsons R, Li GM, Longley MJ et al. Hypermutability and mismatch repair deficiency in RER+ tumor cells. Cell 1993;

75: 1227-36.

24. Liu B, Nicolaides NC, Markowitz S et al. Mismatch repair gene defects in sporadic colorectal cancers with microsatel- lite instability. Nat Genet 1995; 9: 48-55.

25. Fleisher AS, Esteller M, Harpaz N et al. Microsatellite instability in inflammatory bowel disease-associated neo- plastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLHl. Cancer Res 2000; 60: 4864-8.

26. Hsieh CJ, Klump B, Holzmann K, Borchard F, Gregor M, Porschen R. Hypermethylation of the pl6INK4a promoter in colectomy specimens of patients with long-standing and extensive ulcerative colitis Cancer Res 1998; 58: 3942-5.

27. Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns with a complemen- tary DNA microarray. Science 1995; 270: 467-70.

28. DeRisi J, Penland L, Brown PO et al. Use of cDNA microarray to analyse gene expression patterns in human cancer. Nature Genetics 1996; 14: 457-60.

29. Heller RA, Schena M, Chai A et al. Discovery and analysis of inflammatory disease-related genes using cDNA micro- arrays. Proc Natl Acad Sci USA 1997; 94: 2150-5.

30. Chuaqui RF, Cole KA, Emmert-Buck MR, Merino M I Histopathology and molecular biology of ovarian epithelial tumors Ann Diagn Pathol 1998; 2: 195-207.

31. Duggan DJ, Bittner M, Chen Y, Meltzer P, Trent J. M.

Expression profiling using cDNA microarrays. Nat Genet 1999; 21: 10-4.

32. Friend SH. How DNA microarrays and expression profiling win affect clinical practice Br Med J 1999; 319: 1306-7.

33. Golub TR, Slonim DK, Tamayo P et al. Molecular classifi- cation of cancer: class discovery and class prediction by gene expression monitoring. Science 1999; 286: 531-7.

34. Khan J, Saal LH, Bittner ML, Chen Y, Trent JM, Meltzer PS. Expression profiling in cancer using cDNA microarrays.

Electrophoresis 1999; 20: 223-9.

35. Khan J, Bittner ML, Chen Y, Meltzer PS, Trent JM. DNA microarray technology: the anticipated impact on the study of human disease. Biochim Biophys Acta 1999; 1423: M 1 7 - 28.

36. Fozard JB, Quirke P, Dixon MF, Giles GR, Bird C. C. DNA aneuploidy in ulcerative colitis Gut 1986; 27: 1414-8.

37. Fozard JB, Quirke P, Dixon MF. D N A aneuploidy in ulcerative colitis. Gut 1987; 28: 642-4.

38. Levine DS, Rabinovitch PS, Haggitt RC et al. Distribution of aneuploid cell populations in ulcerative colitis with dysplasia or cancer. Gastroenterology 1991; 101: 1198-210.

39. Porschen R, Robin U, Schumacher A et al. DNA aneuploidy in Crohn's disease and ulcerative colitis: results of a com- parative flow cytometric study. Gut 1992; 33: 663-7.

40. Rubin CE, Haggitt RC, Burmer GC et al. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology 1992; 103: 1611-20.

41. Navratil E, Stettler C, Paul G et al. Assessment of dysplasia, mucosal mucins, p53 protein expression, and DNA content in ulcerative colitis patients with colectomy and ileorectal anastomosis. Scand J Gastroenterol 1995; 30: 361-6.

42. Klump B, Holzmann K, Kuhn A et al. Distribution of cell populations with DNA aneuploidy and p53 protein expres- sion in ulcerative colitis Eur J Gastroenterol Hepatol 1997;

9: 789-94.

43. Holzmann K, Klump B, Borchard F et al. Comparative analysis of histology, DNA content, p53 and Ki-ras muta- tions in colectomy specimens with long-standing ulcerative colitis. Int J Cancer 1998; 76: 1-6.

44. Hammarberg C, Slezak P, Tribukait B. Early detection of malignancy in ulcerative colitis. A flow-cytometric DNA study. Cancer 1984; 53: 291-5.

45. Hammarberg C, Rubio C, Slezak P, Tribukait B, Ohman U Flow-cytometric DNA analysis as a means for early detec- tion of mahgnancy in patients with chronic ulcerative colitis.

Gut 1984; 25: 905-8.

46. Melville DM, Jass JR, Shepherd NA et al. Dysplasia and deoxyribonucleic acid aneuploidy in the assessment of precancerous changes in chronic ulcerative colitis. Observer variation and correlations. Gastroenterology 1988; 95: 668- 75.

47. Melville DM, Jass JR, Morson BC et al. Observer study of the grading of dysplasia in ulcerative colitis: comparison with clinical outcome. Hum Pathol 1989; 20: 1008-14.

48. Lofberg R, Tribukait B, Ost A, Brostrom O, Reichard H.

Flow cytometric DNA analysis in longstanding ulcerative colitis: a method of prediction of dysplasia and carcinoma development? Gut 1987; 28: 1100-6.

49. Rutegard J, Ahsgren L, Stenling R, Roos G. DNA content and mucosal dysplasia in ulcerative colitis. Flow cytometric analysis in patients with dysplastic or indefinite morpholo- gic changes in the colorectal mucosa. Dis Colon Rectum 1989; 32: 1055-19.

50. Rutegard J, Ahsgren L, Stenling R, Roos G. DNA content in ulcerative colitis. Flow cytometric analysis in a patient series from a defined area. Dis Colon Rectum 1988; 31: 710- 5.

51. Levine DS, Reid BJ, Haggitt RC, Rubin CE, Rabinovitch PS. Correlation of ultrastructural aberrations with dysplasia and flow cytometric abnormalities in Barrett's epithelium.

Gastroenterology 1989; 96: 355-67.

(6)

52. Reid BJ, Blount PL, Rubin CE et al. Flow-cytometric and histological progression to malignancy in Barrett's esopha- gus: prospective endoscopic surveillance of a cohort. Gas- troenterology 1992; 102: 1212-9.

53. Kahn S, Yamamoto F, Almoguera C et al. The c-K-ras gene and human cancer (Review). Anticancer Res 1987; 7: 639- 52.

54. Bos JL, Fearon ER, Hamilton SR et al. Prevalence of ras gene mutations in human colorectal cancers. Nature 1987;

327: 293-7.

55. Forrester K, Almoguera C, Han K, Grizzle WE, Perucho M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature 1987; 327: 298-303.

56. Burmer GC, Rabinovitch PS, Loeb LA. Analysis of c-Ki-ras mutations in human colon carcinoma by cell sorting, poly- merase chain reaction, and DNA sequencing. Cancer Res 1989; 49: 2141 6.

57. Delattre O, Olschwang S, Law DJ et al. Multiple genetic alterations in distal and proximal colorectal cancer. Lancet

1989; 2: 353 6.

58. Bodmcr WF, Cotlrcll S, Frischauf AM et al. Genetic analysis of colorectal cancer. Princess Takamatsu Symp 1989; 20: 49 59.

59. Mcltzcr SJ, Ahncn DJ, Baltifora H, Yokola J, Cline MJ.

Protooncogcne abnormalities in colon cancers and adeno- matous polyps [Published erratum appears in Gastroenter- ology 1987; 93: 223]. Gastroenterology 1987; 92: 1174 80.

60. Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 1988; 53: 549 54.

61. Meltzcr SJ, Zhou D, Weinstein WM. Tissue-specific expres- sion of c-Ha-ras in premalignant gastrointestinal mucosae.

Exp Mol Pathol 1989; 51: 264 74.

62. Redston MS, Papadopoulos N, Caldas C, Kinzler KW, Kern SE. Common occurrence of APC and K-ras gene mutations in the spectrum of colitis-associated neoplasias.

Gastroenterology 1995; 108: 383 92.

63. Suzui M, Ushijima T, Yoshimi N et al. No involvement of APC gene mutations in ulcerative colitis-associated rat colon carcinogenesis induced by 1-hydroxyanthraquinone and methylazoxymethanol acetate. Mol Carcinogenet 1997;

20: 389 93.

64. Feinberg AP. The molecular biology of human cancer. Prog Clin Biol Res 1985; 198: 279 92.

65. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu onco- gene. Science 1987; 235: 177-82.

66. Lu SH, Hsieh LL, Luo EC, Weinstein IB. Amplification of the EGF receptor and c-myc genes in human esophageal cancers. Int J Cancer 1988; 42: 502-5.

67. Mallet MK, Mane SM, Meltzer SJ, Needleman SW. c-myc amplification coexistent with activating N-ras point muta- tion in the biphenotypic leukemic cell line Red-3. Leukemia 1989; 3: 511-1-5.

68. Bigner SH, Friedman HS, Vogelstein B, Oakes WJ, Bigner DD. Amplification of the c-myc gene in human medullo- blastoma cell lines and xenografts [Published erratum appears in Cancer Res 1990; 50: 3809]. Cancer Res 1990;

50: 2347-50.

69. Jiang W, Kahn SM, Tomita N, Zhang YJ, Lu SH, Weinstein IB. Amplification and expression of the human cyclin D gene in esophageal cancer. Cancer Res 1992; 52: 2980-3.

70. Zhang YJ, Jiang W, Chen CJ et al. Amplification and overexpression of cyclin D l in human hepatocellular carci- noma. Biochem Biophys Res Commun 1993; 196: 1010-6.

71. Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW, Vogelstein B. Oncoprotein MDM2 conceals the activa-

tion domain of tumour suppressor p53. Nature 1993; 362:

857-60.

72. Kauraniemi P, Hedenfalk I, Persson K et al. MYB oncogene amplification in hereditary BRCAl breast cancer. Cancer Res 2000; 60: 5323-8.

73. Weinberg R. Tumor suppressor genes. Science 1991; 254:

1138-461.

74. Vogelstein B, Fearon ER, Hamilton SR et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988; 319: 525-32.

75. Baker SJ, Preisinger AC, Jessup JM et al. p53 gene muta- tions occur in combination with 17p allelic deletions as late events in colorectal tumorigenesis. Cancer Res 1990; 50:

7717-22.

76. Devilee P, van Vliet M, van Sloun P et al. Allelotype of human breast carcinoma: a second major site for loss of heterozygosity is on chromosome 6q. Oncogene 1991; 6:

1705 11.

77. Sato T, Saito H, Morita R, Koi S, Lee J, Nakamura Y.

Allelotype of human ovarian cancer. Cancer Res 1991; 51:

5118 22.

78. Aoki T, Mori T, Du X, Nisihira T, Matsubara T, Nakamura Y. Allelotype study of esophageal carcinoma. Genes Chro- mosomes Cancer 1994; 10: 177 82.

79. Nawroz H, van dcr Riet P, Hruban RH, Koch W, Ruppert JM, Sidransky D. Allelotype of head and neck squamous cell carcinoma. Cancer Res 1994; 54: 1152 5.

80. Seymour AB, Hruban RH, Redston M et al. Allelotype of pancreatic adenocarcinoma. Cancer Res 1994; 54: 2761 4.

81. Field JK, Kiaris H, Risk JM ('/ al. Allelotype of squamous cell carcinoma of the head and neck: fractional allele loss correlates with survival [Published erratum appears in Br J Cancer 1996; 74: 1153]. Br J Cancer 1995; 72: 1180 8.

82. Rosin MP, Cairns P, Epstein JI, Schocnberg MP, Sidransky D. Partial allelotype of carcinoma in situ of the human bladder. Cancer Res 1995; 55: 5213 16.

83. Califano JA, Johns MMr, Westra WH ('/ al. An allelotype of papillary thyroid cancer. Int J Cancer 1996; 69: 442 4.

84. Johns MMr, Westra WH, Califano JA, Eisele D, Koch WM, Sidransky D. Allelotype of salivary gland tumors. Cancer Res 1996; 56: 1151 4.

85. Hammond ZT, Kaleem Z, Cooper JD, Sundaresan R, Patterson GA, Goodfellow PJ. Allelotype analysis of eso- phageal adenocarcinomas: evidence for the involvement of sequences on the long arm of chromosome 4. Cancer Res 1996; 65: 4499 502.

86. Dolan K, Garde J, Gosney J et al. Allelotype analysis of oesophageal adenocarcinoma: loss of heterozygosity occurs at multiple sites. Br J Cancer 1998; 78: 950-7.

87. Yustein AS, Harper JC, Petroni GR, Cummings OW, Moskaluk CA, Powell SM. Allelotype of gastric adenocarci- noma. Cancer Res 1999; 59: 1437-41.

88. Chang M, Tsuchiya K, Batchelor RH et al. Deletion map- ping of chromosome 8p in colorectal carcinoma and dyspla- sia arising in ulcerative colitis, prostatic carcinoma, and malignant fibrous histiocytomas. Am J Pathol 1994; 144:

1-6.

89. Hoque AT, Hahn SA, Schutte M, Kern SE. DPC4 gene mutation in colitis associated neoplasia. Gut 1997; 40: 120- 2.

90. Fogt F, Vortmeyer AO, Stolte M et al. Loss of heterozygosity of the von Hippel Lindau gene locus in polypoid dysplasia but not flat dysplasia in ulcerative colitis or sporadic adenomas. Hum Pathol 1998; 29: 961-4.

91. Zou TT, Lei J, Shi YQ et al. FHIT gene alterations in esophageal cancer and ulcerative colitis (UC). Oncogene 1997; 15: 101-5.

(7)

92. Tomlinson I, Ilyas M, Johnson V ei al. comparison of the genetic pathways involved in the pathogenesis of three types of colorectal cancer. J Pathol 1998; 184: 148-52.

93. Odze RD, Brown CA, Hartmann CJ, Noffsinger AE Fogt F.

Genetic alterations in chronic ulcerative colitis-associated adenoma-like DALMs are similar to non-colitic sporadic adenomas. Am J Surg Pathol 2000; 24: 1209-16.

94. Fearon EKC, Nigro J, Kern S et al. Identification of a chromosome 18q gene that is altered in colorectal cancers.

Science 1989; 247: 49-56.

95. Wang XW, Harris CC. p53 tumor-suppressor gene: clues to molecular carcinogenesis. J Cell Physiol 1997; 173: 247-55.

96. Harris CC. p53: at the crossroads of molecular carcinogen- esis and molecular epidemiology. J Invest Dermatol Symp Procl996; 1: 115-18.

97. Yin J, Harpaz N, Tong Y et al. J. p53 point mutations in dysplastic and cancerous ulcerative colitis lesions. Gastro- enterology 1993;104:1633-9.

98. Brentnall TA, Crispin DA, Rabinovitch PS et al. Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology 1994; 107: 369-78.

99. Harpaz N, Peck AL, Yin J et al. p53 protein expression in ulcerative colitis-associated colorectal dysplasia and carci- noma. Hum Pathol 1994; 25: 1069-74.

100. Groden J, Thliveris A, Samowitz W^/ al. Identification and characterization of the familial adenomatous polyposis coH gene. Cell 1991;66:589-600.

101. Joslyn G, Carlson M, Thliveris A et al. Identification of deletion mutations and three new genes at the familial polyposis locus. Cell 1991; 66: 601-13.

102. Kinzler KW, Nilbert MC, Su LK etal. Identification of FAP locus genes from chromosome 5q21. Science 1991; 253:

661-5.

103. Powell SM, Zilz N, Beazer-Barclay Y et al. APC mutations occur early during colorectal tumorigenesis. Nature 1992;

359: 235-7.

104. Sidransky D. Is human patched the gatekeeper of common skin cancers? Nat Genet 1996; 14: 7-8.

105. Nakamura Y, Nishisho I, Kinzler KW et al. Mutations of the adenomatous polyposis coli gene in familial polyposis coli patients and sporadic colorectal tumors. Princess Taka- matsu Symp 1991; 22: 285-92.

106. Cho KR, Vogelstein B. Suppressor gene alterations in the colorectal adenoma-carcinoma sequence. J Cell Biochem Suppl 1992; 16G: 1 3 7 ^ 1 .

107. Baba S. Recent advances in molecular genetics of colorectal cancer. World J Surg 1997; 21: 678-87.

108. Ilyas M, Tomlinson IP. Genetic pathways in colorectal cancer. Histopathology 1996; 28: 389-99.

109. Miyoshi Y, Nagase H, Ando H et al. Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. Hum Mol Genet 1992; 1: 229-33.

110. Tarmin L, Yin J, Harpaz N et al. Adenomatous polyposis coh gene mutations in ulcerative colitis-associated dyspla- sias and cancers versus sporadic colon neoplasms. Cancer Res 1995; 55: 2035-8.

111. Taken SJ, Petersen GM, Gruber SB et al. Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC.

Nat Genet 1997; 17:79-83.

112. Frayling IM, Beck NE, Ilyas M et al. The APC variants I1307K and E1317Q are associated with colorectal tumors, but not always with a family history. Proc Natl Acad Sci USA 1998; 95: 10722-7.

113. Gryfe R, Di Nicola N, Galhnger S, Redston M. Somatic instability of the APC I1307K allele in colorectal neoplasia.

Cancer Res 1998; 58: 4040-3.

114. Woodage T, King SM, Wacholder S et al. The APCI1307K allele and cancer risk in a community-based study of Ashkenazi Jews. Nat Genet. 20: 62-5, 1998.

115. Yin J, Harpaz N, Souza R F et al. Low prevalence of the APC I1307K sequence in Jewish and non-Jewish patients with inflammatory bowel disease. Oncogene 1999; 18: 3902- 4.

116. lonov Y, Peinado MA, Malkhosyan S, Shibata D, and Perucho M. Ubiquitous somatic mutations in simple re- peated sequences reveal a new mechanism for colonic carcinogenesis. Nature 1993; 363: 558-61.

117. Aaltonen LA, Peltomaki P, Leach FS et al. Clues to the pathogenesis of familial colorectal cancer. Science 1993;

260:812-16.

118. Thibodeau SN, Bren G, Schaid D. Microsatellite instabiUty in cancer of the proximal colon. Science 1993; 260: 816-19.

119. Aaltonen LA, Peltomaki P. Genes involved in hereditary nonpolyposis colorectal carcinoma Anticancer Res 1994;

14:1657-60.

120. Bronner CE, Baker SM, Morrison YY et al. Mutation in the DNA mismatch repair gene homologue hMLHl is asso- ciated with hereditary non-polyposis colon cancer. Nature 1994;368:258-61.

121. Koi M, Umar A, Chauhan DP et al. Human chromosome 3 corrects mismatch repair deficiency and microsatellite in- stability and reduces #-methyl-A^'-nitro-A^-nitrosoguanidine tolerance in colon tumor cells with homozygous hMLHl mutation [Published erratum appears in Cancer Res 1995;

55: 201]. Cancer Res 1994; 54: 4308-12.

122. Vasen HFA, Mecklin J-P, Kahn PM, Lynch HT. Hereditary non-polyposis colorectal cancer. Lancet 1991; 338: 887.

123. Benatti P, Sassatelli R, Roncucci L et al. Tumour spectrum in hereditary non-polyposis colorectal cancer (HNPCC) and in families with 'suspected H N P C C A population- based study in northern Italy. Colorectal Cancer Study Group. Int J Cancer 1993; 54: 371-7.

124. Peltomaki P, Lothe RA, Aaltonen LA et al. Microsatellite instability is associated with tumors that characterize the hereditary non-polyposis colorectal carcinoma syndrome.

Cancer Res 1993;53:5853-5.

125. Meltzer SJ, Yin J, Manin B et al. Microsatellite instability occurs frequently and in both diploid and aneuploid cell populations of Barrett's-associated esophageal adenocarci- nomas. Cancer Res 1994; 54: 3379-82.

126. Rhyu MG, Park WS, Meltzer SJ. Microsatellite instability occurs frequently in human gastric carcinoma. Oncogene 1994; 9: 29-32.

127. Berg PE, Liu J, Yin J, Rhyu MG, Frantz CN, Meltzer, SL Microsatellite instability is infrequent in neuroblastoma.

Cancer Epidemiol Biomarkers Prev 1995; 4: 907-9.

128. Brentnall TA, Chen R, Lee JG et al. Microsatellite instabil- ity and K-ras mutations associated with pancreatic adeno- carcinoma and pancreatitis. Cancer Res 1995; 55: 4264-7.

129. Eshleman JR, Markowitz SD. Microsatellite instability in inherited and sporadic neoplasms. Curr Opin Oncol 1995;

7: 83-9.

130. Boland CR, Thibodeau SN, Hamilton SR et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsa- telhte instability in colorectal cancer. Cancer Res 1998; 58:

5248-57.

131. Perucho M. Correspondence re: C R . Boland et al, A National Cancer Institute workshop on microsatellite in- stability for cancer detection and familial predisposition:

development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res.

1998; 58: 5248-57. Cancer Res 1999; 59: 249-56.

132. Eshleman JR, Markowitz SD. Mismatch repair defects in human carcinogenesis. Hum Mol Genet (5 Spec. No.) 1996;

5:1489-94.

(8)

133. Fishel R, Lescoe MK, Rao MR et al The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer [Published erratum appears in Cell 1994; 77: 167]. Cell 1993; 75: 1027-38.

134. Nicolaides NC, Papadopoulos N, Liu B et al. Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature 1994; 371: 75-80.

135. Nystrom-Lahti M, Parsons R, Sistonen P et al. Mismatch repair genes on chromosomes 2p and 3p account for a major share of hereditary nonpolyposis colorectal cancer families evaluable by linkage. Am J Hum Genet 1994; 55: 659-65.

136. Papadopoulos N, Nicolaides NC, Wei YF et al. Mutation of a mutL homolog in hereditary colon cancer. Science 1994;

263:1625-9.

137. Liu B, Parsons R, Papadopoulos N et al. Analysis of mismatch repair genes in hereditary non-polyposis color- ectal cancer patients. Nat Med 1996; 2: 169-74.

138. Nystrom-Lahti M, Wu Y, Moisio AL et al. DNA mismatch repair gene mutations in 55 kindreds with verified or putative hereditary non-polyposis colorectal cancer. Hum Mol Genet 1996; 5: 763 9.

139. Akiyama Y, Sato H, Yamada T<'/ al. Germ-line mutation of the hMSH6/GTBP gene in an atypical hereditary nonpoly- posis colorectal cancer kindred. Cancer Res 1997; 57: 3920 3.

140. Papadopoulos N, Lindblom A. Molecular basis of HNPCC:

mutations of MMR genes. Hum Mutat 1997; 10: 89 99.

141. Lipkin SM, Wang V, Jacoby R ('/ al. MLH3: a DNA mismatch repair gene associated with mammalian micro- satellite instability. Nat Genet 2000; 24: 27 35.

142. Borresen AL, Lothe RA, Meling GI et al. Somatic muta- tions in the hMSH2 gene in microsatellitc unstable color- ectal carcinomas. Hum Mol Genet 1995; 4: 2065 72.

143. Moslein G, Tester DJ, Lindor NM et al. Microsatellitc instability and mutation analysis of hMSH2 and hMLHl in patients with sporadic, familial and hereditary colorectal cancer. Hum Mol Genet 1996; 5: 1245-52.

144. Cunningham JM, Christensen ER, Tester DJ et al. Hyper- methylation of the hMLHl promoter in colon cancer with microsatellitc instability. Cancer Res 1998; 58: 3455-60.

145. Esteller M, Levine R, Baylin SB, Ellenson LH, Herman JG.

MLHl promoter hypermethylation is associated with the microsatellitc instability phenotype in sporadic endometrial carcinomas. Oncogene 1998; 17: 2413-7.

146. Herman JG, Umar A, Polyak K et al. Incidence and functional consequences of hMLHl promoter hypermethy- lation in colorectal carcinoma. Proc Natl Acad Sci USA 1998;95:6870-5.

147. Esteller M, Catasus L, Matias-Guiu X et al. hMLHl promoter hypermethylation is an early event in human endometrial tumorigenesis. Am J Pathol 1999; 155: 1767 72.

148. Fleisher AS, Esteller M, Wang S et al. Hypermethylation of the hMLHl gene promoter in human gastric cancers with microsatellitc instability. Cancer Res 1999; 59: 1090 5.

149. Eads CA, Danenberg KD, Kawakami K, Saltz LB, Danen- berg PV, Laird PW. CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltrans- ferasc overexpression. Cancer Res 1999; 59: 2302 6.

150. Fleisher AS, Esteller M, Tamura G et al. Hypermethylation of the hMLHl gene promoter is associated with microsa- tellitc instability in early human gastric neoplasia. Onco- gene 2001; 20: 329 35.

Riferimenti

Documenti correlati

[r]

Fig. Discrete correlation function for the light curves of GASP- WEBT and RXTE/ASM for Period 2. The description of data points and contours are given in the caption of Fig.

I componenti presi in esempio hanno consumo medio giornaliero simile, deviazione standard del consumo medio giornaliero simile, Lead Time simile e peso simile

The main purpose is to investigate mixing processes among fluids from different sources flowing into the carbonate formations and to describe how the chemical composition of the gas

Moreover, if the choice for the type of rectal dis- section in patients with a diagnosed rectal cancer is clear, in the case of prophylactic proctocolectomy in males younger than

Direct interaction; I2D predictions of protein protein interactions for Homo sapiens using Krogan-Greenblatt-2006 Saccharomyces cerevisiae data; Pubmed 16554755; I2D

I nuovi elementi emersi in queste ricerche sono stati al centro di un serrato dibattito 11 , che ha visto una gamma di sfumature tra due posizioni estreme: da un lato chi vedeva

techniques and constructive solutions that becomes an unicum in architecture: roofing, masonry, road paving, doors and windows, chimneys, in their geometries and materials