molecular epidemiology of human cancer: contribution of … · [cancer research 58. 4023-4037,...

16
[CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human Cancer: Contribution of Mutation Spectra Studies of Tumor Suppressor Genes S. Perwez Hussain and Curtis C. Harris1 Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland 21201 IS. P. H.¡. and Laboratory of Human Carcinogenesis. National Cancer Institute, N1H, Bethesda, Maryland 20892 ¡C.C. H.] Molecular Epidemiology The identification of individuals at high risk of cancer can offer promising avenues for the prevention of cancer. The majority of human cancer cases are caused by environmental, occupational, and recreational exposures to carcinogens (1). These carcinogens can affect one or multiple stages of carcinogenesis through both genetic and epigenetic mechanisms (2-4). Cancer risk assessment, a highly visible discipline in the field of public health, has historically relied on classical epidemiology, chronic animal bioassays of potential carcin ogens, and the mathematical modeling of these epidemiological and laboratory findings (5). Public health regulators are forced to steer a prudent course of conservative risk assessment because of the limited knowledge of the complex pathobiological processes during carcino genesis: differences in the metabolism of carcinogens and DNA repair capacities, variable genomic stability among animal species, and variation among individuals with inherited cancer predisposition make a definitive analysis of cancer risk extremely difficult. A mo lecular epidemiological approach integrates molecular biology, in vitro and in vivo laboratory models, and biochemistry and epidemi ology to infer individual cancer risk (6-9). Achieving this goal is challenging both current molecular technologies and the epidemio logical designs used to resolve bioethical dilemmas (10, 11). The two major facets of the molecular epidemiology of human cancer risk are the assessment of carcinogen exposure, including biomarkers of effect, and the inherited or acquired host cancer sus ceptibility factors (reviewed in Refs. 12 and 13); thus, it is the gene-environment interaction that determines an individual's cancer risk (Fig. 1). Whereas the external environment is a major source of carcinogens, the cellular microenvironment also contains endogenous carcinogens including oxyradicals generated by cancer-prone chronic inflammatory diseases. Kinzler and Vogelstein (14) have further clas sified cancer susceptibility genes as gatekeepers and caretakers (dis cussed below). The gene-environment paradigm can improve cancer risk assess ment. When combined with carcinogen bioassay in laboratory ani mals, laboratory studies of molecular carcinogenesis, and classical epidemiology, molecular epidemiology can contribute to the four traditional aspects of cancer risk assessment: (a) hazard identification; (b) dose-response assessment; (c) exposure assessment; and (d) risk characterization (Fig. 2). Improved cancer risk assessment has broad public health and economic implications (5). The identification of individuals at high risk of cancer raises com plex bioethical issues (Refs. 10 and 15-17; Fig. 2). One can argue that Received 4/15/98; accepted 7/17/98. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1To whom requests for reprints should be addressed, at Laboratory of Human Carcinogenesis, National Cancer Institute, N1H, Building 37, Room 2C05, Bethesda, MD 20892-4255. Phone: (301) 496-2048; Fax: (301) 496-0497; E-mail: Curtis_Hams@ nih.gov. the knowledge of one's risk can be beneficial. However, more en compassing bioethical issues arise such as an individual's responsi bility to family members and psychosocial concerns regarding the genetic testing of children. Therefore, the uncertainty of the current individual risk assessments and the limited availability of genetic counseling services dictate caution and, many argue, the restriction of genetic testing to those conditions amenable to either preventative or therapeutic intervention. Cancer Susceptibility Genes An intricately balanced control of cellular proliferation and death maintains normal tissue homeostasis and is accomplished by a net work of genes. Many of these genes are implicated in the natural history of human cancer because of their consistent alteration in most types of human cancer. The p53 tumor suppressor gene is a remark able example, because it is mutated in about half of all cancer types arising from a variety of tissues, and missense mutations occur at a high frequency (18-20). Other tumor suppressor genes that are im portant in human oncology, e.g., APC, WT1, or NF1, seem to have a more limited tissue contribution (Table 1). In addition to the consid eration of single genes, recent data indicate the importance of molec ular pathways involving cancer susceptibility genes, e.g., pl6'NK4 and Rb are involved in both the G, checkpoint pathway (21) and the molecular pathogenesis of many types of cancer (22), and APC and ß-catenin are involved in the initiating events in colon carcinogenesis (23). Germline mutations in genes, e.g., p53, RB, pl6'NK4, and APC, have been identified in rare cancer-prone families (Table 1). Somatic mutations in these genes are also frequently found in common spo radic cancers, which attests to the value of studying rare familial cancer syndromes. These cancer susceptibility genes encode proteins that perform diverse cellular functions, including transcription, cell cycle control, DNA repair, and apoptosis. The increased cancer risk of an individual carrying one of these germline mutations can be ex traordinarily high, i.e., more than 1000-fold in xeroderma pigmento- sum (complementation group A-G; Fig. 3), where the "at risk" alÃ-ele is infrequent in the general population. Whereas germline mutations in genes involved in carcinogen metabolism increase cancer risk only severalfold, an at risk alÃ-elecan be very common, e.g., GSTM1 (occurring in about 50% in the Caucasian population), thus making the attributable cancer risk substantial. Caretaker and Gatekeeper Genes Recently, the concept of gatekeeper and caretaker genes character ized by their control of net cellular proliferation or maintenance of genomic integrity, respectively, has been introduced (14, 24). Exam ples of gatekeeper genes include APC and ß-cateninin colon epithe lial cells, Rb in retinal epithelial cells, NF1 in Schwann cells, and VHL in kidney cells. The most prominent example of a gatekeeper is the APC gene in colorectal cancer. It is suggested that an alteration in 4023 on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 28-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

[CANCER RESEARCH 58. 4023-4037, September 15, 1998]

Perspectives in Cancer Research

Molecular Epidemiology of Human Cancer: Contribution of Mutation Spectra

Studies of Tumor Suppressor Genes

S. Perwez Hussain and Curtis C. Harris1

Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland 21201 IS. P. H.¡. and Laboratory of Human Carcinogenesis. National CancerInstitute, N1H, Bethesda, Maryland 20892 ¡C.C. H.]

Molecular Epidemiology

The identification of individuals at high risk of cancer can offerpromising avenues for the prevention of cancer. The majority ofhuman cancer cases are caused by environmental, occupational, andrecreational exposures to carcinogens (1). These carcinogens canaffect one or multiple stages of carcinogenesis through both geneticand epigenetic mechanisms (2-4). Cancer risk assessment, a highly

visible discipline in the field of public health, has historically relied onclassical epidemiology, chronic animal bioassays of potential carcinogens, and the mathematical modeling of these epidemiological andlaboratory findings (5). Public health regulators are forced to steer aprudent course of conservative risk assessment because of the limitedknowledge of the complex pathobiological processes during carcinogenesis: differences in the metabolism of carcinogens and DNA repaircapacities, variable genomic stability among animal species, andvariation among individuals with inherited cancer predispositionmake a definitive analysis of cancer risk extremely difficult. A molecular epidemiological approach integrates molecular biology, invitro and in vivo laboratory models, and biochemistry and epidemiology to infer individual cancer risk (6-9). Achieving this goal is

challenging both current molecular technologies and the epidemiological designs used to resolve bioethical dilemmas (10, 11).

The two major facets of the molecular epidemiology of humancancer risk are the assessment of carcinogen exposure, includingbiomarkers of effect, and the inherited or acquired host cancer susceptibility factors (reviewed in Refs. 12 and 13); thus, it is thegene-environment interaction that determines an individual's cancer

risk (Fig. 1). Whereas the external environment is a major source ofcarcinogens, the cellular microenvironment also contains endogenouscarcinogens including oxyradicals generated by cancer-prone chronic

inflammatory diseases. Kinzler and Vogelstein (14) have further classified cancer susceptibility genes as gatekeepers and caretakers (discussed below).

The gene-environment paradigm can improve cancer risk assess

ment. When combined with carcinogen bioassay in laboratory animals, laboratory studies of molecular carcinogenesis, and classicalepidemiology, molecular epidemiology can contribute to the fourtraditional aspects of cancer risk assessment: (a) hazard identification;(b) dose-response assessment; (c) exposure assessment; and (d) risk

characterization (Fig. 2). Improved cancer risk assessment has broadpublic health and economic implications (5).

The identification of individuals at high risk of cancer raises complex bioethical issues (Refs. 10 and 15-17; Fig. 2). One can argue that

Received 4/15/98; accepted 7/17/98.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordance with18 U.S.C. Section 1734 solely to indicate this fact.

1To whom requests for reprints should be addressed, at Laboratory of Human

Carcinogenesis, National Cancer Institute, N1H, Building 37, Room 2C05, Bethesda, MD20892-4255. Phone: (301) 496-2048; Fax: (301) 496-0497; E-mail: Curtis_Hams@

nih.gov.

the knowledge of one's risk can be beneficial. However, more encompassing bioethical issues arise such as an individual's responsi

bility to family members and psychosocial concerns regarding thegenetic testing of children. Therefore, the uncertainty of the currentindividual risk assessments and the limited availability of geneticcounseling services dictate caution and, many argue, the restriction ofgenetic testing to those conditions amenable to either preventative ortherapeutic intervention.

Cancer Susceptibility Genes

An intricately balanced control of cellular proliferation and deathmaintains normal tissue homeostasis and is accomplished by a network of genes. Many of these genes are implicated in the naturalhistory of human cancer because of their consistent alteration in mosttypes of human cancer. The p53 tumor suppressor gene is a remarkable example, because it is mutated in about half of all cancer typesarising from a variety of tissues, and missense mutations occur at ahigh frequency (18-20). Other tumor suppressor genes that are im

portant in human oncology, e.g., APC, WT1, or NF1, seem to have amore limited tissue contribution (Table 1). In addition to the consideration of single genes, recent data indicate the importance of molecular pathways involving cancer susceptibility genes, e.g., pl6'NK4 and

Rb are involved in both the G, checkpoint pathway (21) and themolecular pathogenesis of many types of cancer (22), and APC andß-catenin are involved in the initiating events in colon carcinogenesis

(23).Germline mutations in genes, e.g., p53, RB, pl6'NK4, and APC,

have been identified in rare cancer-prone families (Table 1). Somatic

mutations in these genes are also frequently found in common sporadic cancers, which attests to the value of studying rare familialcancer syndromes. These cancer susceptibility genes encode proteinsthat perform diverse cellular functions, including transcription, cellcycle control, DNA repair, and apoptosis. The increased cancer risk ofan individual carrying one of these germline mutations can be extraordinarily high, i.e., more than 1000-fold in xeroderma pigmento-sum (complementation group A-G; Fig. 3), where the "at risk" alíele

is infrequent in the general population. Whereas germline mutations ingenes involved in carcinogen metabolism increase cancer risk onlyseveralfold, an at risk alíelecan be very common, e.g., GSTM1(occurring in about 50% in the Caucasian population), thus makingthe attributable cancer risk substantial.

Caretaker and Gatekeeper Genes

Recently, the concept of gatekeeper and caretaker genes characterized by their control of net cellular proliferation or maintenance ofgenomic integrity, respectively, has been introduced (14, 24). Examples of gatekeeper genes include APC and ß-cateninin colon epithe

lial cells, Rb in retinal epithelial cells, NF1 in Schwann cells, and VHLin kidney cells. The most prominent example of a gatekeeper is theAPC gene in colorectal cancer. It is suggested that an alteration in

4023

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

GENE—ENVIRONMENT

/\ /\GatekeeperGenes

CaretakerGenes

ExogenousAgents

EndogenousAgents andMechanisms

Fig. 1. Gene-environment interaction and human cancer risk. Cancer-related genes indifferent human cancers have been classified as gatekeeper (e.g., APC) and caretakergenes (e.g., MSH1 and MLH1). The interaction of these genes with external and internalenvironmental agents can lead to the derangement of regulatory pathways that maintaingenetic stability and cellular proliferation.

APC leads to a derangement of the cellular proliferation pathway thatis important for maintaining a constant cell population. However, thisfunction of APC has a high specificity for colonie epithelial cells butis lacking in most other organs. In murine as well as human coloncancer, APC mutation occurs early on in the process of carcinogenesis(25-27). Although other genes such as K-ras and p53 play important

roles in the later stages of colorectal carcinogenesis, APC mutationand the less common ß-cateninmutations are essential events in the

initiation of neoplasia (reviewed in Ref. 24). If this concept of agatekeeper pathway holds true for the initiation of neoplasia in general, then the identification of other gatekeeper genes can be anticipated.

Unlike gatekeeper genes, caretaker genes, generally maintaingenomic stability and are not involved directly in the initiation of theneoplastic process. Genetic instability due to mutations in caretakergenes enhances the probability of mutation in other genes, includingthose in the gatekeeper pathway. Mismatch DNA repair genes, e.g.,MSH2 and MLH1, are caretaker genes, and abnormalities in thesegenes enhance genomic instability and increase the risk of humancolon cancer. Animal models based on this knowledge of humancolon carcinogenesis have been developed (28-30). Similar in vivo as

well as in vitro models should be developed for other tissue sites ofcarcinogenesis. Breast cancer susceptibility genes BRCA1 and BRCA2have been recently included in the list of caretaker genes (14). In thesame report, it is suggested that a predisposed individual with aninherited mutated alíeleof a caretaker gene is at a lower risk of cancerwhen compared with an individual with a mutated gatekeeper alíele.This difference has been attributed to the finding that three or moreadditional somatic mutations are required to initiate neoplasia in thecaretaker pathway, whereas only one additional somatic mutation isrequired to initiate neoplasia in the gatekeeper pathway. In addition tocancer as an endpoint, the molecular analysis of gatekeeper andcaretaker genes in presumed preneoplastic morphological lesionscould provide short-term and less expensive pathobiological end-

points for hazard identification and molecular epidemiological studies.

Mutator Concept

Multiple mutations are found in cancer cells. The presence of amutator phenotype has been suggested as an important step in tumordevelopment and forms the basis of the mutator hypothesis (Ref. 31,reviewed in Ref. 32). MIS2 arises from either deletions or insertions

in short repetitive sequences in the human genome known as micro-

satellites, and it is used as a measure of genetic instability. Theoccurrence of MIS was initially reported in colorectal cancer, partic-

2 The abbreviations used are: MIS, microsatellite instability; HNPCC, hereditary

nonpolyposis colorectal cancer; BP, benzo(a)pyrene; BPDE. BP-7,8-diol 9,10-epoxide.

ularly HNPCC (33-35). The presence of MIS as a biomarker of a

mutator phenotype has been linked to the defect in mismatch repairgenes, e.g., HMSH2, hMLHl, hPMSl, and HPMS2 (36-39). Tissue ofMSH2-null mice shows hypermutability in a transgenic lacl-reportersystem on exposure to /V-methyl-Ar-nitrosourea (40). A proportion of

HNPCC with MIS does not show a mutation in mismatch repairgenes, which indicates the possible involvement of other mutatorgenes. In a recent study of five Japanese HNPCC families without agermline mutation in MSH2 or MLH1, one family showed MIS and agermline mutation in MSH6/GTP (41). However, tumors in mice withnull mutations in MSH6 do not show MIS (42), which indicatespossible subtle differences in DNA mismatch repair among animalspecies.

Candidate mutator genes are involved in multiple cellular functionsthat are important for maintaining genetic instability, such as DNArepair, DNA replication, chromosomal segregation, cell cycle regulation, and apoptosis. One example is the p53 tumor suppressor gene.Cells lacking expression of wild-type p53 displayed a high frequencyof gene amplification, but the expression of wild-type p53 in Li-

Fraumeni syndrome cells with mutant p53 reduced the frequency ofPALA (a uridine biosynthesis inhibitor)-selected gene amplification

(43, 44). In murine cells, p53 mutations lead to genomic instabilityand the amplification of drug-resistant genes (43). The role of p53 in

DNA repair is less certain. A number of studies have shown p53modulation of nucleotide excision repair in which the loss of p53functions leads to a decrease in DNA repair and thus an increase ingenomic instability (45-51). Suppression of spontaneous homologous

recombination is another suggested function of p53 that contributes tothe maintenance of genetic stability. Isogenic cell pairs from tumorcells or primary fibroblasts showed a 100-fold increase in the recom

bination rate when p53 is inactivated (52). Transfection of normalcells with either the dominant-negative p53ala143 variant expression

vector or the human papilloma virus E6 gene showed a 10-80-fold

higher spontaneous recombination rate (53). p53 is also involved inthe fidelity of centrosoma! duplication, which ensures balanced segregation of the chromosomes during cell division. Mouse embryofibroblasts lacking p53 protein produce multiple copies of centro-

somes (54), resulting in unequal segregation of the chromosomes andgenetic instability. Mutant p53 may increase the sensitivity of cells tomutagens. A 5-fold increase in the mutation frequency, as determinedby the appearance of hypoxanthine guanine phosphoribosyl trans-ferase-deficient (6-thioguanine-resistant) cells (induced by X-rays),was observed in human fibroblast SUSM-I/p53 cells containing amutant p53 gene (codon 273 Arg-His) when compared with theparental SUSM-I cells (55). Human lymphoblastoid cell lines TK6and WTK1 containing wild-type and mutant p53, respectively,showed different levels of (X-ray-induced) mutagenicity and apoptosis (56). A 28-fold increase in the spontaneous mutation frequency atthe heterozygous thymidine locus was observed in WTK1 cells (p53-

mutated) when compared with TK6 (normal) cells (57). TK6 cellswere also more sensitive at the tk locus to the mutagenic effects ofX-rays, ethylmethane sulfonate, methylmethane sulfonate, and mito-

mycin C. Mutant p53 induced an increase in the frequency of chromosomal breaks in dividing LIM12 cells (58). A study of the replication of the pZ402 shuttle vector containing the supF gene inLi-Fraumeni fibroblasts with mutated p53 alíeleO showed a frequentdeletion of a portion of SV40 T antigen (58). A 20-fold increase in

silent mutations has been observed in the mutated p53 gene in humancancer, which indicates its hypermutability (59). These silent mutations were not found to be selected or represented as genetic polymorphisms in the normal population. However, the fact that thealteration in p53 has been described as a relatively late event incolorectal carcinogenesis argues against its candidacy as a mutator

4024

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Laboratory Animal Studies

1Cancer Epidemiology

Molecular Epidemiology

_L_L

Molecular Dosimetry of CarcinogenExposure

— Carcinogen-macromolecular adducts

— Cytogenetic endpoints

— Mutational spectra and frequency

f

Internal Exposure Assessment

1Inherited Cancer Predisposition

— Genetic polymorphism of enzymes involved in

activation and detoxification of carcinogens— Genetic polymorphisms in DMA repair genes, DMA

repair deficiency, and genomic instability— Germline mutations or polymorphisms in tumor

suppressor genes

Host Susceptibility Assessment

_LHuman Cancer Risk Assessment•Hazard Identification•Dose Response Assessment•Exposure Assessment•Risk Characterization

_LBioethical Issues•Autonomy•Privacy•Justice•Equity

QualitySensitivitySpecificityEffectivenessLimit genetic testing to conditionsthat are correctable by successfulintervention

Intervention•Reduce carcinogen exposure•Increase medical surveillance•Therapeutic strategies including chemoprevention•Formulation of health policy

Fig. 2. Human cancer risk assessment and bioethical issues associated with molecular epidemiology and human cancer.

gene involved in the initiation of sporadic colorectal carcinoma (60).For example, there was a lack of any association between p53 statusand MIS or loss of heterozygosity in a recent study analyzing 58sporadic colorectal tumors (61). An analysis of 40 primary gastriccarcinomas showed a negative correlation between p53 mutation andMIS and indicates two distinct pathways for their contributions incancer development (62). In the same study, a positive associationwas found between MIS and mutation in the TGFß-RIIgene. Treatment of wild-type and p53-null mouse embryo fibroblasts with 4-nitroquinoline 1-oxide did not show any difference in accumulation of

point mutations in the lad target gene (63). Also, in the same study,DNA from thymus and thy mie tumors in both p53+/+ and p53—/—

mice did not show any distinct difference in the mutation frequency inthe target gene. However, the spontaneous mutation frequency of thelad gene was high, i.e., 10~5, so that the modulating effects of p53,

if any, might not be detected. In addition, the rate of nucleotideexcision repair is substantially lower in rodent-derived cells than it isin human cells (64, 65), and the repair of UV damage in the nontran-

scribed strand of the active gene is markedly reduced in rodents whencompared with that of humans (66).

Mutational Spectra of Tumor Suppressor Genes

Several endogenous and exogenous mutagens have been describedas inducing a characteristic pattern of DNA alteration. Mutationalspectra analyses are required to study the type, location, and frequency of DNA changes. Alterations of cancer-related genes found in

tumors not only represent the interaction of a carcinogen with DNAand cellular DNA repair processes but also reflect the selection ofthose mutations that provide premalignant and malignant cells with aclonal growth and survival advantage. Study of the frequency, timing,

and mutational spectra of p53 and other cancer-related genes provides

insight into the etiology and molecular pathogenesis of cancer andgenerates hypotheses for future investigations. These include questions regarding carcinogen-DNA interactions, the function of the

affected gene products, the mechanism of carcinogenesis in specificorgans or tissues, and general cell biological processes such as DNAreplication and repair.

Nonsense mutations, deletions, and insertions are the most frequenttypes of mutations in tumor suppressor genes that produce either anabsentee or a truncated protein product. These mutations are clearlyloss-of-function mutations. In contrast, the p53 tumor suppressor gene

shows an unusual spectrum of mutations when compared with othersuppressor genes, e.g., APC, BRCA1. or ATM, in which the mutationslead to a loss of function (Fig. 4). Missense mutations, in which theencoded protein contains amino acid substitutions, are commonlyfound in the p53 tumor suppressor gene. p53 missense mutations cancause both a loss of tumor suppressor function and a gain of onco-

genic function by changing the repertoire of genes whose expressionis controlled by this transcription factor (67-69). This functional

duality may be one explanation for the high frequency of p53 mutations in human cancer.

p53

The p53 gene is well suited for mutational spectrum analysis forseveral reasons: (a) p53 mutations are common in many humancancers, and a sizeable database of more than 8000 entries hasaccrued, so that the analysis of this large database can yield statistically valid conclusions (70, 71) and can be readily accessed on theWorld Wide Web (http://www.iarc.fr/p53/homepage.html); (b) themodest size of the p53 gene ( 11 exons, 393 amino acids) permits the

4025

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Table 1 Examples of tumor suppressor genes involved in human cancers"

Tumorsuppressorgenep53RBIAPCATMChromosomallocus17pl3.113ql45q21Ilq22Location/proposedfunctionNuclear/transcription

factorNuclear/transcription

modifierCytoplasmic/signal

transductionNuclear/kinase

Ser/Major

typesof somatic

mutationMissenseDeletion

andnonsenseDeletion

andnonsense

DeletionSomatic

mutationsExamples

of neoplasms withsomaticmutationsMost

human cancer typesexamined todateRetinoblastoma,

osteosarcoma,and carcinomas ofthebreast,

prostate, bladder,andlungCarcinoma

of the colon,stomach, andpancreasLeukemiaInherited

mutationsSyndromeLi-FraumeniRetinoblastomaFamilialade

nornalous,polyposis

Ataxia telangiectasiaHétérozygotecarrier

rate/IO5births'7~2~2-10

2Typical

neoplasmsCarcinomas

of the breast andadrenal cortex,sarcomas.leukemia,

and braintumorsRetinoblastoma,and

osteosarcomaCarcinomas

of the colon,thyroid, andstomachLeukemia

and lymphoma

WTI

BRCA1

BRCA2

NF1

NF2

pl6INK"

VHL

null p13 Nuclear/transcription

factor17q21 Nuclear/DNA repair

13q12-13 Nuclear/DNA repair

17qll

22q

9p21

3p25

Cytoplasmic/GTPaseactivating protein

Cytoplasmic/cytoskeletal-membrane linkage

Nuclear/cyclin-dependent kinaseinhibitor

Nuclear/adaptor

Missense

Deletionandnonsense

Deletionandnonsense

Deletion

Deletionandnonsense

Deletionandnonsense

Deletion

Wilms' tumor

Breast

Breast

Schwannomas

Schwannomas andmeningiomas

Mesothelioma, pancreasmelanoma, and glioblastoma

Unknown

Wilms' tumor

Familialbreast/ovarian

Familialbreast/ovarian

Neurofibromatosistype 1

Neurofibromatosistype 2

Familial melanoma

von Hippel-Lmdau

-0.5-1

-2-10-1

Wilms' tumor

Carcinoma of the breast andovary

—2-10 Carcinoma of the breast andovary

—30 Neural tumors

~3 Central Schwannomas andmeningiomas

? Melanoma

-3 Hemangioblastoma and renalcell carcinoma

°Reviewed in Ref. 201 (see also Refs. 202-204).

Ref. 205 and A. Knudson, personal communication.

study of the entire coding region, and it is highly conserved invertebrates, allowing the extrapolation of data from animal models(72); and (c) the point mutations that alter p53 function are distributedover a large region of the molecule, especially in the hydrophobicmidportion (Refs. 18, 19, and 73; Fig. 5). These numerous basesubstitutions alter p53 conformation and sequence-specific transacti-

vation activity; thus, correlations between distinct mutants and functional changes are possible. Frameshift and nonsense mutations thattruncate the protein are located outside of these regions (Fig. 5), anddeletions and insertions are most commonly found in exons 2-4 and9-11 (74), so evaluation of the entire DNA sequence yields relevant

data. This situation differs from that of the raíoncogenes, whosetransforming mutations occur primarily in three codons, a few sequence-specific motifs, and a critical functional domain (75). The

diversity of p53 mutational events permits more extensive inferencesof the mechanism of DNA damage involved.

Exogenous mutagenic agents and/or endogenous mutagenic

HIGH GERMLINE MUTATIONSLOW(103-to104

i-fold)CANCER

RISK(2-10-fold),

Rb (Retinoblastoma)p53 (Li-Fraumeni)APC (Familial Polyposis Coll)NF1 (Neurofibromatosis)XPA-G (XerodermaPigmentosum)ATM

(Ataxia Telangiectasia)BRCA1 (Breast-Ovarian Ca)HMLH1 (Hereditary Nonpolyposis

ColorectalCa)CYP1A1"1

CYP2E1 \.CarcinogenNATi Metabolism ,GSTM1 J '(1-5

per 10slivebirths)FREQUENCY

OF AT RISKALLELE(2-50

perlO2livebirths)MODERATE

HIGH

Fig. 3. Examples of the inverse correlation between allelic frequency and cancer riskassociated with inherited cancer susceptibility genes.

mechanisms have been implicated in the induction of mutations.These mutations are archived in the spectrum of p53 mutationsfound in human cancer (18, 19, 73, 76-78). Errors introduced

during DNA replication, RNA splicing, DNA repair, and DNAdeamination are examples of endogenous mutagenic mechanisms.The DNA sequence context is an important factor that determinesthese events. Almost all short deletions and insertions occur atmonotonie runs of two or more identical bases or at repeats of2-8-bp DNA motifs, either in tandem or separated by a short

intervening sequence (74). The mechanism that has been studiedmost is called slipped mispairing, a misalignment of the templateDNA strands during replication that leads to either deletion, if thenucleotides excluded from pairing are on the template strand, orinsertion, if the nucleotides excluded are on the primer strand (79).When direct repeat sequences mispair with a complementary motifnearby, the intervening oligonucleotide sequence may form a loopbetween the two repeat motifs and be deleted (80, 81). Morelengthy runs and sequence repeats are more likely to generateframeshift mutations. The deletions and insertions in the p53 genefound in human tumors also may be biologically selected from thebroad array of such mutations occurring in human cells. Whencompared with the distribution of missense mutations, these typesof mutations occur more frequently in exons 2-4 (54%) and 9-11(77%) than in exons 5-8 (20%; Fig. 5). The NH2 terminus of thep53 protein (encoded by exons 2-4; reviewed in Refs. 82-85) has

an abundance of acidic amino acids that are involved in thetranscriptional function of p53 (86, 87) and binds to transcriptionfactors such as TATA-binding protein in transcription factor IID(82, 88-91). Experimental studies have shown that multiple point

mutations in this domain are required to inactivate its transcriptional transactivation function (92). The COOH terminus (encodedby exons 9-11) of the p53 protein is enriched in basic amino acids

4026

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Fig. 4. Class of mutations in p53, APC, BRCAI,and ATM genes in all human cancers. Missensemutations represent a high proportion of p53 mutations, whereas nonmissense mutations (e.g., frame-

shift, nonsense, and splice site mutations) are common in other tumor suppressor genes. Deletions andinsertions that do not induce frameshifts have beenreferred to as in frame deletions.

p53 (n=6,604)... Nonsense

Frameshift 11% 7o/Splice site 4%

In FrameDel/Ins 2%Silent 2%

Missense74%

ATM (n=130)

Frameshift77%

ARC (n=352)

Frameshift 41%

Splice site 6%

Nonsense24%

Missense16%

Silent 13%

Nonsense15%

Missense6%

In FrameDel/Ins 2%

Frameshift64%

BRCA-1 (n=580)

Nonsense 5%

Missense29%

Splice site2%

that are important in: (a) the oligomerization and nuclear localization ofthe p53 protein (reviewed in Refs. 93-96); (b) the recognition of DNA

damage (97, 98); (c) the negative regulation of p53 binding to promotersequences; (d) the transcription of p53-transactivated genes (99); and (e)

the induction of apoptosis (100). Laboratory studies have shown that at

least two point mutations in the NH2 terminus of p53 are required toinhibit its transcriptional transactivity (92); therefore, deletions and insertions are a more detrimental mutagenic mechanism than single pointmutations for disrupting these NH2-terminal and COOH-terminal func

tional domains.

Transactivation I-Domain

Mutations andSequence Specific

DNA Binding Domain

Tetramerization,Nuclear Localizationand DNA-DamageRecognition Domain

17-29 .97 EVOLUTIONARILY CONSERVED 292, 324 — 352

Frameshift27%

Exons 2-4 Exons 5-8Nonsense Frameshift 10% Nonsense

9% Splice site 3%

Exons 9-11

Splice site15%

Silent 8%

Silent 3%Missense

41%

Frameshift23%

n=236

Splice site \'Missense 15%

78% Silent 7%^n=6177 n=129

Nonsense27%

Missense28%

Fig. 5. Schematic of the p53 molecule. The p53 protein consists of 393 amino acids with functional domains, evolutionary conserved domains, and regions designated as mutationalhotspots. Functional domains include the transactivation region (amino acids 20-42. ^), the sequence-specific DNA-binding region (amino acids 100-293), the nuclear localizationsequence (amino acids 316-325, 01), and the oligomerization region (amino acids 319-360, B). Cellular or oncoviral proteins bind to specific areas of the p53 protein. Evolutionaryconserved domains (amino acids 17-29,97-292, and 324-352; black areas) were determined using the Multiple Alignment Construction and Analysis Workbench (MACAW) program.Seven mutational hotspot regions within the large conserved domain are identified [amino acids 130-142, 151-164, 171-181, 193-200, 213-223, 234-258, and 270-286 (checkered

blocks)}. Vertical lines above the schematic, missense mutations.

4027

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Structure-Function Relationship of p53

The p53 mutation spectrum can also provide clues to the criticalfunctional regions of the gene that, when mutated, contribute to thecarcinogenic process. Because about 75% of the missense mutationsare in the sequence-specific DNA-binding midregion of the protein

(Refs. 18, 19, and 73; Fig. 5), investigators have focused on thetranscription transactivator function of p53. However, these missensemutations and the resultant amino acid substitutions can cause aberrant protein conformations (101) that may alter other functional domains, including those in the COOH terminus of the p53 protein. Thispositively charged region contains the putative major nuclear localization signal (amino acids 316-325), the oligomerization domain(amino acids 319-360), and a DNA damage binding domain (aminoacids 318-393; Refs. 102-105). p53 sequence-specific DNA bindingand transcriptional transactivation can also be modulated by posttrans-

lational mechanisms, including serine phosphorylation (103, 106) andthe redox regulation of the cysteine residues responsible for bindingzinc to p53 (107-109). Recently, it has been demonstrated that acety-lation of the p53 COOH-terminal domain at amino acids 373 and 382stimulates its sequence-specific DNA-binding activity (110). Furthermore, in another recent study, O-glycosylation of the p53 COOHterminus has been shown to activate DNA binding (111). In ML-1cells exposed to Adriamycin or cisplatin, a COOH-terminal cleavage

product of endogenous p53 has been shown to correlate with theup-regulation of p21 (112). The function-structure relationship re

vealed by the analysis of the p53 mutation spectrum (18, 73), itsnuclear magnetic resonance and crystallographic three-dimensionalstructure (93, 94, 113), and functional studies of wild-type versus

mutant p53 activity (reviewed in Ref. 83) have generated both hypotheses for further study and strategies for the development ofrational cancer therapy.

Missense mutations in the p53 gene domains encoding loop 2 or 3have been associated with more aggressive breast cancer (114) thatwas less responsive to chemotherapy with doxorubicin (115). Unlikemutations affecting interaction at the interface between the p53 protein and its consensus sequences in DNA, loop 2 or 3 mutants are lesslikely to be temperature sensitive. Additional studies are warranted todetermine the importance of somatic p53 mutation in specific structural and functional domains and the success of cancer therapy. Thestructural and functional consequences of specific germline p53 mutations in Li-Fraumeni syndrome families should also be investigated.

Mutation spectra studies of DNA repair genes such as XPD haveshown dramatic gene phenotype effects (116). Germline mutations inXPD can exhibit either a phenotype of trichodystrophy without anincreased risk of cancer or xeroderma pigmentosum with a > 1000increased risk of sunlight-induced skin cancer. Therefore, similar genephenotype effects are likely to be found in Li-Fraumeni families.

DNA Methylation and Mutational Hot Spots

Methylated CpG sites of p53 harbor a strikingly high proportion ofmutations that constitute major hotspots in human cancer (reviewed inRef. 73). Deamination of 5-methylcytosine at CpG sites is considered

to be one of the major endogenous mechanisms for the induction ofthese mutations (117). Deamination of 5-methylcytosine forms thy-midine and generates a G-T mismatch which, if not repaired, produces

a C to T transition. Deamination of cytosine can also generate a C toT transition if uracil glycosylase and a G-T mismatch repair are

inefficient. The presence of a high frequency of C to T transitions atCpG dinucleotides in colon carcinomas (18) suggests the involvementof an endogenous deamination mechanism. However, a considerablevariation also has been reported in mutational type at CpG sitesamong different types of cancer. Compared to colon carcinoma, low

frequencies of transition mutations are observed in liver and lungcancer at CpG dinucleotide sites, and, in contrast, high frequencies ofG to T transversions are reported in these cancer sites (73). A distinctpattern of mutation at CpG sites in different tissue types indicateseither the possible involvement of exogenous agents or the selectionof p53 mutants that vary among tissues in their pathobiologicalactivities (118).

Recently, a role for cytosine methylation at CpG sites has beensuggested in determining the preferential site of DNA damage byexogenous carcinogen. Interestingly, treatment of HeLa cells andbronchial epithelial cells with BPDE selectively induced guanineadduci formation at CpG sites inp53 codons 157, 248, and 273 (119).In contrast to nonmethylated cloned p53 gene sequences containingnonmethylated cytosines, treatment of methylated sequences withBPDE preferentially produced BPDE-guanine adducts at CpG sites

(120). In the same study, a similar trend also is observed in the PGK1gene, with different methylation status, derived from an active orinactive X chromosome. BPDE, aflatoxin B,, 8,9-epoxide, /V-acetoxy-2-acetylaminofluorene, and benzo(g)chrysene diol epoxide, which

interact with different moieties within guanine, showed more bindingat methylated CpG sites than at nonmethylated CpG sites in the hot-

spot codons of p53 (121). The precise role by which methylationresults in the preferential interaction of BPDE is not clear; however,its influence in the creation of an intercalation site has been suggested(120). Rates of DNA repair may vary among p53 exons (122).

DNA Strand Bias of p53 Mutation

The transcribed strand of an active gene is usually repaired at amore rapid rate than the nontranscribed (DNA coding) strand (123).For example, DNA damage on the transcribed strand of the p53 geneis repaired faster compared with that of the nontranscribing or codingstrand (124, 125). This preferential DNA repair is suggested as animportant factor in determining the mutation pattern leading to strandbias (123, 126). The positive correlation between cigarette smokingand G:C to T:A transversions on the nontranscribed strand of p53 inlung cancer (127) supports the above-mentioned hypothesis. Labora

tory studies have also shown that the mutations induced by exogenouschemical carcinogens occur preferentially on the nontranscribed coding strand of p53 (128-130). C to T transitions at CpG sites are a

common mutation produced by endogenous mechanisms such asdeamination of 5-methylcytosine (117, 131, 132). It can be hypothe

sized that C:G to T:A transitions arising from the deamination of5-methylcytosine at CpG dinucleotide sites in the p53 gene would not

display a DNA strand bias, whereas C:G to T:A transitions arisingfrom bulky chemical DNA adducts would occur primarily on thenontranscribed strand, due to the efficient repair of the transcribedstrand. Alternatively, the reported association between transcription-

coupled repair and mismatch repair (133) implies a possible strandbias for mismatch repair. Analysis of the p53 mutation database (70)for C to T transitions at CpG sites at mutational hotspot codons on thenontranscribed strand showed either bias for both transcribed andnontranscribed strands or no bias, depending on the specific codonand cancer type (Table 2). Although the total number of mutations issmall, mutations at codon 175 (nucleotide residue 13,202) show asubstantial bias for the transcribed strand in breast, colon, esophageal,head and neck, and lung cancers. In contrast, mutations at codon 282(nucleotide residue 14,513) show a strong bias for the nontranscribedstrand in brain, breast, colon, esophageal, head and neck, and lungcancers. Mutations at codon 248 (nucleotide residue 14,068) show nostrand bias in breast, colon, and head and neck cancers, whereasesophageal and lung cancers show a bias for the nontranscribedstrand. Mutations at codon 248 (nucleotide residue 14,068) only show

4028

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 7: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Table 2 DNA strand bias at somatic and germline mutation hotspots in the p53 tumor suppressor gene

AB.Cancer

type.

Somaticmutation:BrainBreastColonEsophagealHead

andneckLiverLung%

codon157%

codonn175C

to T at CpG sites(nontranscribedNANANANANANANA<5<5<5<5<5<5<5strand)0NA0300NA0n<521732713<5S%

codon24832424586423864n2833101726814%

codon27324654336179248n17606214121221%codon28288100100100100NA100n784086<513G

lo T at CpG siles (nonlranscribedstrand)0BrainBreastColonEsophagealHead

andneckLiverLungC.

GermlinemutationGermlineNA100NANA100100100:C to T atCpGNA<55<5<571123NANA80NANANANA<5<55<5<5<5<5NANANA100NANA100<5<5<56<5<512NANANANA1008388<5<5<5<57524NANANANANANANA<5<5<5<5<5<5<5sites(nontranscribedstrand)"'6<5NA<5205164369

" The number of mutations less than five (<5) was not assessed (NA; Ref. 50).b The germline data do not include multiple mutations from the same family.

a bias for the nontranscribed strand in brain and liver cancers. Mutations at codon 273 (nucleotide residue 2,896) show a transcribedstrand bias in brain, esophageal, and head and neck cancers but showa nontranscribed strand bias in breast and liver cancers. No strand biaswas observed at this codon in colon and lung cancers. One possibilityof the occurrence of strand bias for C to T transitions at CpG sitescould be due to the different rate or the fidelity of G-T mismatch

repair, depending on either the surrounding DNA sequences of themutational hotspot sites or differences in the etiological agents amongcancer types. These findings are consistent with the hypothesis thatchemical carcinogens found in the environment, diet, or tobaccosmoke are responsible for this subset of p53 mutations.

The frequency of G to T transversions at CpG sites is found to berelatively low and exhibits a strong bias for the nontranscribed strandat a number of mutational hotspots. All G to T transversions at CpGsites at codon 157 (nucleotide residue 13,147) occur on the nontranscribed strand in breast, head and neck, liver, and lung cancers.Similarly, at codon 248 (nucleotide residue 14,069), a strong bias forthe nontranscribed strand exists in esophageal and lung cancers. G toT transversions at codon 175 (nucleotide residue 13,203) in coloncancer and codon 282 (nucleotide residue 14,514) in lung, liver, andhead and neck cancers also show a substantial bias for the nontran

scribed strand. The presence of strong bias for the nontranscribedstrand in G to T trans versions at CpG sites of hotspot codons suggeststhe involvement of exogenous chemical carcinogens that form bulkyDNA adducts, rather than the endogenous mechanism such as thedeamination of 5-methylcytosine.

Interestingly, analysis of germline C to T transitions at CpG sites ofhotspot codons in Li-Fraumeni families shows a considerable bias for

the transcribed strand (Table 2). G to T germline mutations at CpGsites are not reported in these families.

pl6'NK4

pl6'NK4, which encodes a cell cycle-inhibitory protein, has been

found to be commonly mutated in a variety of human cancers (134-140). Epigenetic control of pl6INK4 expression by DNA methylation

may also inactivate this cancer susceptibility gene (141, 142). Germ-line mutations in p!6INK4 have been associated with familial cancers

including melanoma, pancreatic cancer, and head and neck cancer(143). Because of the common occurrence of germline mutations inmelanoma families, pl6'NK4 has been considered a melanoma suscep

tibility gene; however, half of the chromosome 9p-linked melanomafamilies do not show pl6'NK4 germline mutations. In vitro studies

Germline(n=19)

Somatic(n=96)

Del. + ins.5%

G:C->C:G

A:T->T:A16%

CC:GG->TT:AA 4%

Del. + ins33%

G:C->A:Tat Non-CpG

26%

G:C->C:G16%

G:C->T:A21%

A:T->C:G 2%G:C->A:T A:T->G:C 3%at CpG 5% A:T->T:A 2%

G:C->T:A23%

Polymorphism(n=37)

G:C->C:GA:T->G:C 8% 3% Q:C->T:AA:T->T:A

3%

G:C->A:Tat Non-CpG

27%

G:C->A:Tat CpG 21%

G:C->A:Tat Non-CpG 11%

G:C->A:Tat CpG

48%

Fig. 6. Mutation spectra of pI6INK4 in human cancers. Germline and somatic pl6INK4 mutations show distinct patterns. Deletions and insertions constitute 33% of the somatic

mutations, whereas only 5% of the germline mutations represent this class of mutation.

4029

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 8: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Fig. 7. Germline mutational spectra in BRCA1and BRCA2. Deletions and insertions represent themajor types of mutations in both BRCA1 andBRCA2. Among the less common missense mutations, G:C to A:T and A:T to G:C transitions represent 17 and 12%, respectively, of the total mutations in BRCA1. G:C to A:T and A:T to G:Ctransitions represent only 6 and 5%, respectively, ofthe total mutations in BRCA2.

BRCA1 (n=889)G:C->C:G

Del. + ins59%

G:C->T:A 6%G:C->A:T

at CpG 5%G:C->A:T at

non-CpG 12%A:T->T:A 1%

A:T->G:C 12%

A:T->C:G 4%

BRCA2 (n=168)G:C->C:G 2%

Del. + ins80%

G:C->T:A 3%G:C->A:TatCpG4%

G:C->A:Tatnon-CpG 2%A:T->T:A 2%A:T->G:C 5%

A:T->C:G 2%

have shown that germline melanoma-predisposing mutants are unableto inhibit the catalytic activity of cyclin D1/CDK4 and cyclin Dl/CDK6 complexes (144). The absence of pl6'NK4 alteration in a largeportion of the melanoma-prone families linked to chromosome 9p21supports the possible existence of additional melanoma susceptibilitygenes. There are some interesting distinctions between the germlineand somatic pl6'NK4 mutational spectra (Fig. 6). Deletions and insertions, which constitute 33% of the somatic pl6INK4 mutation, are

represented by only 5% of germline mutations. CC to TT tandemdouble mutations, which are characteristically induced by UV radiation, constitute 4% of somatic p!6INK4mutations but are never found

as a germline mutation. Although several germline polymorphismsalso have been reported for pl6'NK4, their functional and prognostic

significance is not yet known.

BRCA1 and BRCA2

BRCA1 on chromosome 17ql2-21 and BRCA2 on chromosome13ql2-13 have been recently identified as cancer susceptibility genesthat are involved in the familial predisposition to breast cancer (145-151). In addition to breast cancer, germline mutations in BRCA1 alsopredispose individuals to ovarian cancer (152-154), whereas BRCA2

mutations are implicated in male breast and pancreatic cancers (155-157). Mutations in these genes are considered to be responsible forabout 80% of familial breast cancer cases. In contrast to other cancersusceptibility genes, somatic mutations in BRCA1 or BRCA2 areinfrequent in breast and ovarian cancers (158-162). Mutation spectrastudies of BRCA1 and BRCA2 have revealed that the majority of themutations lead to a loss of function and include deletions and insertions leading to frameshift and nonsense mutations. Deletion andinsertion mutations constitute about 60% of all mutations in BRCA1and 80% of all mutations in BRCA2(Fig. 7). However, a considerablenumber of missense mutations have been reported previously (163).The major missense mutations reported in BRCA1 are G:C to A:Ttransitions (17%, with 12% at non-CpG sites) and A:T to G:C transitions (12%), and in BRCA2,only 6% of the total mutations are G:Cto A:T transversions (with 2% at non-CpG sites), and 5% are A:T toG:C transitions. Although there are 100 different mutations reportedthroughout the BRCA1gene, two mutations, i.e., 185delAG and 5382insC, occur with considerably high frequencies and constitute about20% of the total mutations reported so far. Interestingly, one of thesemutations, 185delAG, has an ethnic commonality and segregates withearly-onset breast cancers among 20% of Ashkenazi Jews (164). In a

CODONI 57

Colon19%

CODON 175 CODON 179

Head &Neck 11%

Lung 7%Liver 2HeadiNeck5%

CODON 248

Colon17%

Head &Neck 27%

Breast20%

(îolon66%

CODON 249

Skin Breast3% 6% Colon

3%Head&

Neck5%

Liver69%

Breast19%

Colon15%

Head&Neck

Liver 5%4%

CODON 278

Breast

Skin59%

Colon5%Head&

Neck16%

Liver2%

Lung9%

Fig. 8. p53 mutational hotspots in human cancers. Most types of human cancers show the domination of specific p53 mutations at particular mutational hotspots. The characteristicpatterns hypothesize molecular linkage between a particular cancer and a specific exogenous or endogenous carcinogen.

4030

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 9: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

LUNG (64%)

BREAST (15%)

HEAD & NECK (20%)

CodonsFig. 9. p53 mutational hotspots of G to T Iransversions in human breast, head and neck, and lung cancers. Codon 157 is one of the mut alumal hotspots in lung cancer. A transversion

of G to T at codon 157 in lung cancer is found more frequently among smokers than never-smokers and is a candidate early marker for identifying individuals at higher cancer risk.

recent study involving 5318 Ashkenazi Jews, the 185delAG and5382insC mutations in BRCA1 and the 6174delT mutation in BRCA2were analyzed. Among 120 carriers oíBRCA1 and BRCA2 mutations,the estimated risk at the age of 70 years was reported to be 56% forbreast cancer and 16% for both ovarian cancer and prostate cancer(165). Similar to BRCA1, a large percentage of known BRCA2 germ-

line mutations (33%) is represented by two deletion mutations,6174delT and 997del5. The 6174delT is found in about 8% of early-

onset breast cancer cases in the Ashkenazi Jewish population (166),whereas 997del5 has been commonly reported in early-onset familial

breast cancer cases from Iceland (153). Another interesting observation is the apparent occurrence of genotype-phenotype correlation

with respect to a specific BRCA1 or BRCA2 mutation and the risk ofbreast and ovarian cancers. The presence of a truncating mutation inthe first two-thirds of the BRCA1 gene instead of in the last third of the

gene may significantly increase the risk of ovarian cancer in comparison with that of breast cancer (153). Likewise, the analysis of 25families with multiple cases of breast and ovarian cancers withBRCA2 mutations also showed a significant genotype-phenotype cor

relation. Mutations leading to the truncation of BRCA2 in familieswith the highest risk of ovarian cancer were all found to be clusteredin a region of approximately 3.3 kb in exon 11 (154).

Although the normal functions of BRCA1 and BRCA2 are notknown, recent studies are providing interesting clues. For example,the transcriptional activation function of BRCAl and BRCA2 has beenreported, and the COOH-terminal region of BRCAl (amino acids1528-1863) showed significant transcriptional activation when fusedto the GAL4 DNA-binding domain; this transactivation function waslost when a mutation was introduced in the COOH-terminal region

(167). BRCA2 exon 3 has been found to have a sequence homologyfor the activation domain of c-Jun and showed transcriptional activation in yeast when linked to the lex-A DNA-binding domain as well

as in two different mammalian cell lines, U20S and NMuMG, whenlinked to the GAL4 DNA-binding domain (168). BRCAl and BRCA2

have been suggested to play a role in embryonic cellular proliferationand development (169, 170). Mutations in BRCAl or BRCA2 causelethality at different stages of development in mouse embryos. Interestingly, BACA5"6 mutants (deletion of the fifth and sixth exons) with

a homozygous null p53 or p21 background showed an enhancedsurvival of embryos (171), suggesting a functional interaction between BRCAl, p53, and p21. An increased expression of p21 hasbeen observed in the BRCA5"6 mutants (169). Furthermore, the

formation of a complex between BRCA1/BRCA2 and Rad 51 andhypersensitivity of BRCA2 mutant mouse embryos to y radiation hasindicated a role for BRCAl and BRCA2 genes in DNA repair pathways (170, 172, 173). These functions of BRCA] and BRCA2, whichhave been suggested to play a role in maintaining genomic stability,give BRCAl and BRCA2 an important place in the cancer susceptibility gene family. The finding of inherited mutations in the BRCAland BRCA2 genes in families with early-onset breast and ovarian

cancers raises the significance of predictive testing or early diagnosiswhile prompting the debate of several technical and bioethical concerns (reviewed in Ref. 151).

Considering the variety of hereditary cancers and alleile deletions,one can predict the discovery of additional tumor suppressor genes,some of which may have a conspicuous role in carcinogenesis. Thefrequency of these cancer susceptibility genes and their attributablecancer risk are important considerations in the development of a

4031

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 10: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Table 3 Assessment of causation by the Bradford-Hill criteria

Hypothesis: Dietary AFB," exposure can cause 249"' (AGG —>ACT) p53 mutations during human liver carcinogenesis

Strength of association•Consistency

•Positive dose-response correlation between estimated dietary AFB!exposure and the frequency of 249*" p53 mutations in three different

ethnic populations on three continents (179, 180, 206)•Specificity

•249s*' p53 mutant cells are observed in nontumorous liver in high HCC

incidence geographic areas (177)

Biological plausibility•AFB ! is a potent mutagen and carcinogen in laboratory studies (207-209)•AFB, is enzymatically activated by human hepatocytes (210, 211), and the

8,9-AFB, oxide binds to the third base (G) in codon 249 (212)•AFB, exposure to human liver cells (178, 184) in vitro produces codon 249"'

p53 mutations•249s" p53 expression inhibits apoptosis (213) and p53-mcdialcd transcription

(118) and enhances liver cell growth in vitro (214)'AFB,, aflatoxin B,.' HCC, hepatocellular carcinoma.

public health policy for genetic screening of the general population.Different public health and bioethical considerations apply to thegenetic screening of family members of individuals carrying a highcancer risk alíelein their germline (16).

Molecular Linkage between Carcinogen Exposure and Cancer

A number of specific p53 mutational hotspots have been recognizedin different types of human cancer (Fig. 8). The occurrence of anidentical mutation that is experimentally induced by a carcinogensupports a causative role of a specific environmental carcinogen incertain tumor types. Molecular linkage between exposure to carcinogens and cancer is best exemplified by the p53 mutational spectra ofhepatocellular carcinoma, skin cancer, and lung cancer. The mostcommon techniques used for p53 mutation analysis include PCR-based assays like single-strand conformational polymorphism, dena

turing gradient get electrophoresis, and DNA sequencing. Recently,another method based on yeast functional assays was developed todetect p53 mutations (174-176). In this assay, loss of DNA binding

and transcriptional transactivation function in mutant p53 is detectedby the colony color of the yeast. Measuring p53 mutation load or thefrequency of mutated alíeles in nontumorous tissue may indicateprevious carcinogen exposure and identify individuals at increasedcancer risk. However, the detection of rare cells with mutations in aproto-oncogene or tumor suppressor gene in normal-appearing human

tissue represents a challenging task. The average spontaneous mutation per base pair in human cells is estimated to be in the range of10~8 to 10~'°, and these frequencies increase only 10- to 1000-fold

on exposure to a mutagen. Therefore, methods that allow the detectionof few altered DNA sequences from IO5 to IO10 copies of the corre

sponding wild-type sequences in the presence of large quantities of

cellular DNA are required. The development of a highly sensitivegenotypic assay is allowing the detection of low-frequency mutationsin normal-appearing human tissues (177, 178). The detection of aparticular mutation in normal-appearing tissue provides further sup

port for the involvement of a specific carcinogen in a particular humancancer and may help identify individuals at increased cancer risk.

In liver tumors from persons living in geographic areas in whichaflatoxin B, and hepatitis B virus are cancer risk factors, most p53mutations are at the third nucleotide pair of codon 249 (179-182). A

dose-dependent relationship between dietary aflatoxin B, intake andcodon 249scr/>53 mutations is observed in hepatocellular carcinoma from

Asia, Africa, and North America (reviewed in Refs. 8 and 183). Themutation load of 249"" mutant cells in nontumorous liver also is posi

tively correlated with dietary aflatoxin B, exposure (177). Exposure ofaflatoxin B, to human liver cells in vitro produces 249"" (AGG to AGT)

p53 mutants (178, 184). These results indicate that expression of the249scr mutant p53 protein provides a specific growth and/or survival

advantage to liver cells (185) and are consistent with the hypothesis thatp53 mutations can occur early in liver carcinogenesis.

Sunlight exposure is a well-known risk factor for skin cancer.Tandem CC to TT transition mutations are frequently found in squa-

mous and basal cell skin carcinoma (186), whereas they are rarelyreported in other types of cancers (73). In vitro studies have shown theinduction of the characteristic CC to TT mutations by UV exposure(187-191). Sunlight-exposed normal skin and precancerous skin con

tain CC to TT tandem mutations (192,193). These results indicate thatCC to TT mutations induced by sunlight exposure may play a role inthe occurrence of skin cancer.

Cigarette smoking has been established as a major risk factor for theincidence of lung cancer. Codons 157,248, and 273 of \hep53 gene havebeen designated as mutational hotspots in lung cancer. The majority ofmutations found at these codons are G to T transversions. Furthermore, inaddition to lung cancer, codon 157 also constitutes one of the hotspots forG to T transversions in breast and head and neck cancers (Fig. 9). Insmoking-associated lung cancer, the occurrence of G to T transversions

has been linked to the presence of BP in cigarette smoke. Interestingly,codon 157 (GTC to TTC) mutations are not found in lung cancer fromnever-smokers (73, 194). A dose-dependent increase in p53 G to T

transversion mutations with cigarette smoking has been reported in lungcancer (127). Recently, it has been shown that BPDE, the metabolicallyactivated form of BP, binds to guanosine residues in codons 157,248, and273, which are mutational hotspots in lung cancer (119). Cigarette smokecondensate or BP also neoplastically transforms human bronchial epithelial cells in vitro (195).

Assessment of Causation by the Bradford-Hill Criteria

Results obtained from molecular epidemiological studies can beused for the assessment of causation. Using the "weight of the

Table 4 Assessment of causation by the Bradford-Hill criteria

Hypothesis: The chemical carcinogen BP in tobacco smoke can cause p53 hotspot mutations at codons 157, 248, and 273 in human lung carcinogenesis

Strength of association•Consistency

•Cigarette smoking is associated with a dose-response increase in p53mutations (G to T transversions) in human lung cancer (127)

•Specificity

•Codon 157 (GTC -> TTC) mutations are uncommon in other types of

cancer, including lung cancer in never smokers (73)•Temporality

•p53 mutations can be found in bronchial dysplasia (215-222)

Biological plausibility•Tobacco smoke and BP are mutagens (223-225)•BP is metabolically activated and forms BPDE DNA adducts in human

bronchus in vitro (75-fold interindividual variation; Refs. 226 and 227)•BP diol epoxide binds to Gs in codons 157, 248, and 273, which are p53

mutational•BP exposure to human cells in vitro produces codon 248 (COG —>CTG) p53

mutations (228)•Cigarette smoke condensâtes or BP can neoplastically transform human

bronchial epithelial cells in the laboratory (195, 229)

4032

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 11: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Table 5 Assessment of causation by the Bradford-Hill criteria

Hypothesis: Sunlight exposure can cause a characteristic CC to TT tandem double mutation in p53 in human skin cancer

Strength of association•Consistency

•Commonality of CC to TT tandem double mutation in squamous and basalcell skin carcinoma (186, 193, 230)

•Specificity•Tandem dipyrimidine CC to TT mutations are uncommon in other types of

cancer(73)•Temporality

•CC to TT mutations are found in normal and precancerous skin (193, 231)

Biological plausibility•UV is a mutagen and carcinogen in laboratory studies (232)•Cylobulane pyrimidine dimers and pyrimidine photoproducts are the two major adducts

produced by UV-irradiation and induce C to T at non-CpG sites and CC to TTmutations (189, 191)

•Skin cancer in xeroderma pigmentosum patients with defective nucleotide excisionrepair contains a high frequency of CC to TT mutations (233)

•UV exposure produces CC to TT mutations in studies using phage, bacteria, and mice(187-190)

evidence" principle, Bradford-Hill (196) proposed criteria in the

assessment of cancer causation including strength of association(consistency, specificity, and temporality) and biological plausibility. Three examples of weight of the evidence consistent withthe hypotheses include the following: (a) dietary aflatoxin B,exposure that can cause codon 249ser (AGG to AGT) p53 muta

tions during human liver carcinogenesis (Table 3); (b) the chemicalcarcinogen BP in tobacco smoke that can cause p53 hotspot mutations in human lung carcinogenesis (Table 4); and (c) sunlightexposure that can cause a characteristic CC to TT double mutationin p53 in human skin cancer (Table 5).

Furthermore, the influence of geographic location, race, and genderneeds to be taken into account when determining an individual's

susceptibility or risk for cancer (13). The p53 mutation spectra inbreast cancer from Japanese women show a different pattern whencompared with that of European and American women (197, 198).Even among African-American and Caucasian-American women, adifferent p53 mutational pattern in breast cancer is observed (199).The difference in mutation spectra and cancer incidence within apopulation or race could be due to either the effect of a regionalenvironmental agent or the polymorphic variation in genes responsible for carcinogen metabolism and DNA repair.

Comprehensive databases of tumor suppressor genes with mutations can be used as an important tool in molecular epidemiology.Examples of databases available on the Internet include the following:(a) for p53, http://www.iarc.fr/p53/home page.html; (b) for germlinemutations in Li-Fraumeni syndrome, http:cuni.cz/win/projects/readme.htm (71, 200); (c) for BRCA1 and BRCA2, http://www.nhgri.nih.gov/Intramural research/Lab transfer/Bic/; and (d) for APC,http://perso.curie.fr/Thierry.Soussi/p53database.html. We have previously discussed the limitations and advantages of the methodologiesto detect mutations and the need for improved epidemiológica!designand quality control (73). Further improvement of these databases byincluding information on geographical location, race, ethnicity, mutagen exposure, clinical course, genetic polymorphism, and so forthwill enhance the analyses of the mutation spectra of these genes.

Acknowledgments

We thank Mohammed Khan and Karman Raja for help with the mutationdatabase analysis and Dorothea Dudek for editorial and graphic assistance.

References

1. Doll, R. Nature and nurture: possibilities for cancer control. Carcinogenesis (Lond.),17: 177-184, 1996.

2. Couch, D. B. Carcinogenesis: basic principles. Drug Chem. Toxicol., 19: 133-148,1996.

3. Yuspa. S. H., and Shields. P. G. Etiology of cancer: chemical. In: V. T. DeVita, S.Hellman. and S. A. Rosenberg (eds.). Cancer: Principles and Practice of Oncology,pp. 185-202. Philadelphia: Lippincott-Raven Publishers, 1997.

4. Weston, A., and Harris, C. C. Chemical carcinogenesis. In: ¡.F. Holland. R. C. Bast,Jr., D. L. Morton, E. Frei III, D. W. Kufe, and R. R. Weichselbaum (eds.). CancerMedicine, pp. 261-276. Baltimore. MD: Williams & Wilkins, 1997.

5. National Research Council. Assessment of toxicology. In: National Academy ofSciences (ed.). Science and Judgement in Risk Assessment, pp. 56-67. Washington,

DC: National Academy Press. 1994.

6. Shields, P. G.. and Harris. C. C. Molecular epidemiology and the genetics ofenvironmental cancer. J. Am. Med. Assoc., 266- 681-687, 1991.

7. Perera, F. P.. and Santella. R. Carcinogenesis. In: P. Schulte and F. P. Perera (eds.).Molecular Epidemiology: Principles and Practices, pp. 277-300. New York: Aca

demic Press, 1993.8. Harris, C. C. The Walter Hubert Lecture. Molecular epidemiology of human cancer:

insights from the mutational analysis of the p53 tumor suppressor gene. Br. J.Cancer, 73: 261-269, 1996.

9. Perera. F. P. Environment and cancer: who are susceptible? Science (WashingtonDC), 278: 1068-1073, 1997.

10. Hunter, D., and Caporaso, N. Informed consent in epidemiologie studies involvinggenetic markers. Epidemiology. 8: 596-599, 1997.

11. Schulte, P. A., Hunter, D., and Rothman, N. Ethical and social issues in the use ofbiomarkers in epidemiological research. IARC Scientific Pubi., 142, 313-318. 1997.

12. Harris, C. C. Chemical and physical carcinogenesis: advances and perspectives forthe 1990s. Cancer Res., 51 (Suppl.): 5023s-5044s, 1991.

13. Perera. F. P. Molecular epidemiology: insights into cancer susceptibility, riskassessment, and prevention. J. Nati. Cancer Insl.. 88: 496-509. 1996.

14. Kinzler, K. W.. and Vogelstein. B. Gatekeepers and caretakers. Nature (Lond.), 386:761-763, 1997.

15. Ponder, B. Genetic testing for cancer risk. Science (Washington DC), 278: 1050-

1054. 1997.16. Li, F. P., Garber. J. E., Friend. S. H.. Strong. L. C.. Patenaude, A. F., Juengst. E. T.,

Reilly, P. R., Correa, P., and Fraumeni. J. F.. Jr. Recommendations on predictivetesting for germline p53 mutations among cancer-prone individuals. J. Nati. CancerInst., 84: 1156-1160, 1992.

17. Soskolne. C. L. Ethical, social, and legal issues surrounding studies of susceptiblepopulations and individuals. Environ. Health Perspect., 105 (Suppl. 4); 837-841,

1997.18. Hollstein. M., Sidransky. D.. Vogelstein, B., and Harris, C. C. p53 mutations in

human cancers. Science (Washington DC). 253: 49-53. 1991.

19. Levine. A. J., Momand. J.. and Finlay, C. A. The p53 tumour suppressor gene.Nature (Lond.). 351: 453-456, 1991.

20. Ponder, B. Cancer. Gene losses in human tumours. Nature (Lond.), 335: 400-402,

1988.21. Sherr, C. J. Cancer cell cycles. Science (Washington DC), 274: 1672-1677, 1996.

22. Fearon, E. R. Human cancer syndromes: clues to the origin and nature of cancer.Science (Washington DC). 278: 1043-1050. 1997.

23. Morin, P. J., Sparks. A. B., Korinek, V.. Barker. N.. Clevers. H.. Vogelstcin. B.. andKinzler. K. W. Activation of ß-catenin-Tcf signaling in colon cancer by mutationsin 0-catenin or APC. Science (Washington DC). 275: 1787-1790, 1997.

24. Kinzler. K. W., and Vogelstein, B. Lessons from hereditary colorectal cancer. Cell,87: 159-170, 1996.

25. Levy, D. B., Smith, K. J., Beazer-Barclay, Y., Hamilton. S. R., Vogelslein. B., andKinzler. K. W. Inactivation of both APC alíelesin human and mouse tumors. CancerRes.. 54: 5953-5958. 1994.

26. Jen, J.. Powell, S. M.. Papadopoulos. N.. Smith. K. J.. Hamilton. S. R., Vogelstein,B.. and Kinzler. K. W. Molecular determinants of dysplasia in colorectal lesions.Cancer Res., 54: 5523-5526. 1994.

27. Luongo, C., Moser, A. R., Gledhill. S.. and Dove. W. F. Loss of A/>r+ in intestinal

adenomas from Min mice. Cancer Res., 54: 5947-5952. 1994.

28. Prolla, T. A., Abuin, A., and Bradley. A. DNA mismatch repair deficient mice incancer research. Semin. Cancer Biol.. 7: 241-247, 1996.

29. de Wind, N., Dekker, M.. Berns. A.. Radman. M.. and te Riele, H. Inactivation ofthe mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance,hyperrecombination. and predisposition to cancer. Cell. 82: 321-330. 1995.

30. Reitmair, A. H.. Schmils. R.. Ewel, A.. Bapat. B., Redston. M., Mitri. A.. Water-

house, P., Mittrucker, H. W., Wakeham. A., and Liu. B. MSH2 deficient mice areviable and susceptible to lymphoid tumours. Nat. Genet., //: 64-70. 1995.

31. Loeb, L. A. Mutator phenotype may be required for multistage carcinogenesis.Cancer Res., 5/: 3075-3080, 1991.

32. Loeb. L. A. Cancer cells exhibit a mutator phenotype. Adv. Cancer Res., 72: 25-56,

1998.33. Aaltonen. L. A., Peltomaki. P.. Leach, F. S.. Sistonen. P.. Pylkkanen. L.. Mecklin.

J. P., Jarvinen, H., Powell. S. M., Jen. J.. Hamilton. S. R.. Peterson. G. M.. Kinzler.K. W., Vogelstein, B., and de la Chapelle, A. Clues to the pathogenesis of familialcolorectal cancer. Science (Washington DC). 260: 812-815, 1993.

34. lonov. Y., Peinado. M. A., Malkhosyan. S.. Shibata. D., and Perucho, M. Ubiquitoussomatic mutations in simple repeated sequences reveal a new mechanism for coloniecarcinogenesis. Nature (Lond.). 363: 558-561, 1993.

4033

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 12: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

35. Thibodeau, S. N., Bren, G., and Schaid. D. Microsatellite instability in cancer of the 58.proximal colon. Science (Washington DC), 260: 816-819, 1993.

36. Fishel. R., Lescoe, M. K.. Rao, M. R., Copeland, N. G.. Jenkins. N. A., Garber, J.,Kane. M.. and Kolodner. R. The human mutator gene homolog MSH2 and its 59.association with hereditary nonpolyposis colon cancer. Cell, 75: 1027-1038, 1993.

37. Leach, F. S., Nicolaides, N. C., Papadopoulos, N., Liu, B., Jen, J., Parsons, R., 60.Peltomaki, P.. Sistonen, P., Aaltonen, L. A., Nyslrom-Lahti, M., Guan, X. Y., Zhang,

J., Meltzer. P. S., Yu, J. W., Kao, F. T.. Chen. D. J., Cerosaletti. K. M.. Fournier, 61.R. E., Todd. S., Lewis, T.. Leach. R. J., Naylor. S. L., Weissenbach. J., Mecklin,J. P.. Jarvinen. H., Petersen, G. M.. Hamilton, S. R., Green, J., Jass. J.. Watson, P..Lynch, H. T., Trent. J. M., de la Chapelle, A., Kinzler. K. W., and Vogelslein, B. 62.Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell, 75:1215-1235, 1993.

38. Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., Ruben, S. M., Carter. K. C., Rosen, 63.C. A., Haseltine, W. A., Fleischmann, R. D., Fraser, C. M., Adams, M. D., Venter,J. C.. Hamilton, S. R., Petersen, G. M., Watson, P.. Lynch, H. T.. Peltomaki, P..Mecklin. J. P., de la Chapelle. A.. Kinzler, K. W., and Vogelstein. B. Mutation of a 64.mutL homolog in hereditary colon cancer. Science (Washington DC), 263: 1625-

1629. 1994.39. Nicolaides, N. C., Papadopoulos, N., Liu, B., Wei, Y. F., Carter, K. C., Rüben,S. M., 65.

Rosen, C. A., Haseltine, W. A., Fleischmann, R. D., and Fräser,C. M. Mutations oftwo PMS homologues in hereditary nonpolyposis colon cancer. Nature (Lond.), 371:75-80, 1994. 66.

40. Andrew, S. E.. McKinnon, M., Cheng, B. S., Francis, A., Penney, J., Reitmair. A. H..Mak, T. W.. and Jirik, F. R. Tissues of MSH2-deficient mice demonstrate hyper-

mutability on exposure to a DNA methylating agent. Proc. Nati. Acad. Sci. USA, 95:1126-1130, 1998. 67.

41. Miyaki. M., Konishi, M., Tanaka, K., Kikuchi-Yanoshila, R., Muraoka, M.. Yasuno,

M., Igari, T., Koike, M., Chiba, M., and Mori, T. Germline mutation of MSH6 as the 68.cause of hereditary nonpolyposis colorectal cancer. Nat. Genet., 17: 271-272, 1997.

42. Edelmann, W.. Yang. K.. Umar, A.. Heyer. J.. Lau. K.. Fan, K.. Liedtke, W., Cohen, 69.P. E., Kane, M. F., Lipford, J. R., Yu, N., Crouse. G. F., Pollard, J. W., Kunkel, T.,Kipkin, M., Kolodner, R.. and Kucherlapati. R. Mutation in the mismatch repairgene Msh6 causes cancer susceptibility. Cell, 91: 467-477, 1997. 70.

43. Livingstone, L. R., White. A., Sprouse, J., Livanos, E., Jacks, T., and Tlsty, T. D.Altered cell cycle arrest and gene amplification potential accompany loss of wild-type p53. Cell, 70: 923-935, 1992. 71.

44. Yin. Y., Tainsky, M. A.. Bischoff, F. Z., Strong, L. C., and Wahl, G. M. Wild-type

p53 restores cell cycle control and inhibits gene amplification in cells with mutantp53 alíeles.Cell, 70: 937-948. 1992.

45. Wang. X. W.. Yeh, H., Schaeffer, L., Roy, R., Moncollin, V., Egly, J. M.. Wang, Z., 72.Freidberg, E. C., Evans, M. K., Taffe, B. G., Bohr, V. A.. Weeda, G., Hoeijmakers,J. H.. Forrester, K., and Harris, C. C. p53 modulation of TFIIH-associated nucleotide 73.excision repair activity. Nat. Genet., 10: 188-195, 1995.

46. Ford, J. M., and Hanawalt, P. C. Li-Fraumeni syndrome fibroblasts homozygous forp53 mutations are deficient in global DNA repair but exhibit normal transcription- 74.coupled repair and enhanced UV-resistance. Proc. Nati. Acad. Sci. USA, 92:8876-8880. 1995.

47. Smith, M. L., Chen, I. T., Zhan, Q., O'Connor. P. M., and Fornace, A. J., Jr. 75.

Involvement of the p53 tumor suppressor in repair of UV-type DNA damage.Oncogene, 10: 1053-1059, 1995.

48. Mirzayans, R., Enns, L., Dietrich, K., Barley, R. D.. and Paterson, M. C. Faulty 76.DNA polymerase a/e-mediated excision repair in response to y radiation or ultraviolet light in p53-deficient fibroblast strains from affected members of a cancer- 77.prone family with Li-Fraumeni syndrome. Carcinogenesis (Lond.), 17: 691-698,

1996. 78.49. Ford, J. M.. and Hanawalt. P. C. Expression of wild-type p53 is required for efficient

global genomic nucleotide excision repair in UV-irradiated human fibroblasts. 79.J. Biol. Chem., 272: 28073-28080, 1997.

50. Ford, J. M., Baron, E. L., and Hanawalt, P. C. Human fibroblasts expressing the 80.human papillomavirus £6gene are deficient in global genomic nucleotide excisionrepair and sensitive to ultraviolet irradiation. Cancer Res., 58: 599-603, 1998. 81.

51. Prost. S., Bellamy, C. O., Cunningham, D. S., and Harrison, D. J. Altered DNArepair and dysregulation of p53 in IRF-1 null hepatocytes. FASEB J., 12: 181-188,

1998. 82.52. Mekeel, K. L., Tang, W.. Kachnic, L. A., Luo, C. M., DeFrank, J. S., and Powell,

S. N. Inaclivation of p53 results in high rates of homologous recombinalion.Oncogene, 14: 1847-1857, 1997. 83.

53. Meyn, M. S.. Strasfeld, L., and Allen, C. Testing the role of p53 in the expressionof genetic instability and apoptosis in ataxia-telangiectasia. Int. J. Radiât.Biol., 66 84.(Suppl.): S141-S149. 1994.

54. Fukasawa, K., Choi, T., Kuriyama, R., Rulong, S., and Vande Woude, G. F.Abnormal centrosome amplification in the absence of p53. Science (Washington 85.DC), 271: 1744-1747, 1996.

55. Kawashima, K., Mihara, K., Usuki, H., Shimtzu, N., and Namba, M. Transfected 86.mutant p53 gene increases X-ray-induced cell killing and mutation in humanfibroblasts immortalized with 4-nitroquinoline 1-oxide but does not induce neoplas-tic transformation of the cells. Int. J. Cancer, 61: 76-79, 1995. 87.

56. Xia, F., Wang. X., Wang. Y. H.. Tsang, N. M., Yandell. D. W., Kelsey, K. T., andLiber, H. L. Altered p53 status correlates with differences in sensitivity to radiation- 88.

induced mutation and apoptosis in two closely related human lymphoblast lines.Cancer Res., 55: 12-15, 1995.

57. Honma. M.. Hayashi, M., and Sofuni, T. Cytotoxic and mutagenic responses to 89.X-rays and chemical mutagens in normal and p53-mutated human lymphoblastoidcells. Mutât.Res.. 374: 89-98, 1997.

4034

Liu, P. K., Kraus, E., Wu, T. A., Strong. L. C.. and Tainsky. M. A. Analysis ofgenomic instability in Li-Fraumeni fibroblasts with germline p53 mutations. Oncogene, 12: 2267-2278. 1996.Strauss, B. S. Silent and multiple mutations in p53 and the question of the hyper-mutability of tumors. Carcinogenesis (Lond.), 18: 1445-1452. 1997.

Fishel, R. Genomic instability, mutators, and the development of cancer: is there arole for p53? J. Nati. Cancer Inst., 88: 1608-1609, 1996.Kahlenberg. M. S., Stoler, D. L., Basik, M., Petrelli, N. J., Rodriguez-Bigas, M.. andAnderson, G. R. p53 tumor suppressor gene status and the degree of genomicinstability in sporadic colorectal cancers. J. Nati. Cancer Inst., 88: 1665-1670, 19%.

Renault, B., Calistri, D., Buonsanti, G.. Nanni, O.. Amadori. D., and Ranzani, G. N.Microsatellite instability and mutations of p53 and TGF-ßRII genes in gastriccancer. Hum. Genet., 98: 601-607, 1996.

Sands, A. T., Suraokar, M. B., Sanchez, A., Marth, J. E., Donehower, L. A., andBradley, A. p53 deficiency does not affect the accumulation of point mutations in aIransgene target. Proc. Nati. Acad. Sci. USA, 92: 8517-8521, 1995.

Vijg, J., Mullaart. E., Van der Schans. G. P., Lohman, P. H.. and Kncok, D. L.Kinetics of ultraviolet induced DNA excision repair in rat and human fibroblasts.Mutât.Res., 132: 129-138, 1984.

van Loon, A. A., Timmerman, A. J., Van der Schans, G. P., Lohman, P. H., andBaan, R. A. Different repair kinetics of radiation-induced DNA lesions in human andmurine white blood cells. Carcinogenesis (Lond.), 13: 457-462, 1992.

Vrieling, H., Venema, J., van Rooyen, M. L., van Hoffen, A., Menichini, P..Zdzienicka, M. Z., Simons, J. W., Mullenders, L. H., and Van Zeeland, A. A. Strandspecificity for UV-induced DNA repair and mutations in the Chinese hamster HPRTgene. Nucleic Acids Res., 19: 2411-2415, 1991.Lane, D. P., and Benchimol, S. p53: oncogene or anti-oncogene. Genes Dev., 4: 1-8,

1990.Dittmer, D., Pâti,S., Zambetti, G., Chu, S., Teresky, A. K., Moore, M., Finlay, C.,and Levine, A. J. Gain of function mutations in p53. Nat. Genet.. 4: 42-46, 1993.

Hsiao. M., Low, J.. Dorn. E.. Ku, D., Pattengale. P.. Yeargin. J.. and Haas, M.Gain-of-function mutations of the p53 gene induce lymphohematopoietic metastaticpotential and tissue invasiveness. Am. J. Pathol.. 145: 702-714, 1994.

Hollstein. M., Shomer. B.. Greenblatt, M., Soussi, T., Hovig, E.. Montesano, R., andHarris, C. C. Somatic point mutations in the p53 gene of human tumors and celllines: updated compilation. Nucleic Acids Res., 24: 141-146, 1996.Hainaut, P., Hernandez, T., Robinson, A., Rodriguez-Tome, P., Flores, T., Hollstein,

M.. Harris, C. C., and Montesano, R. IARC Database of p53 gene mutations inhuman tumors and cell lines: updated compilation, revised formats and new visualisation tools. Nucleic Acids Res., 26: 205-213, 1998.

Soussi, T., Carónde Fromentel, C., and May, P. Structural aspects of the p53 proteinin relation to gene evolution. Oncogene. 5: 945-952, 1990.

Greenblatt, M. S., Bennett, W. P., Hollstein, M., and Harris, C. C. Mutations in thep53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis.Cancer Res., 54: 4855-4878, 1994.

Greenblatt, M. S., Grollman, A. P., and Harris, C. C. Deletions and insertions in thep53 tumor suppressor gene in human cancers: confirmation of the DNA polymeraseslippage/misalignment model. Cancer Res.. 56: 2130-2136, 1996.Park, J. W., Cundy, K. C., and Ames, B. N. Detection of DNA adducts byhigh-performance liquid chromatography with electrochemical detection. Carcinogenesis (Lond.), 10: 827-832, 1989.

Gottlieb, T. M., and Oren, M. p53 in growth control and neoplasia. Biochim.Biophys. Acta, /287: 77-102, 1996.

Harris, C. C. p53: at the crossroads of molecular Carcinogenesis and cancer riskassessment. Science (Washington DC), 262: 1980-1981, 1993.

Soussi, T., Legras, Y.. Lubin, R., Ory, K., and Schlichtholz. B. Multifactorialanalysis of p53 alteration in human cancer: a review. Int. J. Cancer. 57: 1-9, 1994.Kunkel, T. A. Misalignment-mediated DNA synthesis errors. Biochemistry, 29:8003-8011, 1990.

Jego, N., Thomas, G., and Hamelin, R. Short direct repeats flanking deletions, andduplicating insertions in p53 gene in human cancers. Oncogene, 8: 209-213, 1993.

Krawczak, M., and Cooper, D. N. Gene deletions causing human genetic disease:mechanisms of mutagenesis and the role of the local DNA sequence environment.Hum. Genet., 86: 425-441, 1991.

Liu, X., Miller, C. W., Koeffler, P. H., and Berk, A. J. The p53 activation domainbinds the TATA box-binding polypeptide in HoIo-TFIID, and a neighboring p53domain inhibits transcription. Mol. Cell. Biol., 13: 3291-3300, 1993.Vogelstein, B., and Kinzler, K. W. p53 function and dysfunction. Cell, 70: 523-526,

1992.Thut, C. J., Chen, J-L., Klemm, R., and Tjian, R. p53 transcriptional activation

mediated by coactivators TAFÜ40and TAFÜ60.Science (Washington DC), 267:100-104, 1995.

Lu, H., and Levine, A. J. Human TAFII31 protein is a transcriptional coactivator ofthe p53 protein. Proc. Nati. Acad. Sci. USA, 92: 5154-5158, 1995.Raycroft, L., Wu, H., and Lozano, G. Transcriptional activation by wild-type but nottransforming mutants of the p53 anti-oncogene. Science (Washington DC), 249:1049-1051, 1990.

Fields, S., and Jang, S. K. Presence of a potent transcription activating sequence inthe p53 protein. Science (Washington DC), 249: 1046-1048. 1990.Seto, E., Usheva, A., Zambetti, G. P., Momand, J., Horikoshi, N., Weinmann, R.,Levine, A. J., and Shenk, T. Wild-type p53 binds to the TATA-binding protein andrepresses transcription. Proc. Nati. Acad. Sci. USA, 89: 12028-12032, 1992.

Truant, R., Xiao, H., Ingles, C. J., and Greenblatt, J. Direct interaction between thetranscriptional activation domain of human p53 and the TATA box-binding protein.J. Biol. Chem., 268: 2284-2287. 1993.

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 13: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

90. Martin, D. W., Muñoz, R. M., Subler, M. A., and Deb, S. p53 binds to the 117.TATA-binding protein-TATA complex. J. Biol. Chem., 268.- 13062-13067, 1993.

91. Mack, D. H., Vartikar, J., Pipas, J. M., and Laimins, L. A. Specific repression ofTATA-mediated but not initiator-mediated transcription by wild-type p53. Nature 118.(Lond.), 363: 281-283, 1993.

92. Lin, J., Chen, J., Elenbaas, B., and Levine, A. J. Several Hydrophobie amino acidsin the p53 amino-terminal domain are required for transcriptional activation, binding 119.to mdm-2 and the adenovirus 5 El B 55-kD protein. Genes Dev., 8: 1235-1246,

1994.93. Clore, G. M., Omichinski, J. G., Sakaguchi, K., Zambrano, N., Sakamoto, H., 120.

Appella, E., and Gronenborn, A. M. High-resolution solution structure of theoligomerization domain of p53 by multi-dimensional NMR. Science (WashingtonDC), 265: 386-391, 1994. 121.

94. Jeffrey, P. D., Gorina, S., and Pavletich, N. P. Crystal structure of the tetrameri/illunidomain of the p53 tumor suppressor at 1.7 angstroms. Science (Washington DC),267: 1498-1502, 1995. 122.

95. Hupp, T. R., and Lane, D. P. Allosteric activation of latent p53 tetramere. Curi.Biol., 4: 865-875, 1995. 123.

96. Lee, W., Harvey, T. S., Yin, Y., Yau, P., Litchfield, D., and Array/smith, C. H.Solution structure of the tetrameric minimum transforming domain of p53. Nat. 124.Struct. Biol., /: 877-890, 1994.

97. Jayaraman, L., and Prives, C. Activation of p53 sequence-specific DNA binding byshort single strands of DNA requires the p53 C-terminus. Cell, 81: 1021-1029, 125.

1995.98. Bakalkin, G., Yakovleva, T., Selivanova, G., Magnusson, K. P., Szekely, L.,

Kiseleva, E., Klein, G., Terenius, L., and Wiman, K. G. p53 binds single-stranded 126.

DNA ends and catalyzes DNA renaturation and strand transfer. Proc. Nati. Acad.Sci. USA, 91: 413-417, 1994. 127.

99. Horikoshi, N., Usheva, A., Chen, J., Levine, A. J., Weinmann, R., and Shenk, T.Two domains of p53 interact with the TATA-binding protein, and the adenovirus13S EIA protein disrupts the association, relieving p53-mediated transcriptional 128.repression. Mol. Cell. Biol., IS: 227-234, 1995.

100. Wang, X. W., Vermeulen, W., Coursen, J. D., Gibson, M., Lupold, S. E., Forrester, 129.K., Xu, G., Elmore, L., Yeh, H., Hoeijmakers, J. H. J., and Harris, C. C. The XPBand XPD helicases are components of the p53-mediated apoptosis pathway. GenesDev., 10: 1219-1232, 1996. 130.

101. Milner, J., Medcalf, E. A., and Cook, A. C. Tumor suppressor p53: analysis ofwild-type and mutant p53 complexes. Mol. Cell. Biol., //.•12-19, 1991. 131.

102. Brain, R., and Jenkins, J. R. Human p53 directs DNA strand reassociation and isphotolabelled by 8-azido ATP. Oncogene, 9: 1775-1780, 1994. 132.

103. Wang, Y., and Prives, C. Increased and altered DNA binding of human p53 by S andG2/M but not G, cyclin-dependent kinases. Nature (Lond.), 376: 88-91, 1995.

104. Wu, L., Bayle, J. H., Elenbaas, B., Pavletich, N. P., and Levine, A. J. Alternatively 133.spliced forms in the carboxy-terminal domain of the p53 protein regulate its ability

to promote annealing of complementary single strands of nucleic acids. Mol. Cell.Biol., 15: 497-504, 1995. 134.

105. Bakalkin, G., Selivanova, G., Yakovleva, T., Kiseleva, E., Kashuba, E., Magnusson,K. P., Szekely, L., Klein, G., Terenius, L., and Wiman, K. G. p53 binds single-stranded DNA ends through the C-terminal domain and internal DNA segments viathe middle domain. Nucleic Acids Res., 23: 362-369, 1995. 135.

106. Mayr, G. A., Reed, M., Wang, P., Wang, Y., Schwedes, J. F., and Tegtmeyer, P.Serine phosphorylation in the NH2 terminus of p53 facilitates transactivation.Cancer Res., 55: 2410-2417, 1995.

107. Hainaut, P., and Milner, J. Redox modulation of p53 conformation and sequence- 136.specific DNA binding in vitro. Cancer Res., 53: 4469-4473, 1993.

108. Hupp, T. R., Meek, D. W., Midgley, C. A., and Lane, D. P. Activation of the crypticDNA binding function of mutant forms of p53. Nucleic Acids Res., 21: 3167-3174, 137.

1993.109. Rainwater, R., Parks, D., Anderson, M. E., Tegtmeyer, P., and Mann, K. Role of

cysteine residues in regulation of p53 function. Mol. Cell. Biol., 15: 3892-3903, 138.

1995.110. Gu, W., and Roeder, R. G. Activation of p53 sequence-specific DNA binding by

acetylation of the p53 C-terminal domain. Cell, 90: 595-606, 1997.

111. Shaw, P., Freeman, J., Bovey, R., and Iggo, R. Regulation of specific DNA binding 139.by p53: evidence for a role for 0-glycosylation and charged residues at the carboxyterminus. Oncogene, 12: 921-930, 1996. 140.

112. Okorokov, A. L., Ponchel, F., and Milner, J. Induced N- and C-terminal cleavage of

p53: acore fragment of p53, generated by interaction with damaged DNA, promotes 141.cleavage of the N terminus of full-length p53, whereas ssDNA induces C-terminalcleavage of p53. EMBO J., 16: 6008-6017, 1997.

113. Cho, Y., Gorina, S., Jeffrey, P., and Pavletich, N. P. Crystal structure of a p53 tumorsuppressor-DNA complex: a framework for understanding how mutations inactivate 142.p53. Science (Washington DC), 265: 346-355, 1994.

114. Borresen, A. L., Andersen, T. I., Eyfjord, J. E., Comelis, R. S., Thorlacius, S., Borg, 143.A., Johansson, U., Theillet, C., Schemeck, S., and Hartman, S. TP53 mutations andbreast cancer prognosis: particularly poor survival rates for cases with mutations inthe zinc-binding domains. Genes Chromosomes Cancer, 14: 71-75, 1995. 144.

115. Aas, T., Borresen, A. L., Geisler, S., Smith-Sorensen, B., Johnsen, H., Varhaug,

J. E., Akslen, L. A., and Lonning, P. E. Specific P53 mutations are associated withde novo resistance to doxorubicin in breast cancer patients. Nat. Med., 2: 811-814,

1996. 145.116. Taylor, E. M., Broughton, B. C., Botta, E., Stefanini, M., Sarasin, A., Jaspers, N. G.,

Fawcett, H., Harcourt, S. A., Arieti, C. F., and Lehmann, A. R. Xeroderma pigmen-

tosum and trichothiodystrophy are associated with different mutations in the XPD(ERCC2) repair/transcription gene. Proc. Nati. Acad. Sci. USA, 94: 8658-8663,1997.

4035

Ehrlich, M., Zhang. X. Y., and Inamdar, N. M. Spontaneous deamination of cytosineand 5-methylcytosine residues in DNA and replacement of 5-melhylcytosine residues with cytosine residues. Mutât.Res., 238: 277-286, 1990.

Forrester, K., Lupold, S. E., On, V. L., Chay, C. H., Band, V., Wang, X. W., andHarris, C. C. Effects of p53 mutants on wild-type p53-mediated transactivation arecell type dependent. Oncogene, 10: 2103-2111. 1995.Denissenko, M. F., Pao, A., Tang, M., and Pfeifer, G. P. Preferential formation ofbenzo(fl)pyrene adducts at lung cancer mutational hotspots in P53. Science (Washington DC), 274: 430-432, 1996.

Denissenko, M. F., Chen, J. X., Tang, M. S., and Pfeifer, G. P. Cytosine methylationdetermines hotspots of DNA damage in the human P53 gene. Proc. Nati. Acad. Sci.USA, 94: 3893-3898, 1997.

Chen, J. X., Zheng, Y., West, M., and Tang, M. S. Carcinogens preferentially bindat methylated CpG in the p53 mutational hotspots. Cancer Res.. 58: 2070-2075,1998.Tornaletti, S., and Pfeifer, G. P. Slow repair of pyrimidine dimers at p53 mutationhotspots in skin cancer. Science (Washington DC), 263: 1436-1438, 1994.Hanawalt, P. C. Preferential DNA repair in expressed genes. Environ. HealthPerspect., 76: 9-14, 1987.

Ford, J. M., Lommel, L., and Hanawalt, P. C. Preferential repair of ultravioletlight-induced DNA damage in the transcribed strand of the human p53 gene. Mol.Carcinog., 10: 105-109, 1994.

Evans, M. K., Taffe, B. G., Harris, C. C., and Bohr, V. A. DNA strand bias in therepair of the p53 gene in normal human and xeroderma pigmentosum group Cfibroblasts. Cancer Res., 53: 5377-5381, 1993.

Hanawalt, P. C. Preferential repair of damage in actively transcribed DNA sequences in vivo. Genome, 31: 605-611, 1989.

Takeshima, Y., Seyama, T., Bennett, W. P., Akiyama, M., Tokuoka, S., Inai, K.,Mabuchi, K., Land, C. E., and Harris, C. C. p53 mutations in lung cancers fromnon-smoking atomic-bomb survivors. Lancet, 342: 1520-1521, 1993.Bohr. V. A. Gene-specific DNA repair. Carcinogenesis (Lond.), 12: 1983-1992.

1991.Ruggeri, B., DiRado, M., Zhang, S. Y., Bauer, B., Goodrow, T., and Klein-Szanto.A. J. Benzo(a)pyrene-¡nduced murine skin tumors exhibit frequent and characteristicG to T mutations in the p53 gene. Proc. Nati. Acad. Sci. USA, 90: 1013-1017, 1993.

Bohr, V. A., Phillips. D. H., and Hanawalt, P. C. Heterogeneous DNA damage andrepair in the mammalian genome. Cancer Res., 47: 6426-6436, 1987.Holliday, R., and Grigg, G. W. DNA methylation and mutation. Mutât.Res., 285:61-67, 1993.Rideout, W. M., Ill, Coetzee, G. A., Olumi, A. F., and Jones, P. A. 5-Methylcytosine

as an endogenous mutagen in the human LDL receptor and p53 genes. Science(Washington DC), 249: 1288-1290, 1990.Mellon, I., Rajpal, D. K., Koi, M., Boland. C. R.. and Champe, G. N. Transcription-

coupled repair deficiency and mutations in human mismatch repair genes. Science(Washington DC), 272: 557-560, 1996.

Okamoto, A., Demetrick, D. J., Spillare, E. A., Hagiwara, K., Hussain, S. P.,Bennett, W. P., Forrester, K., Gerwin, B., Serrano, M., Beach, D. H., and Harris,C. C. Mutations and altered expression of genes regulating the cell cycle G,checkpoint in human cancer. Proc. Nati. Acad. Sci. USA, 91: 11045-11049, 1994.Kamb, A., Gruis, N. A., Weaver-Feldhaus, J., Liu, Q., Harshman, K., Tavtigian,

S. V., Stocken, E„Day, R. S., Johnson, B. E., and Skolnick, M. H. A cell cycleregulator potentially involved in genesis of many tumor types. Science (WashingtonDC), 264: 436-440, 1994.

Nobori, T., Miura, K., Wu, D. J., Lois, A., Takabayashi, K., and Carson, D. A.Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature (Lond.), J68: 753-756, 1994.

Quesnel, B., Fenaux, P., Philippe, N., Foumier, J., Bonneterre. J., Preudhomme, C.,and Peyral, J. P. Analysis of pl6 gene deletion and point mutation in breastcarcinoma. Br. J. Cancer, 72: 351-353, 1995.Okamoto, A., Hussain, S. P., Hagiwara, K., Spillare, E. A., Rusin, M. R., Demetrick,D. J., Serrano, M., Hannon, G. J., Shiseki, M., and /armala. M. Mutations in thepl6'NK4/MTSl/CDKN2, pl5WK4B/MTS2, and P18 genes in primary and metastatic

lung cancer. Cancer Res., 55: 1448-1451, 1995.Smith-Sorensen, B., and Hovig, E. CDKN2A (pl6INK4A) somatic and germline

mutations. Hum. Mutât.,7: 294-303, 19%.

Foulkes, W. D., Flanders, T. Y., Pollock, P. M., and Hayward, N. K. The CDKN2A(pl6) gene and human cancer. Mol. Med., 3: 5-20, 1997.

Herman, J. G., Merlo, A., Mao, L., Lapidus, R. G., Issa, J. P., Davidson, N. E.,Sidransky, D., and Baylin, S. B. Inactivation of the CDKN2/pl6/MTSl gene isfrequently associated with aberrant DNA methylation in all common human cancers.Cancer Res., 55: 4525-4530, 1995.Issa, J. P., and Baylin, S. B. Epigenetics and human disease. Nat. Med., 2: 281-282,

1996.Yarbrough, W. G., Aprelikova, O., Pei, H., Olshan, A. F., and Liu, E. T. Familialtumor syndrome associated with a germline nonfunctional p!6/NK4a alíele.J. Nati.

Cancer Inst., 88: 1489-1491, 1996.

Ranade, K., Hussussian, C. J., Sikorski, R. S., Varmus, H. E., Goldstein. A. M..Tucker, M. A., Serrano, M., Hannon, G. J., Beach, D., and Dracopoli. N. C.Mutations associated with familial melanoma impair pl6INK4 function. Nat. Genet.,

10: 114-116, 1995.Miki, Y., Swensen, J., Shattuck-Eidens, D., Futreal, P. A., Harshman. K., Tavtigian,

S., Liu, Q., Cochran, C., Bennett, L. M., Ding, W., Bell, R., Rosenthal, J.. Hussey,C., Tran, T., McClure, M., Frye, C., Hattier, T., Phelps, R., Haugen-Strano, A.,

Katcher, H., Yakumo, K., Gholami, Z., Shaffer, D., Stone, S., Bayer, S., Wray, C.,Hogden, R., Dayananth, P., Ward, J., Tonin, P., Narod, S., Bristow, P. K., Noms,F. H., Helvering, L., Morrison. P., Rosteck, P., Lai, M., Barrett, J. C., Lewis, C.,

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 14: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

Neuhausen, S., Cannon-Albright, L., Goldgar, D., Wiseman, R., Kanib. A., and

Skolnick, M. H. A strong candidate for the breast and ovarian cancer susceptibilitygene BRCAI. Science (Washington DC), 266; 66-71, 1994.

146. Wooster, R., Bignell, G., Lancaster, J., Swift, S., Seal, S., Mangion, J., Collins, N.,Gregory, S., Gumbs, C, and Micklem, G. Identification of the breast cancersusceptibility gene BRCA2. Nature (Lond.), 378: 789-792, 1995.

147. Tavtigian, S. V., Simard, J., Rommens, J., Couch, F., Shattuck-Eidens, D.,Neuhausen, S., Merajver, S.. Thorlacius, S., Offit, K., Stoppa-Lyonnet, D., Belanger,

C., Bell, R., Berry, S., Bogden, R., Chen, Q., Davis, T., Dumont, M., Frye, C.,Hattier, T., Jammulapati, S., Janecki, T., Jiang, P., Kehrer, R., Leblanc, J. F., andGoldgar, D. E. The complete BRCA2 gene and mutations in chromosome 13q-linkedkindreds. Nat. Genet., 12: 333-337, 1996.

148. Cannon-Albright, L. A., Kamb, A., and Skolnick, M. A review of inherited predisposition to melanoma. Semin. Oncol., 23: 667-672, 1996.

149. Kamb, A., and Skolnick, M. H. Identification of the BRCAI breast cancer gene andits clinical implications. Important Adv. Oncol., 23-35, 1996.

150. Stratton, M. R., and Wooster, R. Hereditary predisposition to breast cancer. Curr.Opin. Genet. Dev., 6: 93-97, 1996.

151. Gayther, S. A., and Ponder, B. A. Mutations of the BRCAI and BRCA2 genes andthe possibilities for predictive testing. Mol. Med. Today, 3: 168-174, 1997.

152. Easton, D. F., Ford, D., and Bishop, D. T. Breast and ovarian cancer incidence inBRCAI mutation carriers. Breast Cancer Linkage Consortium. Am. J. Hum. Genet.,56: 265-271, 1995.

153. Gayther, S. A., Warren, W., Mazoyer, S., Russell, P. A., Harrington, P. A., Chiano,M., Seal, S., Hamoudi. R., van Rensburg, E. J., and Dunning, A. M. Germlinemutations of the BRCAI gene in breast and ovarian cancer families provide evidencefor a genotype-phenotype correlation. Nat. Genet., 11: 428-433, 1995.

154. Gayther, S. A., Mangion, J., Russell, P., Seal, S., Barfoot, R., Ponder, B. A., Stratton,M. R., and Easton, D. Variation of risks of breast and ovarian cancer associated withdifferent germline mutations of the BRCA2 gene. Nat. Genet., 15: 103-105, 1997.

155. Schutte, M., da Costa, L. T., Hahn, S. A., Moskaluk, C., Hoque, A. T., Rozenblum,E., Weinstein, C. L., Bittner, M., Meltzer, P. S., and Trent, J. M. Identification byrepresentational difference analysis of a homozygous deletion in pancreatic carcinoma that lies within the BRCA2 region. Proc. Nati. Acad. Sci. USA, 92: 5950-

5954, 1995.156. Goggins, M., Schutte, M., Lu, J., Moskaluk, C. A., Weinstein, C. L., Petersen, G. M.,

Yeo, C. J., Jackson, C. E., Lynch, H. T.. Hruban, R. H., and Kem, S. E. GermlineBRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas.Cancer Res., 56: 5360-5364, 1996.

157. Friedman, L. S., Gayther, S. A., Kurosaki, T., Gordon, D., Noble, B., Casey, G.,Ponder, B. A., and Anton-Culver, H. Mutation analysis of BRCAI and BRCA2 ina male breast cancer population. Am. J. Hum. Genet., 60: 313-319, 1997.

158. Futreal, P. A., Liu, Q., Shattuck-Eidens, D., Cochran, C., Harshman, K., Tavtigian,S., Bennett, L. M., Haugen-Strano, A., Swensen, J., Miki, Y., Eddington, K.,McClure, M., Frye, C., Weaver-Feldhaus, J., Ding, W., Gholami, Z., Soderkvist, P.,

Terry, L., Jhanwar, S., Berchuck, A., Inglehart, J. D., Marks, J., Ballinger, D. G.,Barrett, J. C., Skolnick, M. H., Kamb, A., and Wiseman, R. BRCAI mutations inprimary breast and ovarian carcinomas. Science (Washington DC), 266.- 120-122,

1994.159. Merajver, S. D., Pham, T. M., Caduff, R. F., Chen, M., Poy, E. L., Cooney, K. A.,

Weber, B. L., Collins, F. S., Johnston, C., and Frank, T. S. Somatic mutations in theBRCAI gene in sporadic ovarian tumours. Nat. Genet., 9: 439-443, 1995.

160. Miki, Y., Katagiri, T., Kasumi, F., Yoshimoto, T., and Nakamura, Y. Mutationanalysis in the BRCA2 gene in primary breast cancers. Nat. Genet., 13: 245-247,

1996.161. Lancaster, J. M., Wooster, R., Mangion, J., Phelan, C. M., Cochran, C., Gumbs, C.,

Seal, S., Barfoot, R., Collins, N., Bignell, G., Palei, S., Hamoudi, R., Larsson, C.,Wiseman, R. W., Berchuck, A., Iglehart, J. D., Marks, J. R., Ashworth, A., Stratton,M. R., and Futreal, P. A. BRCA2 mutations in primary breast and ovarian cancers.Nat. Genet., 13: 238-240, 1996.

162. Foster, K. A., Harrington, P., Kerr, J., Russell, P., DiCioccio, R. A., Scott, I. V.,Jacobs, I., Chenevix-Trench, G., Ponder, B. A., and Gayther, S. A. Somatic and

germline mutations of the BRCA2 gene in sporadic ovarian cancer. Cancer Res., 56:3622-3625, 1996.

163. Shattuck-Eidens, D., McClure, M., Simard, J., Labrie, F., Narod, S., Couch, F.,

Hoskins, K., Weber, B., Castilla, L., and Erdos, M. A collaborative survey of 80mutations in the BRCAI breast and ovarian cancer susceptibility gene: implicationsfor presymptomatic testing and screening. J. Am. Med. Assoc., 273: 535-541, 1995.

164. FitzGerald, M. G., MacDonald, D. J., Krainer, M., Hoover, I., O'Neil, E., Unsal, H.,

Silva-Arrieto, S., Finkelstein, D. M., Beer-Romero, P., Englert, C., Sgroi, D. C.,

Smith, B. L., Younger, J. W., Garber, J. E., Duda, R. B., Mayzel, K. A., Isselbacher,K. J., Friend, S. H., and Haber, D. A. Germline BRCAI mutations in Jewish andnon-Jewish women with early-onset breast cancer. N. Engl. J. Med., 334: 143-149,

1996.165. Struewing, J. P., Hartge, P., Wacholder, S., Baker, S. M., Berlin, M., McAdams, M.,

Timmerman, M. M., Brody, L. C., and Tucker, M. A. The risk of cancer associatedwith specific mutations of BRCAI and BRCA2 among Ashkenazi Jews. N. Engl.J. Med., 336: 1401-1408, 1997.

166. Neuhausen, S., Gilewski, T., Norton, L., Tran, T., McGuire, P., Swensen, J.,Hampel, H., Borgen, P., Brown, K., Skolnick, M., Shattuck-Eidens, D., Jhanwar, S.,

Goldgar, D., and Offit, K. Recurrent BRCA2 6174delT mutations in AshkenaziJewish women affected by breast cancer. Nat. Genet., 13: 126-128, 19%.

167. Chapman, M. S., and Verma, I. M. Transcriptional activation by BRCA. Nature(Lond.), 382: 678-679, 1996.

168.

169.

170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193.

194.

195.

Milner, J., Ponder, B., Hughes-Davies, L., Seltmann, M., and Kouzarides, T.Transcriptional activation functions in BRCA2. Nature (Lond.), 386: 772-773,

1997.Hakem, R., de la Pompa, J. L., Sirard, C., Mo, R., Woo, M., Hakem, A., Wakeham,A., Potter, J., Reitmair, A., Billia, F., Firpo, E., Hui, C. C., Roberts, J., Rossant, J.,and Mak, T. W. The tumor suppressor gene Brcal is required for embryonic cellularproliferation in the mouse. Cell, 85: 1009-1023, 1996.

Sharan, S. K., Morimatsu, M., Albrecht, U., Lim, D. S., Regel, E., Dinh, C., Sands,A„Bíchele, G., Hasty, P., and Bradley, A. Embryonic lethality and radiationhypersensitivity mediated by RadSl in mice lacking Brca2. Nature (Lond.). 386:804-810, 1997.

Hakem, R., de la Pompa, J. L., Elia, A., Potter, J., and Mak, T. W. Partial rescue ofBrcal (5-6) early embryonic lethality by p53 or p21 null mutation. Nat. Genet.. 16:298-302, 1997.

Scully, R., Chen, J., Plug, A., Xiao, Y., Weaver, D., Feunteun, J., Ashley, T., andLivingston, D.M. Association of BRCA 1 with Rad51 in mitotic and meiotic cells.Cell, 88: 265-275, 1997.Patel, K. J., Yu, V. P. C. C., Lee, H., Corcoran, A., Thistlethwaite, F. C., Evans,M. J., Colledge, W. H., Friedman, L. S., Ponder, B. A., and Venkitaraman, A. R.Involvement of Brca2 in DNA repair. Mol. Cell, /: 347-357, 1998.Scharer, E., and Iggo, R. Mammalian p53 can function as a transcription factor inyeast. Nucleic Acids Res., 20: 1539-1545, 1992.

Ishioka, C., Englert, C., Winge, P., Yan, Y. X., Engelstein, M., and Friend, S. H.Mutational analysis of the carboxy-terminal portion of p53 using both yeast andmammalian cell assays in vivo. Oncogene, 10: 1485-1492, 1995.Moshinsky, D. J., and Wogan, G. N. UV-induced mutagenesis of human p53 in a

vector replicated in Saccharomyces cerevisiae. Proc. Nati. Acad. Sci. USA, 94:2266-2271, 1997.Aguilar, F., Harris, C. C., Sun, T., Hollstein, M., and Cenati, P. Geographicvariation of p53 mutational profile in nonmalignant human liver. Science (Washington DC), 264: 1317-1319, 1994.

Aguilar, F., Hussain, S. P., and Cerutti, P. Aflatoxin B, induces the transversion ofG—>Tin codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proc.Nati. Acad. Sci. USA, 90: 8586-8590, 1993.

Hsu, I. C., Metcalf, R. A., Sun, T., Welsh, J. A., Wang, N. J., and Harris, C. C.Mutational hotspot in the p53 gene in human hepatocellular carcinomas. Nature(Lond.), 350: 427-428, 1991.

Bressac, B., Kew, M., Wands, J., and Ozturk, M. Selective G to T mutations of p53gene in hepatocellular carcinoma from southern Africa. Nature (Lond.), 350: 429-431, 1991.Scorsone, K. A., Zhou, Y. Z., Butel, J. S., and Slagle, B. L. p53 mutations cluster atcodon 249 in hepatitis B virus-positive hepatocellular carcinomas from China.Cancer Res., 52: 1635-1638, 1992.

Li, D., Cao, Y., He, L., Wang, N. J., and Gu, J. Aberrations of p53 gene in humanhepatocellular carcinoma from China. Carcinogenesis (Lond.), 14: 169-173, 1993.

Montesano, R., Hainaut, P., and Wild, C. P. Hepatocellular carcinoma: from gene topublic health. J. Nat!. Cancer Inst., 89: 1844-1851, 1997.Mace, K., Aguilar, F., Wang, J. S., Vautravers, P., Gomez-Lechon, M., Gonzalea,

F. J., Groopman, J., Harris, C. C., and Pfeifer, A. M. A. Aflatoxin B, induced DNAadduci formation and p53 mutations in CYP450-expressing human liver cell lines.Carcinogenesis (Lond.), IS: 1291-1297, 1997.

Puisieux, A., Ji, J., Guillot, C., Legros, Y., Soussi, T., Isselbacher, K., and Ozturk,M. p53-mediated cellular response to DNA damage in cells with replicative hepatitisB virus. Proc. Nati. Acad. Sci. USA, 92: 1342-1346, 1995.

Brash, D. E., Rudolph, J. A., Simon, J. A., Lin, A., McKenna, G. J., Baden, H. P.,Halperin, A. J., and Ponten, J. A role for sunlight in skin cancer: UV-induced p53mutations in squamous cell carcinoma. Proc. Nati. Acad. Sci. USA, 88: 10124-

10128, 1991.Hsia, H. C., Lebkowski, J. S., Leong, P. M., Calos, M. P., and Miller, J. H.Comparison of ultraviolet irradiation-induced mutagenesis of the lad gene inEscherichia coli and in human 293 cells. J. Mol. Biol, 205: 103-113, 1989.

Bredberg, A., Kraemer, K. H., and Seidman. M. M. Restricted ultraviolet mutationalspectrum in a shuttle vector propagated in xeroderma pigmentosum cells. Proc. Nati.Acad. Sci. USA, 83: 8273-8277, 1986.

Brash, D. E. UV mutagenic photoproducts in Escherichia coli and human cells: amolecular genetics perspective on human skin cancer. Photochem. Photobiol., 48:59-66, 1988.

Kress, S., Sutler, C., Strickland, P. T., Mukhtar, H., Schweizer, J., and Schwarz, M.Carcinogen-specific mutational pattern in the p53 gene in ultraviolet B radiation-induced squamous cell carcinomas of mouse skin. Cancer Res., 52: 6400-6403,

1992.Tornaletti, S., Rozek, D., and Pfeifer, G. P. The distribution of UV photoproductsalong the human p53 gene and its relation to mutations in skin cancer. Oncogene, 8:2051-2057, 1993.

Nakazawa, H., English, D., Randell, P. L., Nakazawa, K., Martel, N., Armstrong,B. K., and Yamasaki, H. UV and skin cancer: specific p53 gene mutation in normalskin as a biologically relevant exposure measurement. Proc. Nati. Acad. Sci. USA,91: 360-364, 1994.

Ziegler, A., Jonason, A. S., Leffell, D. J., Simon, J. A., Sharma, H. W., Kimmelman,J., Remington, L., Jacks, T., and Brash, D. E. Sunburn and p53 in the onset of skincancer. Nature (Lond.), 372: 773-776, 1994.Hernandez, T. M., and Hainaut, P. A specific spectrum of p53 mutations in lungcancer from smokers: review of mutations compiled in the IARC p53 database.Environ. Health Perspect., 106: 385-391, 1998.Klein-Szanto, A. J., lizasa. T., Momiki, S., Garcia-Palazzo, I., Caamano. J., Metcalf,R.. Welsh, J., and Harris, C. C. A tobacco-specific N-nitrosamine or cigarette smoke

4036

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 15: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

MOLECULAR EPIDEMIOLOGY OF HUMAN CANCER

condensate causes neoplastic transformation of xenotransplanted human bronchialepithelial cells. Proc. Nati. Acad. Sci. USA, 89.- 6693-6697, 1992.

196. Hill, A. B. The environment and disease: association or causation. Proc. R. Soc.Med., 58: 295-300, 1965.

197. Sasa, M., Kondo, K., Komaki, K., Uyama, T., Morimoto, T., and Monden, Y.Frequency of spontaneous p53 mutations (CpG site) in breast cancer in Japan. BreastCancer Res. Treat., 27: 247-252, 1993.

198. Tsuda, H., Iwaya, K., Fukutomi, T., and Hirohashi, S. p53 mutations and c-erbB-2amplification in intraductal and invasive breast carcinomas of high histologie grade.Jpn. 1. Cancer Res., 84: 394-401, 1993.

199. Blaszyk, H., Vaughn, C. B., Hartmann, A., McGovem, R. M., Schroeder, J. J.,Cunningham, J., Schaid. D., Sommer, S. S., and Kovach, J. S. Novel pattern of p53gene mutations in an American black cohort with high mortality from breast cancer.Lancet, 343: 1195-1197, 1994.

200. Hollstein, M., Rice, K., Greenblatt, M. S., Scussi, T., Fuchs, R., Sorlie, T., Hovig,E„Smith-Sorensen, B., Montesano, R., and Harris, C. C. Database of p53 genesomatic mutations in human tumors and cell lines. Nucleic Acids Res., 22: 3547-3551, 1994.

201. Weinberg, R. A. Tumor suppressor genes. Science (Washington DC), 254: 1138-

1146, 1991.202. Latif, F., Tory, K., Gnarra, J., Yao, M., Duh, F-M., Orcutt, M. L., Stackhouse, T.,

Kuzmin, I., Modi, W., Geil, L., Schmidt, L., Zhou, F., Li, H., Wei, M. H., Chen, F.,Glenn, G., Choyke, P., Walther, M. M., Weng, Y., Duan, D-S., Dean, M., Glavac,D., Richards, F. M., Crossey, P. A., Ferguson-Smith, M. A., Paslier, D. L., Chu-makov, I., Cohen, D., Chinault, A. C., Mäher,E. R., Linehan, W. M., Zbar, B., andLerman, M. I. Identification of the von Hippel-Lindau disease tumor suppressorgene. Science (Washington DC), 260: 1317-1320, 1993.

203. Trofatter, J. A., MacCollin, M. M., Rutter, J. L., Murrell, J. R., Duyao, M. P., Parry,D. M., Eldridge, R., Kley, N., Menon, A. G., Pulaski, K., Haase, V. H., Ambrose,C. M., Munroe, D., Bove, C., Haines, J. L., Martuza, R. L., MacDonald, M. E.,Seizinger, B. R., Short, M. P., Buckler, A. J., and Gusell, J. F. A novel moesin-,ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor.Cell, 72: 791-800, 1993.

204. Rouleau, G. A., Merei, P., Lutchman, M., Sansón, M., Zucman, J„Marineau, C.,Hoang-Xuan, K., Demczuk, S., Desmaze, C., Plougastel, B., Pulst, S. M., Lenoir, G.,

Bijisma, E., Fashold, R., Dumanski, J., de Jong, P., Parry, D., Eldridge, R., Aurias,A., Delattre, O., and Thomas, G. Alteration in a new gene encoding a putativemembrane-organizing protein causes neurofibromatosis type 2. Nature (Lond.), 363:515-521, 1993.

205. Schimke, R. N. Genetics and Cancer in Man, pp. 18-26. New York: ChurchillLivingstone, Inc., 1978.

206. Soini, Y., Chia, S. C., Bennett, W. P., Groopman, J. D., Wang, J. S., DeBenedetti,V. M., Cawley, H., Welsh, J. A., Hansen, C., Bergasa, N. V„Jones, E. A.,DiBisceglie, A. M., Trivers, G. E., Sandoval, C. A., Calderón, I. E., MuñozEspinosa, L. E., and Harris, C. C. An aflatoxin-associated mutational hotspot atcodon 249 in the p53 tumor suppressor gene occurs in hepatocellular carcinomasfrom Mexico. Carcinogenesis (Lond.), 17: 1007-1012, 19%.

207. Wogan, G. N. Aflatoxins as risk factors for hepatocellular carcinoma in humans.Cancer Res., 52 (Suppl.): 2114s-2118s, 1992.

208. Ross, R. K., Yuan, J. M., Yu, M. C., Wogan, G. N., Qian, G. S., Tu, J. T., Groopman,J. D., Gao, Y. T., and Henderson, B. E. Urinary aflatoxin biomarkers and risk ofhepatocellular carcinoma. Lancet, 339: 943-946, 1992.

209. IARC. Mycotoxins: aflatoxins; studies of cancer in experimental animals. In: IARCMonographs on the Evaluation of Carcinogenic Risks to Humans, pp. 263-395.Lyon, France: IARC, 1993.

210. Autrup, H., Harris, C. C., Wu, S. M., Bao, L. Y., Pei, X. F., Lu, S., Sun, T. T., andHsia, C. C. Activation of chemical carcinogens by cultured human fetal liver,esophagus and stomach. Chem.-Biol. Interact., JO: 15-25, 1984.

211. Pfeifer, A. M. A., Cole, K. E., Smoot, D. T., Weston, A., Groopman, J. D., Shields,P. G., Vignaud, J-M., Juillerat, M., Lipsky, M. M., Trump, B. F., Lechner, J. F., andHarris, C. C. SV40 T-antigen immortalized normal human liver epithelial cells

express hepatocyte characteristics and metabolize chemical carcinogens. Proc. Nati.Acad. Sci. USA, 90: 5123-5127, 1993.

212. Puisieux, A., Lim, S., Groopman, J., and Ozturk, M. Selective targeting of p53 genemutational hotspots in human cancers by etiologically defined carcinogens. CancerRes., 51: 6185-6189, 1991.

213. Wang, X. W., Gibson, M. K., Vermeulen, W., Yeh, H., Forrester, K., Sturzbecher,H. W., Hoeijmakers, J. H. J., and Harris, C. C. Abrogation of p53-induced apoptosisby the hepatitis B virus X gene. Cancer Res., 55: 6012-6016, 1995.

214. Ponchel, F., Puisieux, A., Tabone, E., Michot, J. P., Freschi, G., Morel, A. P.,Frebourg, T., Fontaniere, B., Oberhammer, F., and Ozturk, M. Hepatocarcinoma-specific mutant p53-249ser induces mitotic activity but has no effect on transforming growth factor ßI-mediated apoptosis. Cancer Res., 54: 2064-2068, 1994.

215. Vahakangas, K. H., Samet, J. M., Metcalf, R. A., Welsh, J. A., Bennett, W. P., Lane,D. P., and Harris, C. C. Mutations of p53 and ras genes in radon-associated lungcancer from uranium miners. Lancet, 339: 576-580, 1992.

216. Sozzi, G., Miozzo, M., Donghi, R., Piloni, S., Cariani, C. T., Pastorino, U., DellaPorta, G., and Pierotti, M. A. Deletions of 17p and p53 mutations in preneoplasticlesions of the lung. Cancer Res., 52: 6079-6082, 1992.

217. Sundaresan, V., Ganly, P., Hasleton, P., Rudd, R., Sinha, G., Blechen, N. M., andRabbitts, P. p53 and chromosome 3 abnormalities, characteristic of malignant lungtumours, are detectable in preinvasive lesions of the bronchus. Oncogene, 7: 1989-

1997, 1992.218. Chung, G. T., Sundaresan, V., Hasleton, P., Rudd, R., Taylor, R., and Rabbitts, P. H.

Sequential molecular genetic changes in lung cancer development. Oncogene, 11:2591-2598, 1995.

219. Chung, G. T., Sundaresan, V., Hasleton, P., Rudd, R., Taylor, R., and Rabbitts, P. H.Clonal evolution of lung tumors. Cancer Res., 56: 1609-1614, 1996.

220. Mao, L., Lee, J. S., Kurie, J. M., Fan, Y. H., Lippman, S. M., Lee, J. J., Ro, J. Y.,Broxson, A., Yu, R., Morice, R. C., Kemp, B. L., Khun, F. R.. Walsh, G. L.,Hittelman, W. N., and Hong, W. K. Clonal genetic alterations in the lungs of currentand former smokers. J. Nati. Cancer Inst., 89: 857-862, 1997.

221. Awaya, Y., Hiyama, K., and Yamakido, M. Analysis of oncogenes and suppressorgenes in lung cancer and bronchial lesions from high risk group. Nippon KyobuShikkan Gakkai Zasshi, 30 (Suppl.): 39-41, 1992.

222. Wistuba, I. I., Lam, S., Behrens, C., Virmani, A. K., Fong, K. M., LeRiche, J.,Samet, J. M., Srivastava, S., Minna, J. D., and Gazdar, A. F. Molecular damage inthe bronchial epithelium of current and former smokers. J. Nati. Cancer Inst., 89:1366-1373, 1997.

223. IARC. Benzo(a)pyrene. In: IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans, pp. 211-224. Lyon, France: IARC, 1983.

224. Dipple, A. Polynuclear aromatic carcinogens. In: C. E. Searle (ed.). ChemicalCarcinogens, pp. 245-314, Washington, DC: American Chemical Society, 1976.

225. IARC. Tobacco smoking. In: IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans, Vol. 38. Lyon, France: IARC, 1986.

226. Harris, C. C., Genta, V. M., Frank, A. L., Kaufman, D. G., Barrett, L. A., McDowell,E. M., and Trump, B. F. Carcinogenic polynuclear hydrocarbons bind to macromol-ecules in cultured human bronchi. Nature (Lond.), 252: 68-69, 1974.

227. Jeffrey, A. M., Weinstein, I. B., Jennette, K. W., Grzeskowiak, K., Nakanishi, K.,Harvey, R. G., Autrup, H., and Harris, C. C. Structures of benzo(a)pyrene-nucleic

acid adducts formed in human and bovine bronchial expiants. Nature (Lond.), 269:348-350, 1977.

228. Cherpillod, P., and Amstad, P. A. Benzo(a)pyrene-induced mutagenesis of p53hot-spot codons 248 and 249 in human hepatocytes. Mol. Carcinog., 13: 15-20,

1995.229. lizasa. T., Momiki, S., Bauer, B., Caamano, J., Metcalf, R., Lechner, J., Harris,

C. C., and Klein-Szanto, A. J. Invasive tumors derived from xenotransplanted,immortalized human cells after in vivo exposure to chemical carcinogens. Carcinogenesis (Lond.), 14: 1789-1794, 1993.

230. Daya-Grosjean, L., Dumaz, N., and Sarasin, A. The specificity of p53 mutationspectra in sunlight-induced human cancers. J. Photochem. Photobiol. B Bio!., 28:115-124, 1995.

231. Nakazawa, H., English, D., Randell, P. L., Nakazawa, K., Martel, N., Armstrong,B. K., and Yamasaki, H. UV and skin cancer: specific p53 gene mutation in normalskin as a biologically relevant exposure measurement. Proc. Nati. Acad. Sci. USA,91: 360-364, 1994.

232. IARC. Studies of cancer in animals. In: IARC Monographs on the Evaluation ofCarcinogenic Risks to Humans, pp. 139-162. Lyon, France: IARC, 1992.

233. Dumaz, N., Drougard, C., Sarasin, A., and Daya-Grosjean, L. Specific UV-inducedmutation spectrum in the p53 gene of skin tumors from DNA-repair-deficientxeroderma pigmemosum patients. Proc. Nati. Acad. Sci. USA, 90: 10529-10533,1993.

4037

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 16: Molecular Epidemiology of Human Cancer: Contribution of … · [CANCER RESEARCH 58. 4023-4037, September 15, 1998] Perspectives in Cancer Research Molecular Epidemiology of Human

1998;58:4023-4037. Cancer Res   S. Perwez Hussain and Curtis C. Harris  Mutation Spectra Studies of Tumor Suppressor GenesMolecular Epidemiology of Human Cancer: Contribution of

  Updated version

  http://cancerres.aacrjournals.org/content/58/18/4023.citation

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/58/18/4023.citationTo request permission to re-use all or part of this article, use this link

on July 15, 2020. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from