12
Molecular Genetic Basis of Pancreatic Adenocarcinoma Werner Hilgers and Scott E. Kern* Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland Pancreatic ductal adenocarcinoma is a complex genetic disease. As might be expected for a malignancy that is rather homogeneous in clinical presentation and behavior, a distinct subset of genes are found to be genetically inactivated in a majority of the tumors. A yet larger subset of genes experiences genetic inactivation at much lower frequencies. The latter subset could solely reflect a somewhat trivial genetic heterogeneity of the tumor, but more likely will represent the initial insights into pathways whose more widespread importance will be shown in future work. Familial pancreatic cancer susceptibility underlies a significant fraction of the overall incidence. Genetic testing is feasible for many of the causative genes, although the clinical utility remains unsettled. The precursor lesion for pancreatic cancer shares some of the genetic lesions of the more advanced invasive stage, and follows a stepwise progression model both histologically and genetically. Genes Chromosomes Cancer 26:1–12, 1999. r 1999 Wiley-Liss, Inc. INTRODUCTION Pancreatic cancer is among the best-described genetic diseases. The accumulation of past and recent discoveries now offers an opportune time for consideration of some broader issues, such as the likely direction and difficulties of future research. The study of pancreatic cancer has also had general utility in the understanding of the biology of eukaryotic regulatory pathways and of other human neoplasms, many of which are less completely described in genetic terms. Clinically, pancreatic cancer is the fifth leading cause of cancer death in the United States and is remarkably homogeneous, presenting late in the course of the disease and usually fatal within a year or so. Pathologically, most cases are moderately differentiated with abortive duct formation and an intense desmoplastic reac- tion. While unfortunately still of lagging interest in clinical circles, pancreatic cancer is of mainstream interest as a scientific model of a common cancer type in adults. PREINVASIVE NEOPLASIA Pancreatic ductal epithelium constitutes 5% or less of the total cellular population of the pancreas, yet is the origin of the common form of pancreatic carcinoma (Cubilla and Fitzgerald 1976; Kozuka et al., 1979; Pour et al., 1982; Furukawa et al., 1994; Brat et al., 1998; Hruban et al., 1998). It is a low cuboidal epithelium with basal nuclei. In the earli- est form of neoplasia, which is unfortunately but historically termed a ‘‘flat hyperplasia,’’ the epithe- lium becomes columnar. Half of these lesions, when microdissected, prove to contain K-ras muta- tions. A more advanced stage, ‘‘papillary hyperpla- sia,’’ is distinguished by a crowded mucosa that assumes a folded architecture. (This is reasonably reminiscent of the colorectal adenoma, which be- comes so folded that it acquires its own inappropri- ate name, ‘‘villous adenoma.’’ In most cases this appearance is an artifact of a cerebriform, infolded surface pattern that is viewed in cross-section.) Papillary hyperplasia can exhibit varying degrees of cellular and nuclear atypia, or even an architectur- ally disordered amassing of cells sometimes termed (again, unfortunately) as ‘‘carcinoma in situ.’’ The vast majority of papillary hyperplasias contain a mutation of the K-ras (KRAS2) gene (Caldas et al., 1994b). These lesions become a bona fide carci- noma with the development of invasion through the wall of the duct. The carcinoma stage almost always engenders a dense collagenous and inflam- matory reaction termed ‘‘desmoplasia’’ in the in- vaded tissue. Because of the desmoplastic re- sponse, the cancer cells, on average, represent about 25% of the cells of the tumor, and often many fewer (Seymour et al., 1994). The genetic analysis of the primary tumor is therefore an analysis largely of normal cells. Most genetic analyses of pancreatic cancer therefore must first enrich for the neoplastic component, by microdissection or by establishment Supported by: NIH SPORE (Specialized Program of Research Excellence) in Gastrointestinal Cancer; Grant number: CA62924; Deutsche Krebshilfe (W.H.). *Correspondence to: Scott E. Kern, 632 Ross Building, Johns Hopkins University School of Medicine, Baltimore, MD 21205. E-mail: [email protected] Received 30 December 1998; Accepted 17 March 1999 GENES, CHROMOSOMES & CANCER 26:1–12 (1999) 3 REVIEW ARTICLE 3 r 1999 Wiley-Liss, Inc.

Molecular genetic basis of pancreatic adenocarcinoma

Embed Size (px)

Citation preview

Page 1: Molecular genetic basis of pancreatic adenocarcinoma

Molecular Genetic Basis of PancreaticAdenocarcinoma

Werner Hilgers and Scott E. Kern*

Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland

Pancreatic ductal adenocarcinoma is a complex genetic disease. As might be expected for a malignancy that is ratherhomogeneous in clinical presentation and behavior, a distinct subset of genes are found to be genetically inactivated in a majorityof the tumors. A yet larger subset of genes experiences genetic inactivation at much lower frequencies. The latter subset couldsolely reflect a somewhat trivial genetic heterogeneity of the tumor, but more likely will represent the initial insights intopathways whose more widespread importance will be shown in future work. Familial pancreatic cancer susceptibility underlies asignificant fraction of the overall incidence. Genetic testing is feasible for many of the causative genes, although the clinical utilityremains unsettled. The precursor lesion for pancreatic cancer shares some of the genetic lesions of the more advanced invasivestage, and follows a stepwise progression model both histologically and genetically. Genes Chromosomes Cancer 26:1–12,1999. r 1999 Wiley-Liss, Inc.

INTRODUCTION

Pancreatic cancer is among the best-describedgenetic diseases. The accumulation of past andrecent discoveries now offers an opportune time forconsideration of some broader issues, such as thelikely direction and difficulties of future research.The study of pancreatic cancer has also had generalutility in the understanding of the biology ofeukaryotic regulatory pathways and of other humanneoplasms, many of which are less completelydescribed in genetic terms. Clinically, pancreaticcancer is the fifth leading cause of cancer death inthe United States and is remarkably homogeneous,presenting late in the course of the disease andusually fatal within a year or so. Pathologically, mostcases are moderately differentiated with abortiveduct formation and an intense desmoplastic reac-tion. While unfortunately still of lagging interest inclinical circles, pancreatic cancer is of mainstreaminterest as a scientific model of a common cancertype in adults.

PREINVASIVE NEOPLASIA

Pancreatic ductal epithelium constitutes 5% orless of the total cellular population of the pancreas,yet is the origin of the common form of pancreaticcarcinoma (Cubilla and Fitzgerald 1976; Kozuka etal., 1979; Pour et al., 1982; Furukawa et al., 1994;Brat et al., 1998; Hruban et al., 1998). It is a lowcuboidal epithelium with basal nuclei. In the earli-est form of neoplasia, which is unfortunately buthistorically termed a ‘‘flat hyperplasia,’’ the epithe-lium becomes columnar. Half of these lesions,when microdissected, prove to contain K-ras muta-

tions. A more advanced stage, ‘‘papillary hyperpla-sia,’’ is distinguished by a crowded mucosa thatassumes a folded architecture. (This is reasonablyreminiscent of the colorectal adenoma, which be-comes so folded that it acquires its own inappropri-ate name, ‘‘villous adenoma.’’ In most cases thisappearance is an artifact of a cerebriform, infoldedsurface pattern that is viewed in cross-section.)Papillary hyperplasia can exhibit varying degrees ofcellular and nuclear atypia, or even an architectur-ally disordered amassing of cells sometimes termed(again, unfortunately) as ‘‘carcinoma in situ.’’ Thevast majority of papillary hyperplasias contain amutation of the K-ras (KRAS2) gene (Caldas et al.,1994b). These lesions become a bona fide carci-noma with the development of invasion throughthe wall of the duct. The carcinoma stage almostalways engenders a dense collagenous and inflam-matory reaction termed ‘‘desmoplasia’’ in the in-vaded tissue. Because of the desmoplastic re-sponse, the cancer cells, on average, representabout 25% of the cells of the tumor, and often manyfewer (Seymour et al., 1994). The genetic analysisof the primary tumor is therefore an analysis largelyof normal cells. Most genetic analyses of pancreaticcancer therefore must first enrich for the neoplasticcomponent, by microdissection or by establishment

Supported by: NIH SPORE (Specialized Program of ResearchExcellence) in Gastrointestinal Cancer; Grant number: CA62924;Deutsche Krebshilfe (W.H.).

*Correspondence to: Scott E. Kern, 632 Ross Building, JohnsHopkins University School of Medicine, Baltimore, MD 21205.E-mail: [email protected]

Received 30 December 1998; Accepted 17 March 1999

GENES, CHROMOSOMES & CANCER 26:1–12 (1999)

333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333 REVIEW ARTICLE 33333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333333

r 1999 Wiley-Liss, Inc.

Page 2: Molecular genetic basis of pancreatic adenocarcinoma

of cell lines and xenografts. Analysis of the earlier-stage intraductal neoplasms, of course, requiresmicrodissection or in situ labeling techniques.

Because K-ras mutations are not required for theearly flat stage but have a high prevalence in thepapillary stage, it appears that K-ras mutations mayprovide a growth advantage for the middle stages ofthis neoplastic progression, but do not serve a truegatekeeper role. Similarly, alterations of p16 geneticstructure or a loss of expression tend to representonly a subset of lesions, probably those that mighthave higher biologic potential for progression tocancer (Moskaluk et al., 1997; Wilentz et al., 1998).BRCA2 may serve such a late role as well; inpatients with a germline BRCA2 mutation, thepancreas does not harbor an obviously increasednumber of intraductal neoplasms, and the loss ofthe remaining wild-type BRCA2 allele appears toarise well after the development of the hyperplasia(Goggins et al., 1997b). This is perhaps an insight toexplain the observation that in pancreas cancerfamilies, cancer does not arrive at an acceleratedage. If the wild-type allele is lost at later stages ofneoplasia, it represents not a key change but analternate change, i.e., just another in a series ofrate-limiting steps. These genetic lesions them-selves might be in part age-dependent. Theseparticulars would mask the tie that would otherwiselink the germline mutations with a younger age ofdisease onset, a connection seen so reproduciblywith both neoplastic and nonneoplastic diseases ofother organs.

Estimates have been made that as many as half ofthe normal elderly population develop flat hyperpla-sias of pancreatic ducts, whereas less than 10% ofpersons have lesions with significant cellular atypia(Cubilla and Fitzgerald 1976; Kozuka et al., 1979).Considering the low chance that each lesion has todevelop into a cancer (probably near 1 in 500lesions will so progress) and the potentially highrisk of cancer from episodic activation of the pancre-atic enzymes carried by these ducts (as seen inhereditary pancreatitis), humans have truly evolveda remarkable ability to thwart the malignant careerpotential of this neoplastic farm club. It is with thisas a backdrop that one should regard the geneticpatterns seen in pancreatic cancer.

MULTIPLE GENETIC LESIONS

In terms of known mutations per tumor, pancre-atic cancers harbor more than any other tumor type.More than 40% of the chromosomal arms suffer lossof heterozygosity (Hahn et al., 1995). Some tumorshave mutations in five or more genes (Rozenblum

et al., 1997). The changes in pancreatic cancer areoften mirrored in the related biliary carcinomas,although generally at somewhat lower frequenciesin the latter. The genes involved can be divided intwo groups on the basis of genetic alterations intheir signaling/regulatory pathways: those of highfrequency and those of low (Table 1).

High-Frequency Changes

Oncogenes.

Mutations of the K-ras gene are present in over90% of pancreatic cancers (Almoguerra et al., 1988).This is the highest fraction of K-ras alteration foundin any human tumor type.

It is interesting that the K-ras genes of pancreaticcancer do not share the same mutational spectrumseen for this gene in colorectal cancer, the codon 12serine allele being vanishingly rare in the pancreasbut common in the colorectum (Forrester et al.,1987; Vogelstein et al., 1988). This perhaps reflectsan undetermined but surprisingly specific muta-genic influence. On occasion, two differing K-rasmutations are identified in a pancreatic cancer.Most often, this probably represents a focal artifactproduced by a cancer with one type of mutation,invading and obliterating a precursor duct lesionhaving a different mutation. Rarely, however, itdoes appear that two K-ras mutations can coexist ina single cancer clone.

TABLE 1. Genetic Profile of Pancreatic Carcinomaa

GeneGene

locationsFrequency

(%)Mutation

origin

OncogenesK-ras 12p12 95 Som.AKT2 19q13.1 10–20 Som.MYB 6q24 10 Som.

Tumor Suppressorsp16/RB1 9p21/13q14 .90 Som. . germ.p53 17p31 75 Som.DPC4 18q21 55 Som.BRCA2 13q13 5–10 Germ. . som.MKK4 17p13 4 Som.LKB1/STK11 19p13 5 Som. . germ.ALK5 9q21 1 Som.b

RER2/TGFBR2 /3p22 1 Som.b

RER1/TGFBR2 /3p22 3 Som. . germ.c

aReferences are given in the text. Som., (rate of) somatic mutation ormethylation; germ., (rate of) germline mutation.bSingle examples of homozygous deletion of the ALK5 gene and of theTGFBR2 gene have been identified in pancreatic cancer. In colorectalcancer, a single family having a germline mutation of the TGFBR2 gene isreported (Lu et al., 1998).cIn RER1 tumors, the mismatch repair defect is usually somatic in origin;the TGFBR2 alterations are somatic.

2 HILGERS AND KERN

Page 3: Molecular genetic basis of pancreatic adenocarcinoma

Unlike the seldom-mutated H-ras protein, theactivity of the K-ras protein is not solely dependenton farnesylation (James et al., 1995; Lerner et al.,1995). Consequently, the development of farnesyltransferase inhibitor drugs is of uncertain promisein the treatment K-ras mutant tumors. Many compo-nents of signal transduction that lie downstream ofthe ras genes have been discerned in artificialsystems, but no measurable downstream biologicalactivity is yet known that is reliably increasedamong K-ras mutant cells from human tumors.Finally, K-ras mutations are not specific for malig-nancy, being present in a significant number ofbenign human neoplasms and early clonal lesions(Vogelstein et al., 1988; Pretlow et al., 1993; Tada etal., 1993; Yanagisawa et al., 1993; Caldas et al.,1994b; Tada et al., 1996). This unfortunate factdampens (although does not eliminate) enthusiasmfor the application of K-ras mutation detection inscreening programs that will attempt to achieveearly cancer diagnosis. It has been fashionable towonder whether, in the K-ras wild-type cancers,another gene of the same pathway might have acomplementary oncogenic mutation. Yet, the oftendistinctive clinicopathological features of the K-raswild-type cancers suggests instead that they mightshare little in common with the K-ras-mutant tu-mors to hold such an expectation (Goggins et al.,1998a).

Tumor suppressor genes

The p16 (INK4a, CDKN2, MTS1), p53 (TP53), andDPC4 (MADH4, SMAD4) genes are geneticallyinactivated in half or more of pancreatic cancers(Caldas et al., 1994a; Pellegata et al., 1994; Redstonet al., 1994; Hahn et al., 1996b; Schutte et al., 1996;Rozenblum et al., 1997). The tumors with andwithout these mutations are overwhelmingly simi-lar in clinical and pathological features (Rozenblumet al., 1997). This high frequency implies that formost pancreatic neoplasms, in their earlier benignstages, the presence of these genes essentiallyprevents the progression to an invasive stage. If thisholds for most precursors, might it not hold for all?That is, are there genes in these pathways that arealtered in the remaining cancers, so that essentiallyall pancreatic cancers embody inactivation of thesethree pathways? And might this genetic homogene-ity underlie the clinical and pathological homogene-ity of the disease? These are extremely influentialviewpoints and have served to guide the identifica-tion of candidate genes for mutational studies.

For the p16 gene, this expectation is almostentirely borne out. p16 is an inhibitor of the

cyclinD-Cdk4 and cyclinD-Cdk6 mitotic kinasecomplexes that downregulate the activity of the cellcycle inhibitor protein Rb (Serrano et al., 1993).Within this pathway, therefore, mutational activa-tion of any one of the cyclin D, CDK4, and CDK6oncogenes appears to produce a disregulation ofcell division cycle control that is similar to that seenwith mutational inactivation of the p16 or RB1tumor suppressor genes. Tumors such as lungcancer and melanoma almost uniformly select forclones having alterations of any one of these genes,implicitly to remove this regulatory system (Shapiroet al., 1995; Bartkova et al., 1996; Kinoshita et al.,1996; Kratzke et al., 1996; Zuo et al., 1996). Whilepancreatic cancer can sustain mutations of RB1 onoccasion (Ruggeri et al., 1992), it overwhelminglytargets the p16 gene for inactivation, found in over95% of the cancers (Caldas et al., 1994a; Schutte etal., 1997). Germline mutations of p16 predispose topancreatic cancer (Whelan et al., 1995; Ciotti et al.,1996). In sporadic tumors, homozygous deletionsand intragenic mutations (missense, nonsense, andframeshift) of p16 are nearly identical in frequency,accounting for nearly 85% of cases (Caldas et al.,1994a). In most of the remaining cases transcriptionof the p16 gene is turned off in association withmethylation of its promoter (Schutte et al., 1997).Mutations of the CDK4 gene have not been identi-fied (Schutte et al., 1997). Cyclin D amplificationwas observed at a high rate in one study, but not inanother (Schutte et al., 1997).

p53 gene mutations are seen in 50%–75% of cases(Pellegata et al., 1994; Redston et al., 1994; Rozen-blum et al., 1997). Missense mutations vastly out-number the nonsense and frameshift mutations,and homozygous deletions are not described inpancreatic and in most other tumor systems. Theupstream controls and downstream actions of p53appear to be highly complex. The expression of p53is stimulated by a wide variety of cellular stresses.Mdm2 is a major downregulator of p53 that consti-tutes a negative feedback loop for controlling p53activity (Momand et al., 1992; Oliner et al., 1993;Chen et al., 1994) and serves as an oncogene whenactivated by amplification (usually in p53 wild-typetumors of mesodermal origin; Oliner et al., 1992;Leach et al., 1993). There are a large number ofgenes whose transcription is controlled by p53,including an arm effecting cell cycle control (thep21WAF1/CIP1 gene; El-Deiry et al., 1993), a separateapoptotic arm (Yonish-Rouach et al., 1991; Polyaket al., 1997), and many other genes whose tumor-related roles remain yet undefined (Polyak et al.,1997). Perhaps because of this complexity, it has

3MOLECULAR GENETIC BASIS

Page 4: Molecular genetic basis of pancreatic adenocarcinoma

remained unclear whether there is a definable p53pathway such that mutations upstream or down-stream of the MDM2/p53 axis would be observed inthose carcinomas that retain a wild-type p53 gene.

p53 mutations may represent a special case intumor genetics. The ability of p53 to bind DNA atspecific sequences is lost in essentially all mutantsthat are found in tumors, indicating that DNAbinding is central to its tumor suppressive role. Theability of mutant proteins to complex with andinhibit the DNA binding of wild-type p53 isthought to produce an effective impairment of theprotein that is expressed from the remaining wild-type allele (Kern et al., 1992). Consequently, thecompetitive survival of clones having sustained amutation of p53 is aided by the dominant negativeaction of the mutant p53 proteins. The specificityof this mechanism may account for the paucity ofnonsense, frameshift, and homozygous deletions inthe p53 gene, since the development of the firstnull allele would then not allow the formation ofinhibitory complexes and an immediate growthadvantage. The strength of the p53 selection sys-tem (i.e., that biallelic genetic inactivation is notrequired to exert a growth advantage) is not gener-ally considered to be common among tumor suppres-sor genes, and selective pressures for other genes ofan extended p53 suppressive pathway cannot beexpected to match those for p53. Thus, for manyreasons, it may prove difficult technically to demon-strate a universal involvement of the p53 pathwayin pancreatic cancer, although such an effort wouldbe of high impact if successful.

The tumor suppressor DPC4 was cloned uponthe mapping of a hotspot of homozygous deletionsin pancreatic and biliary cancer (Hahn et al., 1996b).It was the fourth marker to be investigated forhomozygous deletions in pancreatic cancer (DPC;Hahn et al., 1996a). DPC4 is inactivated in about55% of pancreatic cancers, being homozygouslydeleted in nearly 35% and suffering intragenicmutations in another 20% (Schutte et al., 1996).Germline mutations of DPC4 cause not pancreaticcancer but a number of nonneoplastic erosive andinflammatory lesions of the intestinal mucosa thatbecome polypoid with time, termed juvenile polypo-sis (Howe et al., 1998). As with any chronic inflam-matory lesions of the bowel, these polyps canstimulate the development of dysplasia and cancer(Giardiello et al., 1991). Here, a direct role for DPC4(due to its mutation) in this progression is sug-gested in studies of accelerated progression of

colorectal neoplasia in transgenic mice mutant forboth the APC and DPC4 genes (Takaku et al., 1998).

DPC4 is a member of the Smad family of genesrelated to the Drosophila Mad and C. elegans Smagenes (Sekelsky et al., 1995; Hahn et al., 1996b;Savage et al., 1996). It is essential for mammaliandevelopment (Sirard et al., 1998). Smad proteinsmediate signals initiated by the binding of TGFb-like cytokines to their receptors (Sekelsky et al.,1995; Savage et al., 1996). Dpc4 protein is cytoplas-mic, redistributes to the nucleus upon receptoractivation (Liu et al., 1997; Zhang et al., 1997), andbinds to a short specific sequence of DNA termedthe Smad-binding element (SBE; Zawel et al.,1998). These functions probably involve the homo-oligomerization of Dpc4 as well as formation offunctional heteromeric complexes with other Smadproteins and with more general transcription factors(Liu et al., 1996; Zhang et al., 1996; Shi et al., 1997).The tumorigenic mutations fall in three majorcategories. The missense mutations of the N-terminal third of the protein abrogate its DNA-binding function, the truncating mutations of theC-terminus remove its ability to activate geneexpression while preserving its DNA-binding prop-erty, and the missense mutations of the C-terminalregion appear to interfere with its nuclear localiza-tion in response to ligand-initiated signaling (Dai etal., 1998). The ability of Dpc4 to act as a transcrip-tional activator at SBE sites is uniformly lost in allmutations found in tumors, strongly suggesting thatit is this property which provides its tumor suppres-sive role (Dai et al., 1998). When Dpc4 is artificiallydirected to the nucleus, it arrests cells in G1 phaseof the cell cycle and induces apoptosis (Dai et al.,1999).

Dpc4 is suspected to act in the TGFb suppres-sive pathway; in some cells Dpc4 has been conclu-sively proven to be required for important TGFb

effects (Zhou et al., 1998). Nonetheless, the geneticinactivation of both DPC4 and a TGFb receptorgene has been observed to coexist in the sametumors (Goggins et al., 1998b). This suggests thatDPC4 has functions that are sufficiently distinctfrom TGFb to require its separate inactivation—one should not simplistically consider them torepresent the same pathway. If the inactivation ofthe DPC4 pathway were indeed to be universallyrequired in pancreatic carcinogenesis, we do not yetknow enough of the upstream and downstreamcomponents to identify the most promising candi-date genes for mutational searches.

4 HILGERS AND KERN

Page 5: Molecular genetic basis of pancreatic adenocarcinoma

Low-Frequency Changes

Oncogenes

Gene amplification is seen at multiple sites.Chromosomal band 19q13 is the most commontarget, being amplified in 10%–20% of the cancers.This hotspot of amplification was first found in ascreen for overexpressed genes (Batra et al., 1991a,1991b). Many years later it was confirmed by acandidate gene approach and separately also by anunbiased genome landmark scanning method(Cheng et al., 1996; Miwa et al., 1996).

When speaking of gene amplification, whichaffects large genomic regions, it is often difficult tospecify with certainty the target gene, and indeedmultiple genes may together provide the survivaladvantage that allows selection for clones that carrythe amplification. A number of genes are known tobe amplified at 19q13, but the signal transducerAKT2 is the leading candidate target gene in thisregion (Cheng et al., 1996).

Amplifications of the MYB gene has also beenobserved in as many as 10% of cases in systematic(positional mapping) studies (Wallrapp et al., 1997).Reports also implicate the amplification of othergenes and chromosomal regions (Yamada et al.,1986; Solinas-Toldo et al., 1996; Ghadimi et al.,1999).

Tumor suppressor and DNA repair genes

There is a considerable and growing number ofgenes known to be inactivated at frequencies of4%–10% of tumors. Some of these clearly fall underthe high-frequency category considered above, onthe basis of pathway memberships. The remainderare in some ways even more exciting, because theyoften represent novel and unexplored regulatorysystems, and also simply because of the sheernumber of such mutant genes. They illustratevividly that pancreatic cancer is not a four-chordsong, but a symphony of large and small parts.

There is a technical point that needs to be made,one of extreme importance. These low-frequencygenetic targets are not random. Genes chosen atrandom have a somatic mutation rate expected atless than 1% of tumors examined (with appropriateexceptions made for tumors having defects in DNAsynthetic fidelity). There are also simple statisticalmeans to discriminate between random mutationsand those patterns that imply the action of selectivepressures during tumorigenesis. Statistical ap-proaches are feasible because random mutationsindeed do occur in dividing cell populations, andthese random mutations have a known pattern. For

example, one could imagine a gene whose muta-tional spectrum included many silent changes (suchas third-base nucleotide substitutions that do notchange the amino acid or other substitutions thatproduce a conservative amino acid change). Onewould be justified to assume that differing degreesof selective pressures had not acted on the sub-clones that sustained these initial random muta-tions. On the other hand, if a mutational spectrumnearly exclusively comprised a skewed spectrum ofnonsense mutations and other inactivating muta-tions, the conclusion becomes the opposite. Itwould then be clear that, of the initial randommutations that are expected to occur in any growingcell population, only those that removed the geneactivity had been able to provide the cells with aselective growth advantage. In other words, a spec-trum statistically enriched in inactivating mutationsclearly indicates a tumor suppressor gene. As in allstatistics, it is a matter of N, the number of samplesstudied. Lower mutational frequencies require ahigher N, but in theory present no special difficul-ties.

Intragenic mutations and homozygous deletionsof the MKK4 (MAP kinase kinase 4) gene are seenin about 4% of pancreatic cancers (Teng et al., 1997;Su et al., 1998). MKK4 is a component of a stress-activated protein kinase cascade and has roles inapoptosis and growth control (Derijard et al., 1995;Lin et al., 1995). MKK4 was localized to a homozy-gous deletion site that was first identified in apancreatic cancer, but is inactivated in other cancertypes as well (Teng et al., 1997). The geneticinactivation of MKK4 can be found to coexist withmutations of p16, p53, and DPC4, and so is thoughtto serve a distinct pathway of tumor suppression(Su et al., 1998).

A somatic homozygous deletion was identified ina pancreatic cancer by an unbiased genomic dele-tion cloning method, the representational differ-ence analysis (Schutte et al., 1995a, 1995b). Thisencompassed the BRCA2 gene, and the map of thisdeletion (markers that included coding sequencesof the BRCA2 gene) was instrumental in the posi-tional cloning of the gene (Wooster et al., 1994).Somatic mutations, however, appear rare. About 5%of unselected pancreatic cancers, and about 10% ofthose from Ashkenazi Jewish patients, have germ-line mutations (Goggins et al., 1996; Ozcelik et al.,1997). This is remarkable in that few of thesepatients had a family history of breast cancer andnone had a relative with pancreatic cancer. Thus,among pancreatic cancers that appear clinically to

5MOLECULAR GENETIC BASIS

Page 6: Molecular genetic basis of pancreatic adenocarcinoma

be sporadic, one can observe an inherited basis. Ifinherited mutations can indeed give rise to ‘‘spo-radic’’ cancer, one must wonder to what extent thecontribution of heritable cancer susceptibility tendsto be overlooked. This brings up the speculativebut undeniable possibility that inherited mutationsof low penetrance might account for much or evenmost of the cancers in the general population.Whatever the wider epidemiologic and theoreticimplications, the precise role of BRCA2 in pancre-atic and other cancers remains largely unknown.The gene is not yet firmly established in either agrowth regulatory or a DNA repair function inhuman tumors, although both are suggested fromexperimental systems.

The Peutz-Jeghers syndrome (PJS) is associatedwith an increased risk of cancer, including pancre-atic (Giardiello et al., 1987). Inherited inactivatingmutations of the LKB1/STK11 gene cause thesyndrome (Hemminki et al., 1998; Jenne et al.,1998). In a pancreatic cancer of a patient havingPJS, loss of the remaining wild-type allele wasobserved, suggestive that the LKB1/STK11 geneacts as a typical tumor suppressor gene in neo-plasms (remember that Peutz-Jeghers polyps arenot neoplastic, but classified as hamartomatous; Suet al., 1999). In pancreatic cancers that are notassociated with the syndrome, somatic mutationsaccompanied by of the LKB1/STK11 gene are seenin about 5% (Su et al., 1999). The gene encodes aserine/threonine kinase, for which the componentsof the signaling pathway and its cellular role arecurrently undefined (Hemminki et al., 1998; Jenneet al., 1998).

The replication error (RER1) phenotype, signify-ing a defect in DNA mismatch repair function, isfound in about 3% of pancreatic cancers (Goggins etal., 1998a). This is an extremely distinctive subsetof pancreatic cancer. Very few tumors have beendescribed, but they are characterized by a sheet-like growth pattern with pushing borders, cells thatare almost syncytial without differentiation, and awild-type K-ras gene. Long-term survivors havebeen observed in this subgroup. In the one tumorcharacterized in detail, p16, p53, and DPC4 genealterations were not found. However, as with theRER1 cancers of the colorectum, alterations ofsimple repeated sequences, including those of thepolyA tract (a mononucleotide run) within thecoding sequence of the TGFBR2 (TGFb type IIreceptor) gene, are widespread (Goggins et al.,1998b). The precise genetic lesions that cause themismatch repair defect in pancreatic cancer are notyet described. Pancreatic cancers are reported in

families with inherited mutations in the knowngenes of this repair system, and some patients havepancreatic cancer combined with multiple colorec-tal cancers, thus showing a connection in somecases with hereditary nonpolyposis colorectal can-cer (Lynch et al., 1985). Some of the repair defectstherefore are undoubtedly on an inherited basis,while others are of somatic origin.

Except for in the RER1 cancers where biallelicinactivation of the TGFBR2 gene is seen, alter-ations in the TGFb receptors are quite rare (Gog-gins et al., 1998b). A single homozygous deletion ofthe TGFBR2 gene is known in a pancreatic cancer.A homozygous deletion of the ALK5 (TGFb type Ireceptor) gene in one pancreatic and one biliarycancer constitutes the only known somatic geneticinactivation of this gene in human cancer. Intra-genic mutations of ALK5 are not identified, and onecould indeed raise questions that challenge theevidence to support ALK5 as a bona-fide tumorsuppressor gene. Homozygous deletions and muta-tions of other related (TGFb superfamily-binding,serine/threonine kinase) receptors, however, havenot been found in pancreatic cancer. Also, defectsin TGFb receptor expression and function arecommon in many human cancers including pancre-atic (Baldwin et al., 1996), and the TGFb receptorsare widely considered to be tumor suppressors.

Abnormalities of one of the chromosome fragilesites (FRA3B) are seen in pancreatic cancers(Shridhar et al., 1996; Simon et al., 1998). Thisregion of chromosomal band 3p14.2 is also the siteof the diadenosine hydrolase gene FHIT, which inmultiple tumors types is affected by occasion homo-zygous deletions (Kastury et al., 1996). Such dele-tions of the FHIT gene were found in about 6% ofpancreatic cancers. In tumors that are competent inmismatch DNA repair (i.e., not RER1), the rate isabout 4%. Intriguingly, two of three RER1 tumorshad such deletions (P , 0.01), suggesting a selec-tive pressure that favors the evolution of clones thathave lost the FHIT gene or that contain a tendencyto sustain breakage in the FRA3B region. Intra-genic mutations of FHIT in pancreatic cancer are sorare as to be considered of no consequence. Thereis one report that found a high rate of FHIThomozygous deletion in a series of pancreatic can-cer (Simon et al., 1998), but it is unconfirmed inother series.

It is important to note that most candidate genesthat are studied are found not to have mutations.Genes known to lack mutations include the tumorsuppressors and related genes: PTEN1 (Kong et al.,1997; Okami et al., 1998), SCH (Hahn et al., 1995),

6 HILGERS AND KERN

Page 7: Molecular genetic basis of pancreatic adenocarcinoma

SMAD genes 1, 2, 3, 5, and 6 (Riggins et al., 1997a,1997b), APC (McKie et al., 1993; Smith et al., 1993;Seymour et al., 1994), b-catenin (Goggins et al.,1997a), ALK1 (Goggins et al., 1998b), p18INK4c

(Rozenblum et al., 1996), p19INK4d (Rozenblum etal., 1996), (in tumors lacking homozygous deletionof the neighboring p16 gene) p15INK4b (Rozenblumet al., 1996), and (in tumors having an intact copy ofthe overlapping p16 gene but without mutationsthat inactivate the p16 reading frame) p14ARF (Cal-das et al., 1994a; Rozenblum et al., 1997).

FAMILIAL PANCREATIC CANCER

It is estimated that as many as 10% of pancreaticcancer may occur in a familial form. Unlike in otherfamilial cancer syndromes, pancreatic cancer doesnot often occur in association with a recognizableclinical syndrome. Kindreds of affected individualsare small. The age of onset in such families isunchanged from that of sporadic pancreatic cancer,about 63 years (Lynch et al., 1990). The recognitionthat familial pancreatic cancer indeed had an inher-ited basis was therefore first proved not by the useof LOD score analysis or other positional ap-proaches, but by the mutational analysis of candi-date tumor suppressor genes. Three such genes(the p16, BRCA2, and LKB1/STK11 genes), as wellas inherited syndromes associated with deficientDNA mismatch repair, are known to be directlyassociated with susceptibility to pancreatic cancer.Inherited mutations of the p16 gene are seen inpatients with pancreatic cancer in the setting of theFAMMM (familial atypical multiple mole and mela-noma) syndrome (Lynch and Fusaro 1991; Gruis etal., 1995; Whelan et al., 1995); but p16-mutantpedigrees having pancreatic cancer alone have notbeen reported. In breast cancer families and in apopulation study of Iceland, breast cancers due toinherited mutations of BRCA2 accompany an in-creased risk of pancreatic cancer (Thorlacius et al.,1996). Unlike with the p16 gene, inherited muta-tions of BRCA2 have such a low penetrance that afamily history may not be apparent. Inheritedoncogenes are not known to predispose to pancre-atic ductal cancer.

There needs to be made special mention of aspecial type of cancer susceptibility gene. Onecould observe that the above list of inheritedmutations is restricted to genes that cause selectiveadvantages for individual cell clones; that is, theyare cell-autonomous. But cells do not live byinternal signals alone. Environmental influences oncancer rates are well known, such as when there isexposure to exogenous carcinogens or mitogenic

agents. This view can be extended to influencesarising within the body, acting on not only indi-vidual cells but entire tissues. In imprecise terms,this can be called a form of field effect. But for moreprecision, we would need an example. The cationictrypsinogen gene, PRSS1, provides it. Inheritedmutations in this gene underlie familial pancreatitis(Whitcomb et al., 1996), wherein there is an inappro-priate activation of trypsinogen to produce theprototype hydrolytic digestive enzyme, trypsin.

Inherited PRSS1 mutations underlie the form ofinherited pancreatic cancer susceptibility havingthe greatest penetrance. The relative risk for thedevelopment of pancreatic cancer in a carrier of aPRSS1 mutation is about 50-fold, the overall life-time risk is nearly 40%, and the average age of onsetis dramatically reduced to 39 years (Lowenfels etal., 1997). This tumor susceptibility is presumablydue to mitogenic stimulation and clonal outgrowthof pancreatic ductal cells as part of the normalhealing responses that occur subsequent to re-peated rounds of tissue destruction. If we were todesignate tumor suppressors as genes whose pres-ence in the germline prevents the formation oftumors (a definition sometimes referred to as thegold standard), then trypsinogen would be in nu-merical terms one of the most convincing examplesof a tumor suppressor gene yet identified.

Obviously, a number of interesting and conten-tious points are hereby raised. The first is thatinherited cancer susceptibility indeed does involvemore than the classic trio of tumor suppressorgenes, repair genes, and oncogenes. The second isthat the study of this particular class of tumorprevention is underdeveloped as a scientific field,but could have major implications for our under-standing of how human populations develop neopla-sia. The arguments that could be offered are quiteopen-ended, and anticipate a fascinating future ingenetic epidemiology. For example, what if a statis-tically higher mean body temperature were to becorrelated to the incidence of cancers of internalorgans? Would the gene(s) that control body tem-perature now be considered oncogenes and suppres-sor genes? The cationic trypsinogen gene is merelythe first clear example of this other form of tumorsuppression. If some genes can be classified asgatekeepers (Kern, 1993; Kinzler and Vogelstein,1997), DNA repair genes become caretaker genes(Kinzler and Vogelstein, 1997), and tumor suppres-sors that act late in tumorigenesis could be termedguardsmen genes (indicating a role after breachingof the neoplasm’s initial regulatory gate), then thecategory of genes represented by PRSSS1 needs a

7MOLECULAR GENETIC BASIS

Page 8: Molecular genetic basis of pancreatic adenocarcinoma

designation as well. Since the activity is specificallyto reduce the destructive processes and inflamma-tion in neighboring or distant tissues, they could betermed diplomat genes.

In the absence of a known gatekeeper function inpancreatic ductal epithelium that controls the pro-pensity to initiate neoplasia, the inherited tumorsuppressor mutations in pancreatic cancer (p16,BRCA2, LKB1/STK11) fit the guardsmen designa-tion. Indeed, a role in later stages is suggested bystudies of these genes in pancreatic tumor progres-sion (Goggins et al., 1997b; Moskaluk et al., 1997;Wilentz et al., 1998). The penetrance of thesegenes is low, producing pancreatic cancer in per-haps 5%–10% of persons carrying the mutant genes.This corresponds to a relative risk of 10- to 20-foldover the incidence rate (less than 0.5% lifetimerisk) of the general population. To date, the knownelevations in incident rates of pancreatic carcinomain African Americans has no association with theknown somatic or germline genetic alterations ofthe cancers (Rozenblum et al., 1997).

It had been estimated that as many as 10% ofpancreatic cancer might occur in a familial form(Lynch, 1994). This estimate was based on theobservation of familial pancreatic cancer, definedwhen at least two first-degree relatives exhibit thedisease. But such an estimate would exclude inci-dents of susceptibility such as demonstrated by theBRCA2 gene mutations, wherein the patient almostinvariably has no familial history of pancreaticcancer. Consequently, the proportion of pancreaticcancer that is inherited is presumably even higherthan that of the above estimate.

Thus, even when we restrict consideration to thepatients that have known germline mutations, someremarkable facts exist for pancreatic cancer. For noother common adult cancer are there as manyknown pathways whose defects give rise to inher-ited susceptibility, nor is there any other organ forwhich the fraction of cancers having an inheritedbasis is higher than for the pancreas.

NO NEWS, JUST SPECULATIVE VIEWS

It is entirely possible that all the major tumorsuppressor genes have already been found. In thisview, for pancreatic and other cancer types, nogenes that have high frequencies of mutation (above50% of cases) await discovery. The future of cancergenetics would then rely in large part in theuncovering of the low-frequency mutation targets.We will piece together entire mutant pathways, butwill do so piecemeal and perhaps at a snail’s pace,5% here and 3% there. Even so, the wonderful

variety and novelty of these new pathways willserve as the fountainhead for individualized riskassessment and for rational drug development.

As this science progresses, low-penetrance muta-tions carried in the germline will be realized toaccount for ever increasing proportions of the adult-onset cancers. This is likely to be an extremelyproductive search, in that evolution has had littleneed to eliminate such mutations from the genepool. Evolution having dropped the ball, the insur-ance companies will take up the offensive task ofgenetic exclusion, to the response of a compensa-tory legislative barrage. Because translational re-search involves real people, researchers will beaffected by these battles.

Positional cloning will become trivialized for twomajor reasons. First, the gene map soon will becompleted. Second, the low-frequency and low-penetrance genetic lesions, those left to be discov-ered, do not readily lend themselves to such posi-tional approaches. While it is true that homozygousdeletions can pinpoint a genetic target where LODscores will fail, not all suppressor and repair genesare affected by these deletions, and not all suchdeletions can reasonably implicate the existence ofan important gene. The use of homozygous dele-tions in a positional strategy is historically vindi-cated as productive, but will benefit greatly fromsome anticipated technical advances.

The consensus mapping of LOH (loss of hetero-zygosity) will prove to be largely disappointing. Ithas perhaps been underappreciated how little hasbeen accomplished through this technique to date.If we restrict ourselves to genes that are somaticallymutated (since germline mutations benefit fromLOD scores and other practical advantages), itmight be interesting to calculate the numerical ratioof LOH-mapping papers to convincing tumor sup-pressor genes cloned as the direct consequence ofthese maps. Our interest is frustrated by the impos-sibility of dividing by zero.

If so, then, how can one use LOH data? Itappears that it is the frequency of deletion at alocus, rather than the map of individual deletionsthat is most helpful. This distinction exists for atleast two major reasons. First, because only raretumor suppressor genes have a pattern of near-universal somatic inactivation when LOH is pres-ent. (A sidelight: The p53 gene is one such ex-ample, but was cloned long before LOH mappingbecame available; nevertheless, one cannot easilyexpect another such universal target. In anotherexample, the DPC4 gene is inactivated in onlyabout half of the pancreatic cancers having LOH of

8 HILGERS AND KERN

Page 9: Molecular genetic basis of pancreatic adenocarcinoma

chromosome 18, but did not emerge from theattempted mapping of consensus LOH.) Second,the existence of a tumor suppressor gene does notimmunize the chromosome against additional ge-netic lesions, whose analysis would assemble astreet map of blind alleys.

For the low-frequency genetic targets, the muta-tion of the known gene constitutes only a minorityof the overall LOH frequency. Indeed, at the lowfrequencies often used in LOH consensus map-ping, the confounding occurrence of random (unse-lected) deletions will not simply overlap, but willcompletely obscure the real gene targets. The highpromise offered by LOH mapping perhaps remainsin special cases only, when a frequency of LOH isfar above background rates (as judged by studies ofother chromosomes) and when a consensus seemsto emerge from a vast majority of the deletionsmapped (in high-resolution studies of a particularchromosome). Chromosomal arm 1p may meetthese criteria in pancreatic cancer (Hilgers et al.,1999). Soberingly, to date the use of hundreds ofgenomic markers has not suggested another tempt-ing locale for such a mapping project in this disease.

Increasingly then, we will see the emergence ofnew low-frequency genetic targets through theapplication of brute-force technologies. Batch muta-tional screening of the hundreds of rational candi-date genes will follow, pursuing the putative candi-date members of signaling pathways, especiallythose pathways already known to have low-frequency mutations in a member gene. As thetranscript map of the genome becomes known, thelists of the gene content of the homozygousdeletions will become triaged as candidate genes.The requirement for technical advances will beacute in the study of inherited susceptibility, wheresmall family sizes and absence of a recognizableclinical syndrome renders the positional approachesineffective. Eventually, tumor studies will imple-ment the automated screening of tens of thousandsof AORGs (any old random genes). This mutationaldatabase will identify a busy grid of genes andnovel pathways, representing both the blind alleysand the true avenues of genetic research. Only theapplication of descriptive genetics, epidemiology,experimentation, and skeptical intellectual argu-ment will appropriately direct the research traffictoward the one as opposed to the other.

As ever, the initial glare and inelegance of thetechnology will fade into the background as thechallenges of progressive discovery remain thesame. Our implicit pact with the taxpayers thatfund this research provides an obligation not for

fascinating and engaging science, but for the actualidentification of clinical breakthroughs. Notwith-standing the gratification in reviewing the accumu-lated progress, by this measure, there is consider-ably more left to do.

REFERENCES

Almoguerra C, Shibata D, Forrester K, Martin J, Arnheim N,Perucho M. 1988. Most human carcinomas of the exocrinepancreas contain mutant c-K-ras genes. Cell 53:549–554.

Baldwin RL, Friess H, Yokoyama M, Lopez ME, Kobrin MS,Buchler MW, Korc M. 1996. Attenuated ALK5 receptor expres-sion in human pancreatic cancer: correlation with resistance togrowth inhibition. Int J Cancer 67:283–288.

Bartkova J, Lukas J, Guldberg P, Alsner J, Kirkin AF, Zeuthen J,Bartek J. 1996. The p16-cyclin D/Cdk4-pRb pathway as a func-tional unit frequently altered in melanoma pathogenesis. CancerRes 56:5475–5483.

Batra SK, Metzgar RS, Hollingsworth MA. 1991a. Isolation andcharacterization of a complementary DNA (PD-1) differentiallyexpressed by human pancreatic ductal cell tumors. Cell GrowthDifferent 2:385–390.

Batra SK, Metzgar RS, Hollingsworth MA. 1991b. Molecular cloningand sequence analysis of the human ribosomal protein S16. J BiolChem 266:6830–6833.

Brat DJ, Lillemoe KD, Yeo CJ, Warfield PB, Hruban RH. 1998.Progression of pancreatic intraductal lesions (high grade PanIN) toinfiltrating adenocarcinoma of the pancreas. Am J Surg Pathol22:163–169.

Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, SeymourAB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE. 1994a.Frequent somatic mutations and homozygous deletions of the p16(MTS1) gene in pancreatic adenocarcinoma. Nat Gen 8:27–31.

Caldas C, Hahn SA, Hruban RH, Redston MS, Yeo CJ, Kern SE.1994b. Detection of K-ras mutations in the stool of patients withpancreatic adenocarcinoma and pancreatic ductal hyperplasia.Cancer Res 54:3568–3573.

Chen CY, Oliner JD, Zhan Q, Fornace AJ Jr, Vogelstein B, KastanMB. 1994. Interactions between p53 and MDM2 in a mammaliancell cycle checkpoint pathway. Proc Natl Acad Sci USA 91:2684–2688.

Cheng JQ, Ruggeri B, Klein WM, Sonoda G, Altomare DA, WatsonDK, Testa JR. 1996. Amplification of AKT2 in human pancreaticcancer cells and inhibition of AKT2 expression and tumorigenec-ity by antisense RNA. Proc Natl Acad Sci USA 93:3636–3641.

Ciotti P, Strigini P, Bianchi-Scarra G. 1996. Familial melanoma andpancreatic cancer. N Engl J Med 334:469–470.

Cubilla AL, Fitzgerald PJ. 1976. Morphological lesions associatedwith human primary invasive nonendocrine pancreas cancer.Cancer Res 36:2690–2698.

Dai JL, Turnacioglu K, Schutte M, Sugar AY, Kern SE. 1998. Dpc4transcriptional activation and dysfunction in cancer cells. CancerRes 58:4592–4597.

Dai JL, Bansal RK, Kern SE. 1999. G1 cell cycle arrest and apoptosisinduction by nuclear Smad4/Dpc4: phenotypes reversed by atumorigenic mutation. Proc Natl Acad Sci USA 96:1427–1432.

Derijard B, Raingeaud J, Barret T, Wu I, Han J, Ulevitch RJ, DavisRJ. 1995. Independent human MAP kinase signal transductionpathways defined by MEK and MKK isoforms. Science 267:682–685.

El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, TrentJM, Lin D, Mercer WE, Kinzler KW, Vogelstein B. 1993. WAF1, apotential mediator of p53 tumor suppression. Cell 75:817–825.

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

Furukawa T, Chiba R, Kobari M, Matsuno S, Nagura H, TakahashiT. 1994. Varying grades of epithelial atypia in the pancreatic ductsof humans: classification based on morphometry and multilvariateanalysis and correlated with positive reactions of carcinoembryonicantigen. Arch Pathol Lab Med 118:227–234.

Ghadimi, B M, Schrock E, Walker RL, Wangsa D, Jauho A, MeltzerPS, Ried T. 1999. Specific chromosomal aberrations and amplifica-tion of the AIB1 nuclear receptor coactivator gene in pancreaticcarcinomas. Am J Pathol 154:525–536.

Giardiello FM, Welsh SB, Hamilton SR, Offerhaus GJA, GittelsohnAM, Booker SV, Krush AJ, Yardley JH, Luk GD. 1987. Increased

9MOLECULAR GENETIC BASIS

Page 10: Molecular genetic basis of pancreatic adenocarcinoma

risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med316:1511–1514.

Giardiello FM, Hamilton SR, Kern SE, Offerhaus GJ, Green PA,Celano P, Krush AJ, Booker SV. 1991. Colorectal neoplasia injuvenile polyposis or juvenile polyps. Arch Dis Child 66:971–975.

Goggins M, Schutte M, Lu J, Moskaluk CA, Weinstein CL, PetersenGM, Yeo CJ, Jackson CE, Lynch HT, Hruban RH, Kern SE. 1996.Germline BRCA2 gene mutations in patients with apparentlysporadic pancreatic carcinomas. Cancer Res 56:5360–5364.

Goggins M, Hilgers W, Hruban RH, Kern SE. 1997a. Normalb-catenin gene in pancreatic cancer. NOGO 1:4.

Goggins M, Hruban RH, Kern SE. 1997b. Relationship of BRCA2germline mutation and pancreatic intraductal lesions. NIH SPOREMeeting 5:30.

Goggins M, Offerhaus GJA, Hilgers W, Griffin CA, Shekher M, TangD, Sohn T, Yeo CJ, Kern SE, Hruban RH. 1998a. Adenocarcino-mas of the pancreas with DNA replication errors (RER1) areassociated with wild-type K-ras and characteristic histopathology:poor differentiation, a syncytial growth pattern, and pushingborders suggest RER1. Am J Pathol 152:1501–1507.

Goggins M, Shekher M, Turnacioglu K, Yeo CJ, Hruban RH, KernSE. 1998b. Genetic alterations of the TGFb receptor genes inpancreatic and biliary adenocarcinomas. Cancer Res 58:5329–5332.

Gruis NA, Sandkuiji LA, van der Velden PA, Bergman W, Frants RR.1995. CDKN2 explains part of the clinical phenotype in Dutchfamilial atypical multiple-mole melanoma (FAMMM) syndromefamilies. Melanoma Res 9:169–177.

Hahn SA, Seymour AB, Hoque ATMS, Schutte M, da Costa LT,Redston MS, Caldas C, Weinstein CL, Fischer A, Yeo CJ, HrubanRH, Kern SE. 1995. Allelotype of pancreatic adenocarcinomausing a xenograft model. Cancer Res 55:4670–4675.

Hahn SA, Hoque ATMS, Moskaluk CA, daCosta LT, Schutte M,Rozenblum E, Seymour AB, Weinstein CL, Yeo CJ, Hruban RH,Kern SE. 1996a. Homozygous deletion map at 18q21.1 in pancre-atic cancer. Cancer Res 56:490–494.

Hahn SA, Schutte M, Hoque ATMS, Moskaluk CA, da Costa LT,Rozenblum E, Weinstein CL, Fischer A, Yeo CJ, Hruban RH,Kern SE. 1996b. DPC4, a candidate tumor-suppressor gene at18q21.1. Science 271:350–353.

Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S,Loukola A, Bignell G, Warren W, Aminoff M, Hoglund P, JarvinenH. 1998. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature 391:184–187.

Hilgers W, Tang DJ, Sugar AY, Shekher MC, Hruban RH, Kern SE.1999. High-resolution deletion mapping of chromosome 1p inpancreatic cancer identifies a major consensus at 1p35. GenesChromosomes Can 24:351–355.

Howe JR, Roth S, Ringold JC, Summers RW, Jarvinen HJ, SistonenP, Tomlinson IP, Houlston RS, Bevan S, Mitros FA, Stone EM,Aaltonen LA. 1998. Mutations in the SMAD4/DPC4 gene injuvenile polyposis. Science 280:1086–1088.

Hruban RH, Yeo CJ, Kern SE. 1998. Pancreatic cancer. In VogelsteinB, Kinzler KW, editors. Metabolic and molecular basis of inheriteddiseases (MMBID): the genetic basis of human cancer. New York:McGraw-Hill, p 603–614.

James GL, Goldstein JL, Brown MS. 1995. Polylysine and CVIMsequences of K-RasB dictate specificity of prenylation and conferresistance to benzodiazepine peptidomimetic in vitro. J Biol Chem270:6221–6226.

Jenne DE, Reimann H, Nezu J, Friedel W, Loff S, Jeschke R, MullerO, Back W, Zimmer M. 1998. Peutz-Jeghers syndrome is causedby mutations in a novel serine threonine kinase. Nat Genet18:38–43.

Kastury K, Baffa R, Druck T, Ohta M, Cotticelli MG, Inoue H,Negrini M, Rugge M, Huang D, Croce CM, Palazzo J, Huebner K.1996. Potential gastrointestinal tumor suppressor locus at the3p14.2 FRA3B site identified by homozygous deletions in tumorcell lines. Cancer Res 56:978–983.

Kern SE. 1993. Clonality: more than just a tumor-progression model.J Natl Cancer Inst 85:1020–1021.

Kern SE, Pietenpol JA, Thiagalingam S, Seymour A, Kinzler KW,Vogelstein B. 1992. Oncogenic forms of p53 inhibit p53-regulatedgene expression. Science 256:827–830.

Kinoshita I, Dosaka-Akita H, Mishina T, Akie K, Nishi M, HiroumiH, Hommura F, Kawakami Y. 1996. Altered p16INK4 and retino-blastoma protein status in non-small cell lung cancer: potentialsynergistic effect with altered p53 protein on proliferative activity.Cancer Res 56:5557–5562.

Kinzler KW, Vogelstein B. 1997. Cancer-susceptibility genes: gate-keepers and caretakers. Nature 386:761–763.

Kong D, Suzuki A, Zou T-T, Sakurada A, Kemp LW, Wakatsuki S.1997. PTEN1 is frequently mutated in primary endometrialcarcinomas. Nat Genet 17:143–144.

Kozuka S, Sassa R, Taki T, Masamoto K, Nagasawa S, Saga S,Hasegawa K, Takeuchi M. 1979. Relation of pancreatic ducthyperplasia to carcinoma. Cancer 43:1418–1428.

Kratzke RA, Greatens TM, Rubins JB, Maddaus MA, NiewoehnerDE, Niehans GA, Geradts J. 1996. Rb and p16INK4a expressionin resected non-small cell lung tumors. Cancer Res 56:3415–3420.

Leach FS, Tokino T, Meltzer P, Burrell M, Oliner JD, Smith S, HillDE, Sidransky D, Kinzler KW, Vogelstein B. 1993. p53 mutationand Mdm2 amplification in human soft tissue sarcomas. CancerRes 53:2231–2234.

Lerner EC, Qian Y, Blaskovich MA, Fossum RD, Vogt A, Sun J, CoxAD, Der CJ, Hamilton AD, Sebti SM. 1995. Ras CAAX peptidomi-metic FTI-277 selectively blocks oncogenic Ras signaling byinducing cytoplasmic accumulation of inactive Ras-Raf com-plexes. J Biol Chem 270:26802–26806.

Lin A, Minden A, Martinetto H, Claret F, Lange-Carter C, MercurioF, Johnson GL, Karin M. 1995. Identification of a dual specificitykinase that activates the Jun kinases and p38-Mpk2. Science268:286–290.

Liu F, Hata A, Baker JC, Doody J, Carcamo J, Harland RM,Massague J. 1996. A human Mad protein acting as a BMP-regulated transcriptional activator. Nature 381:620–623.

Liu F, Pouponnot C, Massague J. 1997. Dual role of the Smad4/DPC4 tumor suppressor in TGF-b–inducible transcriptional com-plexes. Genes Dev 11:3157–3167.

Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK,Jr., Perrault J, Whitcomb DC. 1997. Hereditary pancreatitis andthe risk of pancreatic cancer: International Hereditary PancreatitisStudy Group. J Natl Cancer Inst 89:442–446.

Lu S-L, Kawabata M, Imamura T, Akiyama Y, Nomizu T, MiyazonoK, Yuasa Y. 1998. HNPCC associated with germline mutation inthe TGFb type II receptor. Nat Genet 19:17–18.

Lynch HT. 1994. Genetics and pancreatic cancer. Arch Surg 129:266–268.

Lynch HT, Voorhees GJ, Lanspa SJ, McGreevy PS, Lynch JF. 1985.Pancreatic carcinoma and hereditary nonpolyposis colorectal can-cer: a family study. Br J Cancer 52:271–273.

Lynch HT, Fitzsimmons ML, Smyrk TC, Lanspa SJ, Watson P,McClellan J, Lynch JF. 1990. Familial pancreatic cancer: clinico-pathologic study of 18 nuclear families. Amer J Gastroenterol85:54–60.

Lynch HT, Fusaro RM. 1991. Pancreatic cancer and the familialatypical multiple mole melanoma (FAMMM) syndrome. Pancreas6:127–131.

McKie AB, Filipe MI, Lemoine NR. 1993. Abnormalities affectingthe APC and MCC tumour suppressor gene loci on chromosome5q occur frequently in gastric cancer but not in pancreatic cancer.Int J Cancer 55:598–603.

Miwa W, Yasuda J, Murakami Y, Yashima K, Sugano K, Sekine T,Kono A, Egawa S, Yamaguchi K, Hayashizaki Y, Sekiya T. 1996.Isolation of DNA sequences amplified at chromosome 19q13.1–q13.2 including the AKT2 locus in human pancreatic cancer.Biochem Biophys Res Commun 225:968–974.

Momand J, Zambetti GP, Olson DC, George D, Levine AJ. 1992.The mdm-2 oncogene product forms a complex with the p53protein and inhibits p53-mediated transactivation. Cell 69:1237–1245.

Moskaluk CA, Hruban RH, Kern SE. 1997. p16 and K-ras mutationsin the intraductal precursors of human pancreatic adenocarcinoma.Cancer Res 57:2140–2143.

Okami K, Wu L, Riggins G, Cairns P, Goggins M, Evron E, HalachmiN, Ahrendt SA, Reed AL, Hilgers W, Kern SE, Sidransky D, Jen J.1998. Analysis of PTEN/MMAC1 alterations in aerodigestive tracttumors. Cancer Res 58:509–511.

Oliner JD, Kinzler KW, Meltzer PS, George DL, Vogelstein B. 1992.Amplification of a gene encoding a p53-associated protein inhuman sarcomas. Nature 358:80–83.

Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW,Vogelstein B. 1993. Oncoprotein MDM2 conceals the activationdomain of tumour suppressor p53. Nature 362:857–860.

Ozcelik H, Schmocker B, Di Nicola N, Shi XH, Langer B, Moore M,Taylor BR, Narod SA, Darlington G, Andrulis IL, Gallinger S,Redston M. 1997. Germline BRCA2 6174delT mutations inAshkenazi Jewish pancreatic cancer patients. Nat Genet 16:17–18.

10 HILGERS AND KERN

Page 11: Molecular genetic basis of pancreatic adenocarcinoma

Pellegata S, Sessa F, Renault B, Bonato M, Leone BE, Solcia E,Ranzani GN. 1994. K-ras and p53 gene mutations in pancreaticcancer: ductal and nonductal tumors progress through differentgenetic lesions. Cancer Res 54:1556–1560.

Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. 1997. A modelfor p53-induced apoptosis. Nature 389:300–305.

Pour PM, Sayed S, Sayed G. 1982. Hyperplastic, preneoplastic andneoplastic lesions found in 83 human pancreases. Am J Clin Pathol77:137.

Pretlow TP, Brasitus TA, Fulton NC, Cheyer C, Kaplan EL. 1993.K-ras mutations in putative preneoplastic lesions in human colon.J Natl Cancer Inst 85:2004–2007.

Redston MS, Caldas C, Seymour AB, Hruban RH, da Costa L, YeoCJ, Kern SE. 1994. p53 mutations in pancreatic carcinoma andevidence of common involvement of homocopolymer tracts inDNA microdeletions. Cancer Res 54:3025–3033.

Riggins GJ, Kinzler KW, Vogelstein B, Thiagalinbam S. 1997a.Frequency of SMAD gene mutations in human cancers. CancerRes 57:2578–2581.

Riggins GJ, Thiagalingam S, Goggins M, Kern SE, Kinzler KW,Vogelstein B. 1997b. Normal SMAD2 gene in pancreatic cancer.NOGO 1:40.

Rozenblum E, Schutte M, Kern SE. 1996. INK4 genes in pancreaticcarcinoma. Oncol Reports 3:743–745.

Rozenblum E, Schutte M, Goggins M, Hahn SA, Lu J, Panzer S,Zahurak M, Goodman SN, Hruban RH, Yeo CJ, Kern SE. 1997.Tumor-suppressive pathways in pancreatic carcinoma. Cancer Res57:1731–1734.

Ruggeri B, Zhang SY, Caamano J, DiRado M, Flynn SD, Klein-Szanto AJP. 1992. Human pancreatic carcinomas and cell linesreveal frequent and multiple alterations in the p53 and Rb-1tumor-suppressor genes. Oncogene 7:1503–1511.

Savage C, Das P, Finelli A L, Twonsend SR, Sun C-Y, Baird SE,Padgett RW. 1996. Caenorhabditis elegans genes sma-2, sma-3,and sma-4 define a conserved family of transforming growth factorbeta pathway components. Proc Natl Acad Sci USA 93:790–794.

Schutte M, da Costa LT, Hahn SA, Moskaluk C, Hoque ATMS,Rozenblum E, Weinstein CL, Bittner M, Meltzer PS, Trent JM,Yeo CJ, Hruban RH, Kern SE. 1995a. A homozygous deletionidentified by representational difference analysis in pancreaticcarcinoma overlaps the BRCA2 region. Proc Natl Acad Sci USA92:5950–5954.

Schutte M, Rozenblum E, Moskaluk CA, Guan X, Hoque ATMS,Hahn SA, da Costa LT, de Jong PJ, Kern SE. 1995b. An integratedhigh-resolution physical map of the DPC/BRCA2 region at chro-mosome 13q12–13. Cancer Res 55:4570–4574.

Schutte M, Hruban RH, Hedrick L, Molnar’Nadasdy G, WeinsteinCL, Bova GS, Isaacs WB, Cairns P, Nawro H, Sidransky D, CaseroR, Meltzer PS, Hahn SA, Kern SE. 1996. DPC4 gene in varioustumor types. Cancer Res 56:2527–2530.

Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabin-dran SK, Moskaluk CA, Hahn SA, Schwarte-Waldhoff I, Schmie-gel W, Baylin SB, Kern SE, Herman JG. 1997. Abrogation of theRb/p16 tumor-suppressive pathway in virtually all pancreaticcarcinomas. Cancer Res 57:3126–3130.

Sekelsky JJ, Newfeld SJ, Raftery LA, Chartoff EH, Gelbart WM.1995. Genetic characterization and cloning of mothers against dpp,a gene required for decapentaplegic function in Drosophilamelanogaster. Genetics 139:1347–1358.

Serrano M, Hannon GJ, Beach D. 1993. A new regulatory motif incell-cycle control causing specific inhibition of cyclin D/CDK4.Nature 366:704–707.

Seymour AB, Hruban RH, Redston M, Caldas C, Powell SM, KinzlerKW, Yee CJ, Kern SE. 1994. Allelotype of pancreatic adenocarci-noma. Cancer Res 54:2761–2764.

Shapiro GI, Edwards CD, Kobzik L, Godleski J, Richards W,Sugarbaker DJ, Rollins BJ. 1995. Reciprocal Rb inactivation andp16INK4 expression in primary lung cancers and cell lines. CancerRes 55:505–509.

Shi Y, Hata A, Lo RS, Massague J, Pavletich NP. 1997. A structuralbasis for mutational inactivation of the tumour suppressor Smad4.Nature 388:87–93.

Shridhar R, Shridhar V, Wang X, Paradee W, Dugan M, Sarkar F,Wilke C, Glover TW, Vaitkevicius VK, Smith DI. 1996. Frequentbreakpoints in the 3p14.2 fragile site, FRA3B, in pancreatictumors. Cancer Res 56:4347–4350.

Simon B, Bartsch D, Barth P, Prasnikar N, Munch K, Blum A, ArnoldR, Goke B. 1998. Frequent abnormalities of the putative tumor

suppressor gene FHIT at 3p14.2 in pancreatic carcinoma cell lines.Cancer Res 58:1583–1587.

Sirard C, de la Pompa JL, Elia AJ, Itie A, Mirtsos C, Cheung A, HahnS, Wakeham A, Schwartz L, Kern SE, Rossant J, Mak TW. 1998.The tumor suppressor gene Dpc4/Smad4 is required for gastrula-tion and later for anterior development of the mouse embryo.Genes Dev 12:107–119.

Smith KJ, Johnson KA, Bryan TM, Hill DE, Markowitz S, WillsonJK, Paraskeva C, Petersen GM, Hamilton SR, Vogelstein B, et al.1993. The APC gene product in normal and tumor cells. Proc NatlAcad Sci USA 90:2846–2850.

Solinas-Toldo S, Wallrapp C, Muller-Pillasch F, Bentz M, gress T,Lichter P. 1996. Mapping of chromosomal imbalances in pancre-atic carcinoma by comparative genomic hybridization. Cancer Res56:3803–3807.

Su GH, Hilgers W, Shekher M, Tang D, Yeo CJ, Hruban RH, KernSE. 1998. Alterations in pancreatic, biliary, and breast carcinomassupport MKK4 as a genetically targeted tumor-suppressor gene.Cancer Res 58:2339–2342.

Su GH, Hruban RH, Bova GS, Goggins M, Bansal RK, Tang DT,Shekher MC, Entius MM, Yeo CJ, Kern SE. 1999. Germline andsomatic mutations of the STK11/LKB1 Peutz-Jeghers gene inpancreatic and biliary cancers. Am J Pathol 154:1835–1840.

Tada M, Omata M, Kawai S, et al. 1993. Detection of ras genemutations in pancreatic juice and peripheral blood of patients withpancreatic adenocarcinoma. Cancer Res 53:2472–2474.

Tada M, Ohashi M, Shiratori Y, Okudaira T, Komatsu Y, Kawabe T,Yoshida H, Machinami R, Kishi K, Omata M. 1996. Analysis ofK-ras gene mutation in hyperplastic duct cells of the pancreaswithout pancreatic disease. Gastroenterol 110:227–231.

Takaku T, Oshima M, miyoshi H, Matsui M, Seldin MF, TaketoMM. 1998. Intestinal tumorigenesis in compound mutant mice ofboth Dpc4 (Smad4) and Apc genes. Cell 92:645–656.

Teng DH-F, Perry III WL, Hogan JK, Baumgard M, Bell R, Berry S,Davis T, Frank D, Frye C, Hattier T, Hu R, Jammulapati S,Janecki T, Leavitt A, Mitchell JT, Pero R, Sexton D, Schroeder M,Su PJ, Swedlund B, Kyriakis JM, Avruch J, Bertel P, Wong AK,Tavtigian SV. 1997. Human mitogen-activated protein kinasekinase 4 as a candidate tumor suppressor. Cancer Res 57:4177–4182.

Thorlacius S, Olafsdottir G, Tryggvadottir L, Neuhausen S, JonassonJG, Tavtigian SV, Tulinius H, Ogmundsdottir HM, Eyfjord JE.1996. A single BRCA2 mutation in male and female breastcarcinoma families from Iceland with varied cancer phenotypes.Nat Genet 13:117–119.

Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC,Leppert M, Nakamura Y, White R, Smits AMM, Bos JL. 1988.Genetic alterations during colorectal-tumor development. N EnglJ Med 319:525–532.

Wallrapp C, Muller-Pillasch F, Solinas-Toldo S, Lichter P, Friess H,Buchler M, Fink T, Adler G, Gress TM. 1997. Characterization ofa high copy number amplification at 6q24 in pancreatic canceridentifies c-myb as a candidate oncogene. Cancer Res 57:3135–3139.

Whelan AJ, Bartsch D, Goodfellow PJ. 1995. Brief report: a familialsyndrome of pancreatic cancer and melanoma with a mutation inthe CDKN2 tumor-suppressor gene. N Engl J Med 333:975–977.

Whitcomb DC, Gorry MC, Preston RA, Furey W, Sossenheimer MJ,Ulrich CD, et al. 1996. Hereditary pancreatitis is caused by amutation in the cationic trypsinogen gene. Nat Genet 14:141–145.

Wilentz RE, Geradts J, Offerhaus GHA, Kang M, Goggins M, YeoCJ, Kern SE, Hruban RH. 1998. Inactivation of the p16 (INK4A)tumor-suppressor gene early in pancreatic neoplasia: loss ofintranuclear expression. Cancer Res 58:4740–4744.

Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D, Collins N,Nguyen K, Seal S, Tran T, Averill D, Fields P, Marshall G, NarodS, Lenoir GM, Lynch H, Feunteun J, Devilee P, Cornelisse CJ,Menko FH, Daly PA, Ormiston W, McManus R, Pye C, LewisCM, Cannon-Albright LA, Peto J, Ponder BAJ, Skolnick MH,Easton DF, Goldgar DE, Stratton MR. 1994. Localization of abreast cancer susceptibility gene, BRCA2, to chromosome 13q12–13. Science 265:2088–2090.

Yamada H, Sakamoto H, Taira M, Nishimura S, Shimosato Y, TeradaM, Sugimura T. 1986. Amplifications of both c-Ki-ras with a pointmutation and c-myc in a primary pancreatic cancer and itsmetastatic tumors in lymph nodes. Jpn J Cancer Res 77:370–375.

Yanagisawa A, Ohtake K, Ohashi K, Hori M, Kitagawa T, Sugano H,Kato Y. 1993. Frequent c-Ki-ras oncogene activation in mucous

11MOLECULAR GENETIC BASIS

Page 12: Molecular genetic basis of pancreatic adenocarcinoma

cell hyperplasias of pancreas suffering from chronic inflammation.Cancer Res 53:953–956.

Yonish-Rouach E, Resnitzky D, Lotem J, Sachs L, Kimchi A, OrenM. 1991. Wild-type p53 induces apoptosis of myeloid leukaemiccells that is inhibited by interleukin-6. Nature 352:345–347.

Zawel L, Dai JL, Buckhaults P, Zhou S, Kinzler KW, Vogelstein B,Kern SE. 1998. Human Smad3 and Smad4 are sequence-specifictranscription activators. Mol Cell 1:611–617.

Zhang Y, Feng X, We R, Derynck R. 1996. Receptor-associated Madhomologues synergize as effectors of the TGF-b response. Nature383:168–172.

Zhang Y, Musci TAU, Derynck R. 1997. The tumor suppressor

Smad4/DPC 4 as a central mediator of Smad function. CurrentBiol 7:270–276.

Zhou S, Buckhaults P, Zawel L, Bunz F, Riggins G, Dai JL, Kern SE,Kinzler KW, Vogelstein B. 1998. Targeted deletion of Smad4shows it is required for transforming growth factor-beta and activinsignaling in colorectal cancer cells. Proc Natl Acad Sci USA95:2412–2416.

Zuo L, Weger J, Yang Q, Goldstein AM, Tucker MA, Walker GJ,Hayward N, Dracopoli NC. 1996. Germline mutations in thep16INK4a binding domain of CDK4 in familial melanoma. NatGenet 12:97–99.

12 HILGERS AND KERN