You are on page 1of 25

Mutations in the p53 Tumor Suppressor Gene: Clues to Cancer

Etiology and Molecular Pathogenesis


M. S. Greenblatt, W. P. Bennett, M. Hollstein, et al.

Cancer Res 1994;54:4855-4878. Published online September 1, 1994.

Updated Version Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/54/18/4855.citation

Citing Articles This article has been cited by 100 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/54/18/4855.citation#related-urls

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications
Department at permissions@aacr.org.

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
[CANCERRESEARCH54, 4855-4878, Septemberi5, 1994J

Perspectives in Cancer Research

Mutations in the p53 Tumor Suppressor Gene: Clues to Cancer Etiology


and Molecular Pathogenesist
M. S. Greenblatt, W. P. Bennett, M. Hollstein, and C. C. Harris2
Laboratory of Human Carcinogenesis, Division of Cancer Etiology, National Cancer institute, Bethesda, Maryland 20892 (M. S. G., W. P. B., C. C. HI, and Division of
Toxicology, German Cancer Research Center, D-69120 Heidelburg, Germany [M. H.]

I. Introduction

The p53 tumor suppressor gene has come to the forefront of cancer Ref. 10). Important sources of the spontaneous generation of point
research because it is commonly mutated in human cancer and the mutations in human cells include deamination of cytosine and 5-meth
spectrum of p53 mutations in these cancers is providing clues to the ylcytosine (1 1), DNA polymerase infidelity (12), depurination (13),
etiology and molecular pathogenesis of neoplasia (1—3).Detection of and oxidative damage from free radicals generated by biological
p53 abnormalities may have diagnostic, prognostic, and therapeutic processes (14, 15).
implications (4). The most well-studied endogenous mechanism of DNA damage at
The 15-year history of p53 investigations is a paradigm in cancer this time is the phenomenon of deamination of 5-methylcytosine.
research, illustrating the convergence of previously parallel lines of Methylation of DNA is one epigenetic mechanism involved in regu
basic, clinical, and epidemiological investigation and the rapid trans lating gene expression. Cytosine and 5-methylcytosine residues can
fer of research findings from the laboratory to the clinic. p53 is clearly spontaneously deaminate to uracil and thymine, respectively, which if
a component in biochemical pathways central to human carcinogen not repaired will result in G:C—>A:T transitions. These mutations
esis; p53 protein alterations due to missense mutations and loss of p53 occur most frequently at CpG dinucleotides (a cytosine followed by a
protein by nonsense or frameshift mutations provide a selective ad guanine), which are frequently methylated (9, 11, 16).
vantage for clonal expansion of preneoplastic and neoplastic cells (5). DNA replication in humans is performed by multicomponent protein
The potential for a missense mutation to cause loss of tumor suppres machines that include several DNA polymerases, which vary in charac
sor function and gain of oncogenic activity, i.e., to transform cells by teristics such as exonuclease activity and fidelity of replication. Error
two mechanisms, is one explanation for the commonality of p53 rates vary from 112,000 for DNA polymerase-@3,which repairs only short
mutations in human cancer. Recent studies investigating the mecha gaps, to 1/50,000- 1/1,000,000 for other polymerases which contain
nisms underlying the biological activity of p53 indicate that the exonuclease activity (17). The most common base substitutions observed
protein is involved in gene transcription, DNA synthesis and repair, are G:C—*
A:T and A:T—@ G:C transitions and G:C—*
T:A transversions.
genomic plasticity, and programmed cell death (1—6).These complex Imbalances in deoxynucleoside triphosphate pools (18), mutations in
biochemical processes are performed by multicomponent protein ma DNA polymerase-a (19), and slippage of DNA polymerase at nucleotide
chines; therefore, it is not surprising that the p53 protein forms repeats (Ref. 8; section II.B.2) are examples of mechanisms contributing
complexes with other cellular proteins (Fig. 1) and that some viral to infidelity of DNA synthesis. Considering the multiplicity of proteins
oncoproteins alter the functions of these machines by binding to p53 involved in DNA synthesis, DNA repair, mitosis, and the cell cycle, the
and perturbing its interaction with other cellular protein components. number of potential gene targets which could interfere with replication
In this Perspective, we will focus on the origin of p.53 mutations, fidelity in somatic cells is large.
the mutational spectrum of p.53 in human cancers, and the hypotheses Abnormalities in some of these processes could result in the mu
generated by the analysis of p53 mutations in premalignant and tator phenotype (20, 21), which could increase the probabilities of
malignant cells. The interpretation ofp53 mutations in human cancers both neoplastic transformation and the generation of increasingly
is based on observations of the patterns of DNA damage induced by malignant subclones during tumor progression (22, 23). If such
chemical and physical mutagens in model systems. In this Introduc mutations occur in germ cells and are nonlethal, they could lead to
tion, we will review these data, which provide the background for heritable syndromes of increased cancer risk. The hypothesis that
many of the inferences drawn from p53 mutational analysis. inherited genomic instability may be associated with increased cancer
risk is supported by studies of two diseases. Children with Bloom's
A. Origins of Mutation syndrome have an increased risk of childhood cancers, and their cells
show an increase in cytogenetic abnormalities (24). Germline muta
Both exogenous carcinogens and endogenous biological processes
are known to cause mutations (7—9).Classes of DNA damage include
tions in two DNA mismatch repair genes, hMSH2 (25, 26) and
hMLHJ (27, 28), recently have been identified as the genetic abnor
deletion, insertion, and base substitution, either transition (change of
malities responsible for HNPCC.3 Mutations of these genes are
a pyrimidine to another pyrimidine or a purine to another purine) or
transversion (change of a pynmidine to a purine or vice versa;
3 The abbreviations used are: HNPCC, hereditary nonpolyposis colorectal carcinoma;
aprt, adenine phosphoribosyl transferase; dhfr, dihydrofolate reductase; hprt, hypoxan
Received 4/27/94; accepted 7/20/94. thine-guanine phosphoribosyl transferase; CHO, Chinese hamster ovary; BPDE, 75,8R-
Thecostsof publicationof thisarticleweredefrayedin partby the paymentof page dthydroxy-9R,1OS-epoxy-7,8,9,10-tetrahydrobenzo(a)pyrene; BP, benzo(a)pyrene; IHC,
charges. This article must therefore be hereby marked advertisement in accordance with immunohistochemistry; SSCP, single-stranded conformation polymorphism; PCR, polymer
18 U.S.C. Section 1734 solely to indicate this fact. sac chain reaction; XP-C, xeroderma pigmentosum, complementation group C; PAH, poly
i Due to space limitations and the breadth of this topic, comprehensive reviews which cyclic aromatic hydrocarbon; AFB, aflatoxin B@; HCC, hepatocellular carcinoma; HBV,
are the source of additional citations will be cited frequently. hepatitisB virus; SCL@ small cell lung carcinoma;NSCLC, ron-small cell lung carcinoma
2 To whom requests for reprints should be addressed, at Laboratory of Human Carcino NPC, nasopharyngeal carcinoma EBV, Epstein-Barr virus; HPV, human papilloma virus;
genesis, Building 37, Room 20)5, National Cancer Institute, Bethesda, MD 20892. CAT, chloramphemcol acetyltransferase; LFS, Li-Fraumeni Syndrome; MP, mean pairwise.
4855

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

flG.1 FIG. 2A
Tr.lnsdc:' @atic@n
[ Mutations and -@Oligom.rlzat$on and

@
4.― Dvrnairi Sequence
DNA Binding
Specific
Domain
NucI@r Localization
Domains
MUTATIONAL
SPECTRUM
OF
110 273 ALLHUMANCANCER
14O@
120
100 1 24•
2% 8%
@ I 235 282
@ I 17%
4O@
20 .kli .a

.4 as
.4@V @P $V@ LARGE T-Ag ______
ISBIS H$P70 HSX 543
@ 1 1.4 EISMK £RC@.0
I IIYNPA NPA@ 17%
15@41 MOM2

SKINCANCERS
@ 2B SQUAMOUS CELL BASAL CELL MURINE (UV-INDUCED)
3% 2% 7%
1O%@ @j1% 13%

HEADANDNECKCANCERS
2E 2F 2G
NASOPHARYNX ORAL PHARYNX/LARYNX
6% 10/
0 10%

29%

LEGENDFORFiGURE2 A-O
U G:CtoC:G•
G:CtoT:A
•
G:CtoA:T
•
A:TtoT:A•A:Tto
G:C
@ U A:T
toC:G U-Non-CpG
•
CC:TT
toThAA Othir
Fig. 1. Schematic of the p53 molecule. Verticallines, distribution of mutations; horizontal bars, regions of protein binding. The p53 protein consists of 393 amino acids with functional
domains, evolutionarily conserved domains, and regions designated as mutational hotspots. Functional domains include the transactivation region (amino acids 20-42, orange area),
sequence-specific DNA binding region (amino acids 100—293),nuclear localization sequence (amino acids 316—325,dark green area), and oligomenzation region (amino acids 319—360,light
green area). Cellular or oncoviral proteins bind to specific areas of the p53 protein. Evolutionarily conserved domains (amino acids 17—29,97—292,and 324—352;rose-colored area) were
determined using the MACAW program as described in the text. 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. Functional domains and protein binding sites were compiled from references in text and from X. Wang and C. Harris(unpublished data). Vertical
lines above the schematic, missense mutations;lines below schematic, nonmissense mutations. The majority of missense mutations are in the conserved hydrophobic midregion, while
nonmissensemutations(nonsense,frameshift,splicing,and silent mutations)are distributedthroughoutthe protein, determinedprimarilyby sequencecontext, as described in the text.
Fig. 2. A-G. spectrum of p53 mutations in various tumor types. (n) the number of mutations for which exact base change or deletion was confirmed by DNA sequencing. A, the mutational
spectrumofp53 in all reportedtumorsand cell lines(n = 2567) in the database.B, squamous(n 32 induding Bowen's disease)and (C)basal cell (n 52)carcinomas ofthe skin in humans
and (D) UV light-inducedmurine skin tumors (n = 30) exhibit frequent G:C—÷A:T transitions and occasional tandem mutations at dipynmidine sites (usually CC:GG—@1T:AA),
both
characteristicof UV-induceddamage.Squamouscell carcinomashave been linkedwith occupationalexposureto PAHa,and one-fourthcontainG:C-@T:A transversions,which are consistent
with the mutations caused by these carcinogens in model systems. The mutational spectra differ by site of origin of head and neck squamous carcinomas [(E) nasopharynz n 17; (F) oral,
n 69; (G) pharvnxilar,nx. n 42J. 4856

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MIJFATIONS AND CANCER

FIG.2H-0 2H ADENOCARCINOMA 21 SQUAMOUS


LUNG
CANCERS 14%
BYCELL
TYPE 45%

8%

2J SMALL CELL 2K LARGECELL


4%

5%

13%

LUNG
CANCERS
a SMOKERS 2M NON-SMOKERS
6% 6%
6%
/0

11%
@ I 3%

8%

COLONSOMATICMUTATIONS LI-FRAUMENI
GERMUNEMUTATiONS
2N

8%

5%

15%

Fig. 2. (continued) H-O, spectrumof p53 mutationsin various tumortype&The mutationalspectra in lung cancers differ among histological types [(H) adenocarcinonia n 61; (1) sqisimous
n = 75; (J) 5771011cell (SfLC), n = 92; (K) large @dL n 27]. Menocarcinomasdisplaya lowermutationprevalence, a kwer percentage of G:C—@TA tranavetsions, aixi a higherpercentage
of
G:C-÷Altaiultians.The mutational specus in lungeancervaiy byanddnghistccy[(L)nnokers n = 169;(M)noiwnoke,@ n = 161.Themostcommon mutations in(N)coloncarcinoma(n = 324)
and (0) LFS (germline p53 mutations, n = 47) are GC-*A@T transitions at @pG dinucIeOtideS,which may be the result of spontaneous deamination of 5-inethylcytosine.
4857

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

associated with deletions and insertions in microsatellite sequences of experimental systems include differences in the genes and cell types
somatic cells and a genetic predisposition to tumors of the colon and studied, interspecies variability of DNA repair, and the potential
other tissues (29). Somatic mutations in !ZMSH2 have also been mutagenicity of transfection and shuttle vectors. The interpretation of
observed. Therefore, the mutator phenotype can be both a predispos mutational spectra is complicated, and observed mutations may not
ing inherited factor initiating carcinogenesis or a secondary somatic reflect all events at the DNA level. The DNA sequence can influence
change produced during this multistage process. both carcinogen-DNA adduct formation and DNA repair, and some
A well-studied mechanism by which chemical carcinogens and mutations may be either more readily detected due to clonal expansion
their ultimate carcinogenic metabolites cause mutations is by forming or less readily detected due to inhibition of the mutant cell population.
covalent adducts with the nucleotides in DNA, increasing the proba
bility of errors during DNA replication (30). Some small carcinogen
DNA adducts, such as 06-methylguanine resulting from alkylating 1. Mutational Spectra in Housekeeping Genes
agents, may cause DNA polymerase to misread the base pairing due The majority of spontaneous mutations in the model systems used
to the altered hydrogen bonding properties of a base which contains an are single base substitutions. Studies in prokaryotes indicate that
additional methyl or ethyl group. Laboratory studies have established carcinogen-induced mutations occur at nucleotides which are targets
that the most common mutations caused by alkylating agents are for carcinogen-DNA interactions and alterations (37). Differences
G:C—+A:T transitions, consistent with 06-methylguanine mispairing
have been observed between the spontaneous mutational spectra
with thymine (31). Bulky carcinogen-DNA adducts may render the of the endogenous aprt gene of CHO cells and both bacterial genes
bases unreadable and stall the replication machinery. DNA poly and exogenous genes carried by shuttle vectors in mammalian cells
merases may fill these noninstructive sites preferentially with adenine. (38). This may reflect either cell-specific or gene-specific biological
If this occurs opposite to a guanine (a common target for bulky properties.
carcinogens), a thymine will pair with the adenine in the next round A comparison of the spontaneous mutational spectrum at the aprt
of DNA synthesis, resulting in a G:C—*T:A transversion (32). Studies locus in CHO cells with the spectra induced by UV radiation, ionizing
using prokaryotic and eukaryotic cells and site-specific mutagenesis radiation, or BPDE illustrates carcinogen-specific damage (Table 1).
assays (13, 33) have shown that some carcinogenic agents produce a
The mutational spectrum of UV light is consistent with the mutagen
“fingerprint―
(1, 34, 35); specific types and locations of DNA adducts esis models of the promutagenic cyclobutane and pyrimidine-pyrimi
have been linked with the mutational spectrum of an agent. These done (6—4)photoproducts. Ionizing radiation often causes chromo
patterns have been studied in eukaryotes using both exogenous genes somal abnormalities, deletions, and losses (39, 40). BPDE, which
introduced into the appropriate host cell by a shuttle vector (33, 36) primarily binds to the 2-amino group of deoxyguanine, produces
and endogenous genetic loci (34) such as apri, dhfr, and hprt. predominantly the G:C—*T:A transversions predicted of a bulky car
B. Mutations in Model Systems cinogen-DNA adduct; a similar mutational spectrum for BPDE has
been observed in human cell assays (41). The hprt locus allows
The carcinogen-induced mutational spectra in prokaryotes and in comparison of mutational spectra obtained by exposing cultured cells
target genes in eukaryotes can be compared to spontaneous mutational to carcinogens with those observed in human donor lymphocytes
spectra, although uncertainty exists in the extrapolation of in vitro data exposed in vivo to environmental carcinogens (42). A recent com
to human carcinogenesis. Confounding variables which could affect prehensive database has been developed which contains more than

modeli'Gene Table ISpontaneous and induced mutational spectra: in vitro and anima!
[Ref.]G:C-@A:TG:C-'T:AG:C—@C:GA:T—*G:CA:T-@T:AA:T—@C:GCC:GG—'TT:AADe1C@dCHO
and Mutagen (n)―
aprt gene
Spontaneous (30) [281]
UV light (31) [282] 52 6 6 3 10 6 6 3 3
Ionizing radiation (16) [39] 19 6 6 6 13 19 0 31 0
BPDE (21) [283]71
5Human 513 623 140 07 90 00 06 50
aprt gene
Spontaneous (74) [43]
UV light (55) [43] 47 13 7 15 9 7 NR 2 0
BPDE (102) [43] 14 69 13 2 1 1 NR 1 0
MNNG (103)(43] 63 13 2 10 7 5 NR 0 1
0Animal
ENU (81) [43]28 3112 98 520 178 258 9NR NR15 50
p53 gene@
Mouse skin
UV light (33) [44, 451
BP (7) [46] 14 71 14 0 0 0 0 0 0
Butadiene (10)e 10 40 10 30 0 0 0 10 0
DMBA (18) [46, 48] 22 17 11 11 17 6 0 11 6
0Rat
MCA (7) [284]42 2918 570 140 018 00 021 00 00
esophagus
[285]90000000100Rat
NMBA (10)
liver
choline-deficient diet (9) 11 101 101 10
[286]5501
a Values, percentagesof each class of mutation;MNNG, N-methyl-N'-nitro-N-nitrosoguanidine;ENU, N-ethyl-N'-nitrosourea;DMBA, dimethylbenzanthracene;MCA, methyl
cholanthrene; NMBA, nitrosomethylbenzylamine; CHO< Chinese Hamster Ovary; Ref., reference no; NR, not reported.
h Number of mutations sequenced.
C Deletions.

d Insertions.
e R. Wiseman, personal communication.
I Examples of mutation prevalence may be found in Refs. 287 and 288.

4858

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

1000 human hprt mutations (43). The mutational spectra found at induced mutation patterns that override the spontaneous profiles are
the aprt locus of CHO cells and the hprt locus of human cells are more likely to reflect mutagen-DNA interactions.
detailed in Table 1. The most striking similarities are the high
percentages of G:C—'T:A transversions at both loci in cells ex II. p53 Mutational Spectrum Analysis in Human Cancers
posed to BPDE and high percentages of G:C—*A:T transitions in
UV-exposed cells, consistent with models predicted for these One of the goals of carcinogenesis research is to identify the precise
agents. The largest difference between the two systems is in the molecular alterations responsible for neoplastic transformation, ab
spontaneous mutational spectrum. In the aprt locus, G:C— A:T normal differentiation, and growth control. Molecular epidemiology
transitions account for 71% of the 30 spontaneous alterations, has developed to attempt to integrate traditional epidemiological
whereas in the hprt locus, only 28% of the 74 mutations are investigation of cancer risk factors with the substantial expansion of
G:C—t@
A:T (P = 0.001, Fisher's exact test). Thus, cell-specific or knowledge of the molecular mechanisms of cellular processes (49).
gene-specific features may affect spontaneous mutation patterns. Mutational spectrum analysis, the study of the types and locations
of DNA alterations, describes the often characteristic patterns of DNA
changes caused by endogenous and exogenous mutagens. Alterations
2. Mutational Spectra in Animal Models
of cancer-related genes found in tumors not only represent the inter
Animal models provide opportunities for mutational spectrum stud actions of carcinogens with DNA and cellular DNA repair processes
ies that cannot be performed with human tumors. For example, dose but also reflect the selection of those mutations which provide pre
response experiments can define the mutational spectra of carcinogens malignant and malignant cells with a growth advantage. Study of the
in vertebrate neoplasms. The spectrum of carcinogen-induced muta frequency, timing, and mutational spectra of abnormalities of p53 and
tions in cancer-related and housekeeping genes can be compared in other cancer-related genes can thus generate and test a variety of
rodent and human cells, assessing the relevance of these model hypotheses in carcinogenesis. These include questions regarding car
systems to human carcinogenesis. Furthermore, transgenic animals cinogen-DNA interactions, functions of the affected gene products,
can provide mechanistic insights into the interplay between environ mechanisms of carcinogenesis in specific organs or tissues, and fea
mental carcinogens and host susceptibility factors such as carcinogen tures of general cell biology such as DNA replication and repair.
metabolism and alterations of oncogenes and tumor suppressor genes. The information derived from this type of mutational spectrum
The frequency of p53 alterations in murine primary tumors is analysis is most relevant when it addresses alterations of genes vital to
generally lower than in human tumors. p53 mutations have been found carcinogenesis. A burgeoning literature (reviewed in Refs. 1, 2, 5, and
in tumors of the breast (42%), lung (8%), skin (30%), and other tissues 6) attests to the important role of p53 protein in normal cell function
(Table 1). Alterations generally have not been found in colon or liver and neoplastic transformation. Mutations of p53 are the most common
tumors. The low prevalences in lung and colon tumors are surprising genetic abnormality yet found in human cancers. The prevalence of
for several reasons: (a) the gene is highly conserved between mice p53 mutations varies among tumor types, ranging from 0 to 60% in
and humans; (b) p.53 mutation frequency is high in human lung and major cancers (Table 2), and is over 80% in some histological sub
colon cancers; (c) the morphological features of lung and colon types. All classes of mutations occur in the p53 gene (Fig. 2A);
adenocarcinomas are similar in humans and mice; and (d) lung and three-fourths of substitutions occur at G:C base pairs.
colon tumors from both species frequently contain ras mutations. The The p53 gene is well-suited to mutational spectrum analysis for
mutation database in laboratory animals is still limited (Table 1), and several reasons. First, since p53 mutations are common in many
more studies of different carcinogens and dose regimens are needed. human cancers, a sizable database has accrued whose analysis can
The most well-studied system is skin carcinogenesis in the mouse. yield statistically valid conclusions (50). Its modest size (1 1 exons,
Several carcinogens have been examined in rat, hamster, and nonhu 393 amino acids) permits study of the entire coding region, and it is
man primate models, but mutation rates are generally too low to highly conserved in vertebrates, allowing extrapolation of data from
generate meaningful spectra. animal models (51). Point mutations which alter p53 function are
Uv exposure efficiently induces squamous skin carcinomas in distributed over a large region of the molecule, especially in the
several strains of inbred mice (44, 45). In these studies, as in human hydrophobic midportion (1), where many base substitutions alter p53
tumors, there is a striking preference for single base transitions and conformation and sequence-specific transactivation activity (sec.
tandem mutations at dipyrimidine sites (30 of 33; 91% of mutations). II.C.1.); thus, correlations between distinct mutants and functional
BP, a carcinogen found in tobacco smoke and burning fossil fuels, changes are possible. Frameshift and nonsense mutations which trun
induces papillomas and squamous carcinomas in Sencar mice when cate the protein can be located outside of these regions, so evaluation
applied topically. p53 mutations were identified in 1 of 8 papillo of the entire DNA sequence yields relevant data. This situation differs
mas and 6 of 12 carcinomas (46) with 5 G:C—@ T:A transversions, from that of the ras oncogenes. Transforming mutations of ras occur
1 G:C—t@C:G transversion, and 1 G:C—@. A:T transition (Table 1). primarily in three codons and are thus limited to a few sequence
These data provide the first characterization of the p53 mutation motifs which identify a critical functional domain (52). The diversity
spectrum of BP-induced tumors in vertebrates. Skin tumors in of p53 mutational events permits more extensive inferences regarding
duced by 7,12-dimethylbenz(a)anthracene provide a contrasting mechanisms of DNA damage and mutation.
mutation spectrum. Eighteen p53 mutations have been reported For this Perspective, we compiled 2567 mutations in human carci
(46—48), and the mutation pattern is notable for a high frequency nomas or cell lines from over 300 papers published or in press through
(40%) of mutations at A:T base pairs and a low frequency of January 1, 1994, as identified by searches of Medline and Current
G:C—―
T:A transversions. This is consistent with the mutagenic Contents in January, 1994. Only mutations confirmed by DNA
specificity of 7,12-dimethylbenz(a)anthracene (48). sequencing were entered into the database. Selected additional reports
These mutational spectra of aprt, dhfr, and hprt genes and animal were added to illuminate specific discussions. An updated version of
models serve as a foundation with which to compare the spectra the database is available from the European Molecular Biology Lab
observed in cancer-related genes in humans. Differences in spontane oratory Data Library through e-mail; details of its compilation
ous mutation patterns may reflect differences in inherent characteris have been reported separately (50). Here we analyze these data with
tics of various cell types (e.g., DNA repair capacities), whereas attention to: (a) questions regarding carcinogenesis which have been
4859

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MLflA11ONS AND CANCER

cancers'@Tumor Table 2 Preyalence and spectra ofp53 mutations in human

type p.53
(n)―Prey. Mut.c %G:C-a A:T %G:C—aT:A %G:C-+ C:G %A:T—'G:C %A:T—*T:A %A:T-@ C:G %Del/Ins/ OtherCpG W HOTSPOTS1
All tumors 37 41 17 8 11 6 4 13 24 •175
lb H
(2567) •245G> S,D,C,v
•248lb W,Q
249 R> S,M
•273
lb H,C
Lung 56 24 40 9 7 5 3 11 9 157 V> F
(897) •24@
lb W,L,Q
249 R> M,S
*273 R> L,H,C
Colon 50 63 9 3 11 4 1 8 47 ‘175@>H
(960) •245G> S,D,V
•248lb W,Q
p273 lb H,C
p282 lb W
Esophagus 45 38 16 3 15 14 3 12 18 None
(279)
Ovary 44 42 9 12 18 3 6 9 23 @273@>C,H
(386)
Pancreas 44 41 13 6 17 8 5 10 21 @273@>H
(170)
Skin 44 41 12 6 4 4 2 31 17 @248R>W
(220) 278 P> S,F
Gastric 41 47 6 3 17 5 6 16 35 None
(314)
Headandneck 37 31 18 11 14 5 3 19 13 @@4flbQWJ@
(524)
Bladder 34 37 14 21 9 3 2 14 16 280R>T
(308)
Sarcoma 31 47 14 6 7 6 1 19 21 None
(339)
Prostate 30 44 13 6 16 0 6 16 28 None
(87)
Hepatocellular 29 20 37 8 11 12 3 9 9 249 lb S,M,W
(716)
Brain 25 50 11 5 14 1 3 15 30 *175@>H
(456) •248
R> Q,W
Adrenal 23
(31)
Breast 22 36 13 8 11 7 6 16 23 @175R>H
(1536) •248R> Q,W
*273 R> H,C
Endometrium 22 51 10 4 14 0 6 14 37 •24@B.> W,Q
(224)
Mesothelioma 22
(23)
Renal 19
(102)
Thyroid 13 48 9 13 9 0 4 17 28 *248R>Q,G,W
(299) 273 lb.H
Hematological 12 49 7 7 12 4 5 15 31 •175P.> H
(1916) •24@lb. Q,W
Carcinoid 11
(61)
Melanoma 9
(70)
Parathyroid 8
(13)
Cervix 7 35 35 10 0 0 10 10 25 ‘273@>C
(350)
Neuroblastoma 1
(212)
Wilms 0
(41)
Testes 0
(40)
Pituitary 0
(27)
Pheochromocytoma 0
(47)
a Mutationalspectrum of a tumor type was calculated only if 20 mutationswere reported.
b n, the number of tumors of each cell type evaluated for p53 mutation by PCR-based techniques as compiled in the p53 mutation database (50).
C Prevalence of p53 mutations in cancers from each organ/tissue, as detected by PCR-based techniques, screening at least exons 5—8. Premalignant lesions such as adenomas or

dysplastic lesions are not included in prevalence data. Prevalence in all cancers is a crude estimate derived from statistics on worldwide cancer incidence and the prevalence of p53
mutations for each type as calculated from the reports in the mutation database (see text).
d Del, deletion; Ins, insertions.
@ e of mutationswhich are G:C—@
A:T transitionsat CpG dinucleotides.
I Hotspots for the entire database are defined as codons at which at least 50 mutations have been reported. Hotspots for individual tumor types include codons at which at least 10
mutations, or 10% of all mutations for that tumor, have been reported. Letters refer to amino acid change, using standard one letter code. Amino acid substitutions which occur more
than once are listed in descending order of frequency at each codon.
*, CpG site.

4860

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MIThATIONS AND CANCER

addressed by mutation analysis, and conclusions reached to date; (b) could interfere with a feedback loop which regulates expression of
how mutational spectrum analysis and in vitro experiments provide wild-type p53. This can be tested by searching for mutations in genes
complementary information on carcinogenesis; and (c) new hypoth downstream in the p53 biochemical pathway in the cancers in which
eses generated from the accumulated information to be specifically staining is frequently detected but which rarely contain p53 mutations.
addressed by future studies of p53 mutations. Other methods of screening tumors to identify those likely to
contain mutations involve detection of altered electrophoretic mobil
A. Analytical/Technical Considerations ity patterns of DNA sequences containing point mutations. The most
1. Methods ofAnalysis for p53 Abnormalities common techniques, SSCP (65—67),denaturant gradient gel electro
phoresis, and constant denaturant gel electrophoresis (68, 69), have a
The methods of analysis for mutations can bias conclusions regard sensitivity and specificity of approximately 90% in detecting muta
ing the prevalence and nature of p53 mutations. A few caveats tions confirmed by DNA sequencing (67, 69, 70). Sequencing should
concerning techniques and experimental design are warranted before be done to confirm that positive results represent mutations and not
examining the data in detail. technical artifacts or germline polymorphisms, which have been
Solid tumors are heterogeneous, consisting of tumor cells mixed described at codons 72 and 213 (71, 72).
with nonneoplastic stromal cells and necrotic debris. Since tumors PCR-based techniques, including SSCP, constant denaturant gel
undergo clonal evolution, different sites within a malignancy may also electrophoresis, and DNA sequencing, are subject to false negative
vary in genotype and phenotype (21); a false negative result might and false positive results. The polymerases used in PCR to amplify
arise from sampling error. Microscopic assessment is important to specific gene sequences are subject to error rates from 1/10,000 to
ensure that tissue samples contain adequate amounts of the desired more than 1/500 base pairs, depending on reaction conditions (73).
histological features and to avoid analysis of necrotic debris. Samples Large deletions might not be detected if the missing segment includes
may be enriched for tumor content by several methods, such as primer sites, and the wild-type gene of stromal tissue is amplified.
manual microdissection of slides (removing only the tumor tissue for Contamination of a PCR reaction with other airborne PCR products
study) and cell sorting using DNA flow cytometry (53, 54). may also lead to false positive or negative sequencing results (74).
DNA sequencing is required for precise identification of mutations, Therefore, it is imperative that all mutations found by direct sequenc
but an altered p53 gene or protein can be detected by other immuno ing of PCR products be confirmed by sequencing of a second, mdc
logical or chemical tests (55—58).Because DNA sequencing is time pendent PCR product. This practice has not been followed uniformly;
and labor intensive, studies evaluating human cancers for p53 muta hence, some mutations in the database may represent false positives.
tions often use screening tests, performing DNA sequencing only on There also is considerable inconsistency among papers in the extent of
tumors which are likely to contain mutations. The sensitivity and analysis. Mutation prevalence data are included in this report only if
specificity of these techniques are critical factors which could intro a minimum of exons 5—8was sequenced. Due to underdetection,
duce bias into p53 mutation prevalence data. mutation prevalence rates are likely underestimated in the database,
The wild-type p53 protein usually resides in the cell nucleus, but although reported rates for some tumors were probably inflated due to
has a very short half-life and is present in such small quantities that it preselection of samples by screening tests. Thus, absolute mutation
cannot be detected by routine IHC. Missense mutations often increase prevalence rates are difficult to assess, given the heterogeneity of
the half-life and quantity of the p53 protein, allowing its detection by techniques in cited reports.
IHC (58). Many investigators have used IHC as a screening test prior
to DNA sequencing, and staining is often considered a surrogate
marker for gene mutation even in the absence of confirmatory DNA 2. Extent ofp53 Gene Analysis
studies. In the 84 studies we identified which report IHC and sequenc
ing in the same tumor sets, positive staining was found in 44% of The p53 gene consists of 11 exons; exons 2—1
1 code for the protein
tumors by IHC, while 36% contained mutations. However, the sen of 393 amino acids. Early studies noted that most p53 mutations
sitivity of IHC in these studies was only 75% (range, 36—100%),and occurred in the regions of the gene which are highly conserved
the positive predictive value was only 63% (range, 8—100%), with through evolution and presumably of functional importance, primarily
considerable variation among tumor types. Thus, the status of the p53 in exons 5—8(codons 126—306) (55). Based on these preliminary
protein by IHC should not be equated with wild-type or mutant data, most investigators have confined their analyses to exons 5—8
genotype. (and sometimes exon 4). Partly because of this bias in searching, 95%
Discrepancies between IHC and sequencing results may be due to of the reported mutations have been found in exons 5—8and their
false negative and positive results from technical aspects of sample intervening introns (mutations in introns which affect splicing of the
preparation and analysis or the result of biological phenomena respon next exon have been classified as mutations of the deleted exon). We
sible for the discordance (reviewed in Ref. 58). Mutations which have identified 50 studies in which sequencing of the entire coding
result in deletion or truncation of the protein (nonsense and frame region of p53 was reported. Of the 560 mutations reported in these
shift) do not cause protein accumulation. Therefore, IHC will be less papers, 87% were in exons 5—8,and most of the others were in exons
sensitive in detecting mutations in tumor types with high proportions 4 (8%) and 10 (4%). The distribution varied among tumor types.
of these nonmissense mutations. Some tumors, including melanoma Mutations outside exons 5—8were most frequent in bladder (28%)
(59, 60) and testicular carcinoma (61, 62), are frequently immuno and hepatocellular (24%) carcinomas and least frequent in head and
positive for p53 in the absence of mutation. Potential mechanisms for neck and hematopoietic malignancies (2% each). Over 70% of these
positive IHC due to accumulation and staining of nonmutant p53 mutations generated stop codons or frameshifts; therefore, they likely
protein include stabilization of p53 protein by binding to viral or would be missed by IHC screening.
cellular proteins and DNA damage from chemical or physical geno Thus, evaluation of only exons 5—8is likely to underestimate the
toxic agents (58). Another hypothesis is that mutations are occurring prevalence of p53 mutations, especially nonmissense mutations, by
in a gene downstream of p53, such as WAF1/Cipi, a Mr 21,000 over 20% in some tumors. Analysis of exons 4 and 10 is therefore
protein which is transactivated by the p53 protein and is an inhibitor warranted when mutations are not found in exons 5—8.The tissue
of cyclin-dependent kinases (63, 64). Interruption of the p53 pathway specific differences in the prevalence of mutations in various exons
4861

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

are clues that detailed p.53 mutational spectrum analysis can reveal induced on the nontranscribed (coding) strand by UV light or BP are
subtle patterns of genetic damage. much more likely to be CC-+ U than GG—AA and G—@ T than
Another potential source of underestimating the prevalence of p.53 C—@
A, respectively. This difference is thought to be an evolutionary
mutations might be the presence of mutations in unevaluated DNA adaptation which ensures that structure and function of essential
sequences, such as its 2 promoters or its 10 introns. Although the proteins with short half-lives are maintained, increasing the likelihood
intron bases adjacent to exons, which are responsible for splicing, are of cell survival. Repair of the nontranscribed strand can occur later
often examined, complete evaluation of introns is not common. Re and still preserve the fidelity ofthe gene in the daughter cells. If repair
verse transcription-PCR, a technique using reverse transcriptase to of the nontranscribed strand does not take place, then daughter cells
amplify mRNA and sequence the resulting complementary DNA, might arise with mutations that are lethal or that confer a survival
cannot evaluate introns, which have been removed by splicing. Mu advantage on the mutant clone.
tations in intronic regulatory regions may be prevalent and undetected by The hypothesis that deficient DNA repair can affect mutation
screening with IHC (if protein does not accumulate) and PCR-based patterns in humans was tested by measuring the repair of UV-induced
techniques (if primers do not encompass the relevant intron sequence). cyclobutane pyrimidine dimers in the individual DNA strands of p53
in a normal, repair-proficient human fibroblast strain and in fibro
3. Epidemiokgkal Considerations blasts from a patient with the repair-deficient disorder XP-C (78).
Selective repair of the transcribed DNA strand of p53 is observed in
Specific features of populations evaluated for p53 mutations may both normal and XP-C fibroblasts, and the strand bias of repair is
also create bias in the conclusions. For some cancer types, differences more pronounced in XP-C cells. Since mutations may occur due to
in frequency or spectrum have been detected between groups which replication errors at the sites of unrepaired DNA damage, these results
differ in ethnicity or nationality, possibly due to specific carcinogen predict that the majority of p.53 mutations in skin cancers, especially
exposures or inherited features in these populations (sec. II.D.3.). If those from patients with XP-C, would occur on the nontranscribed
atypical subgroups are overrepresented, features of the mutational strand. Indeed, 100% of tandem mutations in skin cancers from XP-C
spectrum of other groups may be obscured. patients and four of seven from non-XP patients exhibit such a strand
In order to yield valid conclusions, molecular epidemiology must bias (80, 81). More data in non-XP cancers are needed. Tumors from
adhere to standard epidemiological principles of statistical analysis: XP-Cpatientsare muchmorelikelythan sporadiccancersto contain
i.e., selection of appropriate control and study groups, assessment for tandem pyrimidine transitions (58 versus 11%), suggesting a differ
potential confounding variables (which may be different for each ential ability to repair this type of DNA damage. This sequence of studies
tumor type), and adequate sample sizes to minimize type I errors (e.g., is an example of testing in the laboratory of a hypothesis generated by
the premature assignment of relationships between putative carcino analysis of the p53 mutation spectrum and then predicting the result of
gens and cancers) and provide the statistical power to detect important further molecular epidemiological investigation of human tumors.
differences and avoid type II errors (the erroneous conclusion that no Some tumor types have been linked by epidemiological and labo
difference exists). Although these admonitions may seem elemental, ratory evidence to specific carcinogens with known mutational spec
the literature contains many examples of preliminary conclusions tra, such as lung cancer and exposure to cigarette smoke (section
based on small sample sizes, which often pass into general opinion II.D.5.C). Ninety-one percent of G:C—+T:A transversions in lung
without sufficient critique. The database provides a powerful tool to cancer occur on the nontranscribed DNA strand (i.e., are G—+
T rather
highlight the areas in which data are inadequate and focus future endeav than C—+
A), supporting the hypothesis that DNA repair is an impor
ors. tant determinant of mutation specificity of cancer-related genes in
human cancer. The hypothesis may be extended to postulate that a
B. Patterns of Mutation: Insights into DNA Replication nontranscribed strand bias in the mutation pattern of a specific base
and Repair substitution or tumor type implies that an exogenous carcinogen may
1. DNA Repair and Strand Bias of Mutations be responsible. Evaluating the entire p53 database for strand bias may
therefore provide clues to suspected and unrecognized carcinogens.
Since all organisms are constantly exposed to exogenous and en Four patterns of base substitution exhibit strand bias: G—+
T (87%
dogenous mutagens, sophisticated systems of DNA repair to maintain on the nontranscribed strand) and T—* G (67%) transversions, and
the integrity of the genome have evolved. DNA repair may be defined A—p
G (70%) and nonCpG site G-+ A (65%) transitions (Table 3).
as a cellular response which restores the normal nucleotide sequence G:C—*
A:T transitions occurring at CpG dinucleotides are reported
and stereochemistry of DNA following damage. DNA damage often separately from other G:C—@
A:T transitions because of differences in
causes a physical block to transcription; therefore, repair is generally mechanism discussed in section I. Bulky carcinogens such as BP,
faster in actively transcribed genes and is closely linked to other other PAHs (82), and AFB (82—84)have been associated with tran
cellular pathways which control gene expression (75). Several re sitions and transversions at both G:C and A:T base pairs (85). The
cently described genes code for enzymes exhibiting both DNA repair relative contributions of these and other agents to human somatic
and gene transcription activities, suggesting a potential molecular mutations are unknown.
mechanism for control of cellular proliferation until damage is cor In support of the hypothesis that tumors which are associated with
rected (76, 77). In addition, the p53 gene is repaired faster than the environmental carcinogens should contain a high proportion of base
dhfr gene, and much more efficiently than the inactive genomic substitutions with bias toward the nontranscribed strand, Table 3 lists
regions (78). In vitro studies and observations in repair-deficient organ all tumor types in descending frequency of the prevalence of the base
isms indicate that repair is important in determining mutation patterns. substitution types which exhibit strand bias (G—* T, non-CpG site
Importantly, repair is faster on the transcribed (template) strand of G—+A, A-t' G, and T—t'G). Among the tumors which most frequently
DNA than on the nontranscribed (coding) strand (75). Experiments in contain these types of mutations are tobacco-associated cancers of the
bacteria, yeast, and mammalian cells and in vivo mouse skin carcino lung (most frequent), cervix, esophagus, head and neck (fourth, fifth,
genesis models have shown that mutations induced by exogenous and sixth most frequent, respectively), and hepatocellular carcinoma
carcinogens preferentially occur on the nontranscribed strand of genes (second most frequent), which is associated with dietary AFB expo
such as dhfr, hprt (79), and p53 (46, 75). That is, the mutations sure in certain geographic regions (section II.D.5.B). Most of these
4862

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

Table 3DNA strandbias by tumor%

G-+ATumorTotal
T—Gtype % of% G—TonNon-CpG on% A-@G%
at strand G—aA nontran nontran
strandAlltumors50171711487657067(2567)Lung6640157395529164(328)HCC61381011389777571(224)Pancreas56132117510069730(63)Cervix55351001086100NAe50(20)Esophagus53161915
(n)°Muts bias site―% G—'D% non-CpG% A—@G%T—*Gnontran strand―nontran strandon strandon

Skin401222527742'@20100(110)Gastric4061117671693857(121)Thyroid39917941003810050(46)Colon36915II268646650(324)

a ,@ number of mutations detected in benign and malignant tumors at each site. These numbers differ from values obtained from Table 2 by multiplying the total number
tested X prevalence rates because: (a) some tumors contain multiple mutations; (b) mutations in nonmalignant precursors are included here but not in Table 2 prevalence data; and
(c) only sequenced mutations are included here, whereas mutations determined by other techniques may be included in prevalence statistics.
b Percentage of p53 mutations that are one of the indicated base substitutions associated with strand bias.
C Percentage of p53 mutations of indicated base substitution on both strands.

d Percentage of indicated base substitution on nontranscribed strand.


e NA, not applicable.
@ I light-induced transitions at tandem pyrimidine sites are a cause of C—*
T transitions with nontranscnbed strand bias in experimental models. Therefore, in skin cancer, C—'T
transitions are considered to be carcinogen-induced nontranscribed strand mutations.

substitutions exhibit marked strand bias. When strand bias is not the DNA sequence context is the most important factor determining
apparent, it may reflect a mechanism of mutation which does not these events. Almost all short deletions and insertions occur at mono
exhibit strand bias [such as non-bulky DNA adducts of alkylating tonic runs of two or more identical bases or at repeats of 2- to 8-base
agents, which cause mainly G:C—A:T transitions due to mispairing pair DNA motifs, either in tandem or separated by a short intervening
(31, 86)]. Alternatively, multiple carcinogens could be involved, and sequence. Several mechanisms are probably involved (90). The most
the bias patterns could negate each other (e.g., in esophageal and lung well-studied is called slipped mispairing, a misalignment of the tem
cancer, one tobacco-associated carcinogen might cause G—A and plate DNA strands during replication that leads to either deletion, if
another, C—p
T transitions). In vitro studies which establish the mu the nucleotides excluded from pairing are on the template strand, or
tational spectrum of a mutagen can be complementary to analyses of insertion, if they are on the primer strand. When direct repeat se
clinical specimens for strand bias in base substitutions and can im
quences mispair with a complementary motif nearby, the intervening
plicate potential tumor-specific carcinogens (section II.D.5). In the
oligonucleotide sequence may form a loop between the two repeat
lower half of Table 3 (i.e., tumors which exhibit few mutations with
motifs and be deleted (88, 89). More lengthy runs and sequence
strand bias) are cancers which have not been strongly associated with
repeats are more likely to generate frameshift mutations. In vitro
environmental carcinogens (87), supporting the nontranscribed strand
bias-exogenous carcinogen hypothesis. research which detects errors in replication of reporter genes has
helped quantify this phenomenon (reviewed in Ref. 17).
The database contains 285 deletions or insertions of 30 base pairs,
2. Deletions and Insertions: DNA Replication and the Mispairing
including 98 single base pair deletions, 40 single base pair insertions,
Hypothesis
and 217 total events of @6 base pairs. Almost all (265; 90%) are at
Previous analyses of short deletions and insertions in p53 (88) and monotonic runs and short direct repeats, supporting the prior obser
other genes associated with genetic disease (89) have concluded that vations that mispairing is the primary mechanism of short deletions
4863

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONSAND CANCER

and insertions in human genes. Both deletions and insertions increase cated proteins. Some of these inferences have been corroborated by
in frequency as the number of repeated bases in a run increases. The recent evidence describing the nuclear magnetic resonance analysis
region of p53 which most frequently contains deletions or insertions and the crystal structure of several p53 domains (96, 97).
is codons 151—159.Twenty-seven (9% of all deletions and insertions A schematic of the p53 molecule, indicating the functional regions
and 1% of all p53 mutations) have been reported at this G:C-rich most frequently evaluated, is shown in Fig. 1. The NH2-terminal of
sequence with multiple runs and direct repeats, confirming in vitro p53 contains an acidic domain similar to those found in other fran
observations regarding sequence motifs which predispose to mispair scription factors (5). Chimeric proteins containing amino acids 20—42
ing and other mechanisms of frameshift mutations. fused to the DNA binding site of Gal4 will transactivate reporter
Much remains unknown regarding the cellular phenomena which genes linked to Gal4-binding DNA sequences (98). Interestingly, this
influence the risk of mispairing. Factors which affect DNA polymer region also is responsible for binding to cellular protein Mdm2, and
ase function, e.g., binding to polymerase accessory proteins, are areas direct inhibition of transactivation is the likely mechanism by which
of interest for mechanistic study (89). Some bulky chemical mutagens Mdni2 interferes with p53 function (99—102).A proline-rich linking
induce frameshift mutations in prokaryotic models through mecha region between amino acids 60 and 92 begins the hydrophobic mid
nisms related to adduct-induced deformation of the DNA helix. These section of the protein. Between amino acids 100 and 295 lies the
adduct-induced mutations occur more frequently in monotonic or critical region (delineated by studies of truncated and chimeric pro
tandem base runs and at G:C-rich sequences (82, 90, 91). The con teins) which determines p53 conformation, specific binding to DNA
tribution of chemical carcinogens to frameshift mutations of p53 in consensus sequences, and sequence-specific transcriptional activity
eukaryotic cells is potentially significant. Tumor-related differences in and which may be essential for growth suppression (103). This region
deletion prevalence may reflect organ-specific carcinogens or tissue also binds to SV4O T-antigen (104). The recently described crystal
specific properties that regulate replication fidelity. structure of the DNA-binding domain identifies several motifs which
It is uncertain whether the recently described human mismatch directly contact DNA, including nine amino acids which abut the p53
repair genes hMSH2 and hMLHJ (25—28), which are responsible for consensus binding sequence (97). The positively charged carboxy
causing mutations at tandem repeat DNA sequences (microsateilites) terminus contains the basic nuclear localization sequence (amino
in families with FINPCC (section I.A; Refs. 92 and 93), play a role in acids 316—325)and oligomerization domain (amino acids 319—360;
p53 deletions and insertions. Most events in colon and other cancers Refs. 5, 96, 105, and 106). Oligomerization into dimers or tetramers
occur at consecutive base runs, but 40% are at short tandem repeats. appears to be required for DNA binding and transactivation function
None of the seven deletions at the most mutable tandem repeat (97, 107, 108). The observation that the nonsense mutation
sequence in the p53 gene (GTG GTG GTG at codons 216—218)are CGC—@
TGA at codon 342 has been reported 10 times in human
found in colon cancers. Future studies might include evaluation of tumors supports the experimental evidence that truncation of the last
HNPCC tumors for slippage mutations at repeat sequences, assess 50 amino acids does lead to a selection advantage in vivo.
mont of the frequency of microsatellite changes in cancers which Many mutations in the hydrophobic midregion change the confor
exhibit frequent deletions and insertions, and evaluation of mutations mation of the molecule, perhaps by partial denaturation. This exposes
in hMLHJ, hMSH2, and other mismatch repair genes. an epitope between amino acids 213—217,which is the target for
mAb24O, a monoclonal antibody that binds specifically to conforma
C. Determinationof Functionsand Propertiesof p53 tionally altered or denatured p53 (97). This antibody has been used as
a marker of altered p53 conformation in immunoprecipitation and
Mutational spectrum analysis complements in vitro studies of the immunoblotting studies. The importance of conformation in p53 func
relationship between p53 structure and function and suggests many tion has been demonstrated using truncated and mutant p53 molecules
experiments which can test the hypothesis that some mutants of p53 (including temperature-sensitive mutants whose conformation
alter its function and confer upon cells a growth advantage. In other changes from wild-type at 32°Cto mutant at 37°C)and by modulating
tumor suppressor genes (e.g., RB and APC), the most common ab wild-type conformation by oxidation. Molecules whose conformation
normalities found are deletions or nonsense mutations which elimi is altered by mutation or oxidation (109) lose sequence-specific DNA
nate protein expression, indicating that loss of their function promotes binding and transactivation activity; temperature-sensitive mutants
tumorigenesis (94, 95). The presence of mutated p53 protein in tumors exhibit this loss only at 37°C,when they exist in mutant conformation
suggests that cells acquire a selective growth advantage from not only (110, 111). Chimeric proteins, with the central p53 conformation
loss but also missense alteration of p53, frequently accompanied by region linked to foreign nuclear localization and transactivation re
loss or mutation of the second allele. Analysis of all p53 mutations in gions, maintain sufficient p53 transactivating properties to sup
human cancers reveals a nonrandom distribution within the p53 mol press growth of lung and colon carcinoma cells (103, 112), indicating
ecule, implying that specific mutant proteins possess different growth that the amino and carboxy termini contribute generic functions but not
stimulatory properties (Fig. 1). Identification of these locations has specificity.
focused research of p53 structure and function toward specific regions Humans and laboratory animals which develop tumors carrying p53
of the protein, such as exons 5—8.What is the functional importance of mutations may also develop antibodies against the protein, and this
these regions? Are mutations more frequent here because of the gain and phenomenon is now being investigated as a possible marker for
loss of specific functions? Are the types of p53 mutations seen in other diagnosis and prognosis. Anti-p53 antibodies occur in patients whose
regions of the molecule different than those in the conserved regions? tumors contain missense mutations, but they are usually directed
against epitopes in the amino and carboxy termini and not the
conformation-determining midregion, suggesting that the native pro
1. Structure-Function Relationships ofps3 Protein
tein can be immunogenic. Antibodies have been detected in the serum
Homologies have been noted between p53 domains and other of patients with carcinomas of the bladder (28%), lung (15%), breast
proteins, providing clues to both structure and function. The confor (12%), esophagus (16%), liver (25%), pancreas (19%), and lympho
mations of wild-type and some mutant p53 proteins and their rela mas (5%) and at least seven other types of cancer, as well as in
tionship to the critical functions of sequence-specific DNA binding patients with precursor lesions such as Barrett's esophagus (5%; Refs.
and transactivation have been examined using full-length and trun 113—118). In several anecdotal cases, the antibodies have been de
4864

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATION5AND CANCER

tected in serum samples in high-risk patients (Barrett's esophagus, patterns of mutation clusters in conserved domains Il—Vand else
tobacco smokers, and occupational vinyl chloride exposure) several where adds to the evolutionary data and provides clues to amino acids
years before the clinical detection of cancer.4 Hypotheses regarding of greater and lesser importance.
anti-p53 antibodies include: (a) does the immune response affect The predominance of missense over nonmissense mutations begins
carcinogenesis, e.g., does presence of anti-p53 antibody affect rates of and ends abruptly at codons 130 and 286 (Fig. 1), suggesting that
progression to malignancy of preneoplastic lesions? (b) can the de these are the boundaries of the conformation-determining region. This
tection of anti-p53 antibodies be used as a screening test in patients at is shorter than the span identified by in vitro and evolutionary studies
high risk for specific cancers (e.g., smokers, Barrett's esophagus, and as important for DNA binding specificity (amino acids 93—293)and
occupational carcinogens)? and (c) can the presence of serum anti-p53 suggests that changes to amino acids 93—130may not alter p53
antibodies identify individuals most responsive to immunotherapy? conformation significantly. This region contains three short (3-sheets
and an unstructured loop, of which only one amino acid (Lys 120)
2. Distribution of Mutations and the Relationship to p53 interacts with DNA (97). Forty-four codons are relative hotspots for
Conservation through Evolution missense mutation, arbitrarily defined as more than 12 mutations
reported in the database (>0.5% of all mutations); 40 of the 44 hotspot
The clonal selection theory of carcinogenesis posits a growth ad
amino acids (91%) are conserved in all 8 species. This represents only
vantage for clones containing mutant p53; therefore, sites where
42% of the 96 conserved AM, suggesting that mutations at many
missense mutations frequently occur presumably identify amino acids
other conserved sites confer little or no growth advantage; this hy
whose replacement leads to loss of p53 suppressor function and
pothesis can be tested by functional assays of mutants created by
perhaps gain of oncogenic function. Previous reviews have noted that
site-directed mutagenesis.
major mutational “hotspots―
occur in four of five domains labeled I—V
Some amino acids in the previously identified hotspot domains are
(I, amino acids 13—19; II, amino acids 117—142; III, amino acids
affected much more frequently than others, and within each domain
171—181;IV, amino acids 234—258;and V, amino acids 270—286),
are some highly conserved amino acids which are rarely mutated. For
which are highly conserved in evolution and are presumed to contain
amino acids essential for p53 function (Fig. 1; Ref. 51). Of the 393
example, only 3 of the 25 amino acids in domain II (132 Lys, 135 Cys,
amino acids in human p53, 96 (24%) are conserved in 8 species for and 141 Cys) are relative hotspots. All three of these residues begin or
which the p53 sequence is reported. By comparing all hotspot sites to end (3-strands, although other amino acids which flank (3-strands are
the pattern of AA conservation in all species, we can address the not hotspots (97). Of the six hotspot codons which contain 23% of
hypothesis that common mutations affect conserved amino acids reported p53 mutations (Fig. 1), five contain CpG dinucleotides
important to p53 function. (codons 175, 245, 248, 273, and 282 but not codon 249), suggesting
Fig. 1 demonstrates the distribution of missense and nonmissense a shared mechanism (deamination of 5-methylcytosine) for the prey
mutations in the database. Missense mutations are the most common alent G:C—@ A:T transitions reported at these sites. The resulting
type (79% of mutations) in the conserved midregion but are uncom mutants at some of these codons have been shown to alter p53 transcrip
mon (23%) in the amino and carboxy termini, where nonmissense tional activity and growth suppression (section II.D.5.F). Only two of
mutations (nonsense, frameshift, splicing, and silent mutations) pre these amino acids (Arg 248 and 273) directly contact DNA, but all appear
dominate. The rarity of missense mutations in the amino and carboxy to be Critical in stabilizing the protein-DNA interface (97).
termini suggests that in these regions the integrity of a single amino Some relative hotspots for missense mutations occur outside the
acid residue is not essential to function or conformation. Of note, no previously recognized conserved domains, marking three unrecog
mutations have been reported in conserved domain I. Nonconserved nized regions of potential functional significance (Fig. 1). Between
residues within exons 5—8also show a low proportion of missense domains II and III lies a stretch of 14 amino acids, 151—164,of which
mutations, supporting the hypothesis that substitutions of noncon one-half are conserved and at which 167 missense mutations (and 33
served amino acids do not provide cells with a positive selection nonmissense mutations) have been reported. This region forms a
advantage. The most commonly mutated amino acids in the DNA (3-strand which is not adjacent to DNA (97). Its DNA sequence is rich
binding region are close to the protein-DNA interface (97). The in G:C base pairs, often repetitive. No codons in this span are mutated
distribution of deletions and insertions in p53 appears to be related as frequently as the six hotspots noted above. Despite the fact that five
primarily to DNA sequence repeats (Fig. 1; section II.B.2) and not to CpG sites occur here (section II.C.2), G:C—t'A:T transitions are rare.
characteristics of the protein; as predicted, they occur frequently at This suggests that these sites may not be methylated or that the AA
both conserved and nonconserved codons. substitutions resulting from deamination (three of which are silent)
We analyzed patterns of evolutionary conservation using the Mul offer little growth advantage, perhaps because they are not adjacent to
tiple Alignment Construction & Analysis Workbench (MACAW) DNA (97). These hypotheses also are testable using in vitro mutagen
program (119). This method identifies the most highly conserved esis, evaluating the mutants for transactivation, growth suppression,
portion as amino acids 97—292(MP-scores, 359.4 and 471.2 for two and other properties, as has been done for other CpG site mutations
contiguous conserved regions). This area corresponds almost exactly (section II.D.5.F).
to the region which determines sequence-specific DNA binding and Two other highly mutated areas lie between domains III and IV.
transactivation (section II.C.1). When lower stringency for sequence Amino acids 193—200are almost completely conserved; missense
similarity is allowed, secondary conserved blocks are noted between mutations are common at the first three codons (His-Leu-Ile). Codons
amino acids 17—29(MP-score, 40.4), corresponding to the transacti 213—223also define a region of high conservation, with missense
vation region identified through Gal4 binding experiments, and amino mutational hotspots at codons 216 (Val) and 220 (Tyr). Two other
acids 324—352(MP-score 114.3), encompassing the oligomerization conserved moieties which are frequently altered are 205 Tyr and 266
domain. Higher stringencies fail to identify subregions within amino Gly. Of these mutational hotspots, five contain a single favored
acids 97—292,including domains II—V,that are significantly more mutant: 157 G:C—@
T:A, Val>Phe; 194 G:C—*
T:A, Lys>Arg; 195
homologous than the sequence as a whole. Detailed analysis of the T:A—*C:G, Ile>Thr; 205 A:G-+ G:C, Tyr>Cys; and 220 A:T-+ G:C,
Tyr>Cys. Codon 157 mutations, especially G:C—*T:A transversion,
4 G. Trivers and T. Soussi, personal communication. are especially frequent in lung cancer, perhaps indicating a tobacco
4865

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

specific carcinogen (section II.D.5.C) or a growth advantage peculiar of mutation patterns of p53 and other genes in clinical tumors can be
to bronchial cells. an important part of this process.
Taken together, these specificities do not exclude that certain re Some cancers are thought to arise from a single cell, while for
gions of DNA may be susceptible to specific mutagenic events, with others a “field
defect―of the entire mucosal surface is thought to result
only favored mutants providing a selection advantage. The finding in multiple clones of transformed cells and multiple primary neo
that tyrosine residues are affected at several hotspots suggests that plasms (130). Field cancerization is thought to occur in epithelia of the
they are important to p53 function. Potential reasons include contri upper aerodigestive tract, bladder, and ulcerative colitis-associated
bution to p53 conformation or modulation of phosphorylation status, colon (131—133).Careful identification of the stages of this process
although a mechanism has not yet been demonstrated. and molecular analysis at each stage can help illuminate the processes
Some other specific residues with potential importance are serines of clonal evolution in specific tumors. The multiplicity of p53 muta
9, 15, 37, 315, and 392, targets for phosphorylation by DNA-activated tional events indicates that identical mutations are unlikely to occur
protein kinase, p34cdc kinase, and casein kinase II. In vitro mutagen independently, and specific mutants may be used as clonal markers.
esis studies creating alanine residues which are not phosphorylated For example, the field cancerization hypothesis for head and neck
have implicated serine 15 in human p53 as playing a role in suppres squamous carcinoma is supported by molecular analyses which have
sor function; conflicting results have been found by altering serine found discordant mutations among nonneoplastic respiratory mucosa,
392 (120—122). That no mutations of these amino acids have been primary cancers, and second upper aerodigestive primary lesions,
reported is inconsistent with a strong selective advantage and argues suggesting that p53 is an important early target for mutations which
against the hypothesis that these residues contribute significantly to can occur at multiple mucosal sites (132, 133).
suppressor function.
Our conclusions regarding evolutionary aspects of p53 are: (a) the 2. Timing ofps3 Mutations in Human Cancer
most highly conserved region corresponds to the area of p53 respon
sible for sequence-specific DNA binding functions (amino acids 97— Several human cancers develop through multiple, morphologically
292); (b) the transactivation, nuclear localization, and oligomerization defined stages, such as the adenoma-carcinoma sequence in the colon
domains are only moderately conserved, provide generic functions and the dysplasia progression in the oral and bronchial mucosae. The
which are not p53-specific, and are not significantly functionally anatomically defined stages provide milestones for marking the tim
altered by single amino acid substitutions; and (c) based on mutational ing of p53 mutation, and there is evidence that some tumor types
spectrum analysis, essential conserved residues and favored mutants undergo early and others late p53 mutation. The issue has both
occur at previously described domains Il—Vand three additional mechanistic and practical implications, including the use of p53 as a
regions (1) codons 151—164,especially codons 151—152(Pro-Pro), potential marker for early diagnosis and as an intermediate biomarker
157—158(Val-Arg), and 163 (Tyr); (2) codons 193—195,especially for chemoprevention studies. This section reviews current data assess
the codon 194 Lys>Arg and 195 Ile>Thr mutants; and (3) codons ing early and late alterations in human tumors. Early is defined here
213—220,especially the Tyr>Cys mutant at codon 220. Two other as more than one morphological stage before invasion of the basement
residues which are both conserved and frequently mutated are codon membrane, which is the traditional hallmark of malignancy. Late
205, another tyrosine which often mutates to a cysteine, and glycine at means just before, or after, crossing the basement membrane. Alter
codon266.A searchof the SwissProt,PIR International,andtranslated ations in the p53 pathway (mutation or epigenetic inactivation) are
Genbank files found no other reported sequences homologous to these thought to be generally early events in cancers of the lung, esophagus,
three regions; therefore, their specific properties remain obscure. head and neck, breast, cervix, and stomach and generally late events
in cancers of the brain, thyroid, liver, and ovary (134—139).Cancers
D. Clues to Carcinogenesis of the colon, bladder, liver, and perhaps other organs may develop
1. p53 in the Context of Multistep Carcinogenesis through multiple pathways; p53 alterations may be early events in
some pathways and late events in others for these tumor types. Some
Is alteration of function of the p53 pathway a prerequisite for of the better characterized examples are discussed here.
neoplastic transformation? The frequency of p53 mutations argues for Squamous cell carcinomas account for 25% of lung cancers and the
a critical role, but for most tumor types mutations have been found in large majority of cancers of the upper aerodigestive tract. These
only 20—50% of cases, with a maximum of 81% in oral mucosa tumors usually progress through multiple, histologically recognizable
squamous cell, 70% in small cell lung cancers, and 68% in anaplastic/ stages of dysplasia (usually subdivided as mild, moderate, and se
undifferentiated thyroid cancer. Current techniques are unlikely to vere), carcinoma in situ, and transgression of the basement membrane,
miss more than 15—20% of coding sequence abnormalities. How which marks the beginning of invasive cancer. Despite the small size
frequent and important to functional abrogation are mutations in the of preneoplastic lesions, which frequently measure less than 100 p@m,
p53 promoter or unevaluated introns, epigenetic inactivation of p53, some genetic studies have been done, including allelic deletion, SSCP,
and/or alteration of genes or proteins downstream in the p53 pathway? and sequence analysis. However, the bulk of the data come from IHC
Is p53 only one of several pathways whose disruption is sufficient for using p53 protein accumulation as a surrogate marker for missense
neoplastic transformation? Support for this concept comes from the mutation (section II.A.1).
observations that both p53 and ras mutations are common events but Genetic and cytogenetic analyses of preinvasive bronchial lesions
often occur independently (67, 123—127),and either may be associ have shown that p53 mutations can precede invasion. Three reports
ated with aggressive tumor behavior (4, 128, 129). Does neoplastic (140—142) document missense mutations, p53 allelic deletion, and
transformation require a critical mass of genetic injury, in which p53 chromosome l'7p deletion in dysplastic mucosa associated with car
is a frequent but not essential casualty? In this scenario, loss of p53 cinoma. However, composite IHC data show p53 protein accumula
genomic stabilization properties would predispose a cell to an accel tion in 0% of normal bronchial mucosae, 7% of squamous metapla
eration in the rate of genetic damage and greatly increase the likeli sias, 25% of mild dysplasias, 32% of moderate dysplasias, 69% of
hood of neoplastic transformation and/or malignant progression. Re severe dysplasias, 57% of carcinomas in situ, 70% of microinvasive
search which attempts to unify molecular and cellular theories of carcinomas, and 76% of fully invasive lung carcinomas (140—144).
carcinogenesis should address these and other questions. Evaluation These results support a multistage model for squamous lung carcinoma in
4866

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

which p53 mutation occurs in 25% of the earliest neoplastic lesions, and tumors evolve from the low grade precursors and are subject to the
the large majority of mutations occur before invasion develops. same mutagens. As 45% of mutations are G:C—@ A:T transitions at
The timing of p53 alterations in the oral cavity, oropharynx, and CpG dinucleotides plus short deletions, the mutagens responsible for
larynx is addressed by at least 10 published reports, usually using IHC progressive genetic damage in these tumors may be endogenous
as a marker for putative missense mutation in small progenitor lesions. cellular processes (section II.D.5.F).
Several groups have reported p53 alterations (jositive IHC or muta Recent studies have correlated p53 mutation with metastasis and
tion) at or before the stage of severe dysplasia, including some other important but less well-studied tumor phenotypes, such as
histologically normal mucosa (132, 145—149).Future studies could angiogenesis (169). The regulation of specific genes which contribute
contribute to this area by providing additional histological detail to to these behaviors [such as MDR-1 (170) and thrombospondin (171)]
IHC studies, further molecular data, or correlating additional factors is one area in which the known transcriptional activating function of
(e.g., DNA content and other genetic changes) with p53 alterations. p53 could contribute to acquisition of the metastatic phenotype. Since
The timing of p53 mutation in squamous neoplasms in the esophagus p53 regulates genomic stability, and prevents cell cycle entry in
appears similar to those in the oropharynx (150—152). response to DNA damage, loss of p53 function would be expected to
Sporadic colorectal cancers generally develop through an adenoma correlate with aneuploidy and increased proliferative rates, two fea
carcinoma sequence which has been extensively analyzed for alter tures often associated with tumor progression and poor prognosis.
ations in oncogenes and tumor suppressor genes (reviewed in Ref. Preliminary studies have noted a statistically significant association
153). Allelic deletion and p53 point mutation rarely occur until the between aneuploidy and p53 mutation for ovarian (172), colorectal
late adenoma/carcinoma boundary (154; reviewed in Ref. 153). In (173), and gastric (54) cancers and a positive trend in esophageal
contrast, in ulcerative colitis, an inflammatory condition predisposing cancer (53). Other properties which are being studied for a relation
to colorectal cancer in non-polypoid, non-adenomatous, dysplastic ship to p53 function include radiosensitivity and hormone responsive
mucosa, p53 mutation and allelic deletion are found in the earliest ness (161, 174—176).
recognized dysplastic lesions (131, 155, 156). It is notable that several These data support a critical role for p53 in tumor progression. The
IHC studies find protein accumulation at earlier stages of sporadic results to date of p53 mutation analysis indicate that the details of this
colon cancer, suggesting a nonmutational mechanism of protein process are complex and probably mutant- and tissue-specific. Spe
accumulation during incipient neoplasia (157, 158). cific p53 functions potentially involved include the pathways of gene
transactivation, cell cycle checkpoint control, and programmed cell
death and suggest unbounded opportunities for in vitro testing. Future
3. p53 Genotype and Cancer Phenotype
research might address p53 mutation and mechanisms of tumor inva
The conventional method for describing the phenotype and predict sion, metastatic properties such as angiogenesis, and drug resistance.
ing the clinical behavior of a cancer has been histological grading as Such studies could clarify questions generated by clinicopathological
assessed by light microscopy of tumor cells. IHC, which identifies observations, which are often inconclusive due to unavoidable
specific cellular antigens, and other specialized staining techniques confounding variables or sample size limitations.
can yield more detailed phenotypes and sometimes clinically useful
information, but these traditional measures are merely crude descrip 4. Immortalization and Neoplastic Transformation:
tions of essential tumor behaviors. Correlation of phenotypes, such as In Vitro Features
histological grade or proliferative rate, with precise molecular events,
such as alterations of p53 or other genes (e.g., metastasis-related gene Much in vitro carcinogenesis research uses cultured human cells
nm23, RB, and ras), promises to expand our understanding of tumor derived from normal or malignant tissues (177). Tumor cell lines often
behavior and perhaps foster new paradigms of risk assessment, preven differ from the corresponding in vivo normal tissues in properties such
tion, early detection, determination of prognosis, and antineoplastic ther as protein or marker expression, response to growth modulators, and
apies. oncogene abnormalities. p53 mutations are found twice as frequently
The relationship of p53 mutations to tumor grade and stage has in all cell lines compared with all clinical tumor specimens (53% vs
been evaluated in many tumor types. Associations of p53 mutation or 26%); 15 of 20 tumor types have an excess of mutations in cell lines.
positive IHC staining with higher grade and more advanced stage are Potential explanations for this observation include: (a) mutations
frequent, although sample sizes are often too small to draw statisti might be underdetected in clinical specimens due to technical reasons;
cally valid conclusions from individual studies. A relationship be (b) mutations could arise frequently during the process of establishing
tween p53 mutation and advanced tumor stage has been noted for a cell line and, therefore, reflect in vitro mutagenic events which are
cancers of the endometrium (159), cervix (159), ovary (138, 139, not relevant to in vivo carcinogenesis; (c) clones with mutated p53
160), liver (136, 137), prostate (161), and bladder (162, 163), indi which comprise a minority of the in situ tumor could have an in vitro
cating that for these tumors p53 mutation may be a late event con growth advantage over clones without such mutations. According to
tributing to tumor progression, but not for lung (67, 164, 165), head this hypothesis, the mutations would reflect in vivo mutagens, but p53
and neck (132, 149, 166), and breast (134, 135), in which mutations mutation would not be an important early event. The genetic insta
are known to occur in early stage tumors and precursor lesions. bility triggered by loss of p53 function would lead eventually to clonal
Studies of brain neoplasms are archetypes for establishing the role evolution and tumor progression.
of p53 in tumor progression and clonal dedifferentiation. Low grade Mutational spectrum analysis supports the hypothesis that cell line
gliomas, which are indolent and often curable by resection, may mutations do reflect in vivo mutagens. Distribution of mutation sites
transform to high grade, aggressive, rapidly lethal glioblastomas. Two and percentages of each type of base substitution, frameshift muta
studies evaluating both low grade tumors and recurrent high grade tions, and CpG site mutations are similar in clinical tumors and cell
tumors document the acquisition of p53 mutations in five of six lines. Experience in the establishment of cell lines from cervical
tumors after transformation from low to high grade, although low (178—180),thyroid (181, 182), nasopharyngeal (183, 184), and other
grade tumors may also contain p53 mutations (167, 168). The p53 cancers strongly suggests that cells with abnormal p53 function have
mutational spectra of low grade astrocytomas versus high grade a selective growth advantage in vitro. This advantage also may exist
glioblastomas are similar, supporting the concept that the high grade in athymic nude mice, as suggested by the higher prevalence of p53
4867

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

mutations in xenografts than clinical specimens of head and neck supports a role for the p53 biological pathway in tumorigenesis.
carcinomas (183, 185). This hypothesis warrants further investigation Evaluation in melanomas of other genes and proteins which interact
into the features of in vitro cell growth and what properties of with and stabilize p53 (e.g., Mdm2) or act downstream in the p53
wild-type p53 may be inhibitory. pathway [e.g., WAF1/Cipi (63)] would be informative. The p1611――
gene, which like WAF1/Cipi encodes an inhibitor of cyclin-depend
5. Carcinogens and Specific Organ Carcinogenesis
ent kinases, is another candidate tumor suppressor gene involved in
melanomas and other cancers (122, 195, 196).
The narrow mutational spectra exhibited by some mutagens has B) Hepatocellular Carcinoma, Aflatoxin, and Hepatitis. The
popularized the idea that each agent might leave a specific identifying viral-chemical etiology and multistage pathogenesis of HCC are ar
“fingerprint―
of site and type of DNA damage (35). It is probably chetypal of human carcinogenesis. It is estimated that 75—90%of
more realistic to expect that carcinogens will produce mutation pat HCC cases are attributable to HBV (197). The relative risk of HCC is
terns which are characteristic and instructive but not as unique as elevated in viral hepatitis carriers with chronic active hepatitis (198).
fingerprints. Table 2 contains the mutational spectra of all cancers for These findings suggest that cell proliferation and/or inflammatory
which more than 20 mutations were reported at the time of analysis of response associated with chronic active hepatitis are the critical fac
the database, in descending order of mutation prevalence. tors responsible for the increased probability of neoplastic transfor
A) Skin Carcinoma and UV Light. A clear example of the value mation of the precursor cells of HCC.
of mutational spectrum analysis in identifying carcinogen-specific HBVDNAintegratesintoHCCcellsat randomsitesin thegenome.
mutations is seen in skin carcinomas, the most common type of human It contains the X gene, which codes for a protein (HBX) that modu
cancer, in which the role of UV light as the major carcinogen is lates the transactivation of many cellular genes and is a candidate viral
unquestioned. Exposure to UV light increases the risk of both basal oncoprotein (199). Transgenic mice containing HBX gene in their
cell and squamous cell carcinoma, as well as the less common but germline have an increased frequency of HCC (200). HBX protein
more lethal melanoma (186, 187). This physical mutagen produces binds with p53 in vitro and in vivo (201), inhibits p53 sequence
distinctive pyrimidine dimers that, if unrepaired, can produce tandem specific DNA binding and transactivation activities, partially disrupts
mutations, most characteristically CC—tTT transitions. Tandem p53 oligomerization, and prevents p53 binding to transcription-repair
dipyrimidine mutations are infrequently caused by mutagens other coupling factor ERCC3 (202). These mechanisms of epigenetic p53
than UV light (188, 189) and have rarely been observed in noncuta inactivation could account for the transactivation properties of HBX.
neous malignancies, i.e., 10 in over 2400 tumors with p53 mutation. Correlation of p53 staining and mutation data with HBX protein
Thus, the observation that these tandem mutations are common in expression in HCCS could help assess the interactions between HBV
squamous cell (9%) and basal cell (12%) carcinomas of the skin (Fig. and p53.
2, B-D) directly incriminates both exposure to UV light as the cause AFB also is considered to be a significant etiological factor in
of damage to the p53 gene and the loss of its tumor-suppressor certaingeographicareas (e.g., southernAfricaand Asia),wherethis
function in the development ofthe cancers (190). The detection of rare mycotoxin is consumed in food contaminated by Aspergillus flavis
cells with p53 mutations at dipynmidine sites in sun-exposed nonma (reviewed in Ref. 203). Epidemiological studies conducted in the
lignant skin indicates that UV light initiated these mutations at the 1970s and 1980s provided statistical evidence that dietary consump
earliest stage of skin carcinogenesis (191). tion of AFB was positively correlated with incidence of HCC and
Mutations at dipyrimidine sites in skin carcinomas show a nonran suggested a synergistic interaction with alcoholic beverage consump
dom distribution among sites within the p.53 gene. Of the 30 CC:GG tion (204) and chronic active viral hepatitis (205—207).
dinucleotides (18 on the nontranscribed strand) in conserved domains, Many investigators have reported data on the presence of chronic
transitions are common (5 or more) at only five (codons 151—152, HBV infection and p53 mutations in the same tumors (208—216).
247—248,178—179,278, and 281—282);frequency of mutations also Their results indicate that HBV infection alone does not influence the
varies at other dipyrimidine sites (CT:GA, TT:AA, and TC:AG). rate of p53 mutation and that AFB exposure is the most important
Recent work shows that rates of cyclobutane dimer repair vary among influence on mutation prevalence (28 versus 29% mutation prevalence
codons within p53 (192). Comparison of the frequency of transition in HBV-positive versus HBV-negative cases in low AFB exposure
mutations to rates of repair shows that 94% of C:G—@ T:A or CC: areas; 54% prevalence in high AFB exposure areas, where almost all
GG—TT:AA transitions occur at sites where more than 10% of cases were HBV positive). The combination of these exposures may
cyclobutane pyrimidine dimers remain 24 h after UV treatment, be synergistic in causing mutation, but data are insufficient for defm
suggesting that slow repair of these dimers is responsible for some of itive conclusions (28 of 49 AFB positivefHBV-positive cases versus 1
the site specificity of UV-induced mutations. The dipyrimidine sites of S AFB positive/HBV-negative cases contain mutations; Refs. 208,
which are frequently mutated in XP-C patients were not evaluated in 211, and 216).
this study. It remains unknown whether variations in site-specific The results from a recent prospective cohort study of 18,244 people
DNA repair in XP patients follow the same patterns as normal provide convincing evidence that AFB has an etiological role in
individuals and whether these patterns explain the different dipyrimi hepatocellular carcinogenesis and indicate a synergy between HBV
dine mutation site specificity seen in XP-C. Mutant selection due to and AFB (217, 218). This nested case-control analysis shows statis
functional changes of specific amino acid substitutions also is likely to tically significant associations among the presence of AFB and its
play a role in determining this specificity. To further define the metabolites in urine specimens, serum HBV surface antigen positiv
mechanisms responsible for site specificity of mutations, study of the ity, and HCC risk. The finding of the promutagenic AFB-N7-guanine
functional consequences (e.g., transactivation) of the commonly adduct in the urine is further evidence that AFB has been activated to
found mutants in skin and other cell types would be of interest. its electrophilic ultimate carcinogenic metabolite, AFB 8,9-oxide.
Whereas p53 mutations are common in basal cell carcinoma and Human hepatocytes in vitro can enzymatically activate AFB to its
squamous cell carcinoma, they are infrequent in melanomas (Table 2). 8,9-oxide (219); the interindividual variation in forming the AFB-N7-
Since sunlight also is a risk factor for melanomas (reviewed in Ref. guanine adduct may be 10-fold or greater (220). Since several iso
193), the rarity of p53 mutations is surprising, although the frequent forms of cytochrome P450 enzymes can activate AFB (221), the
accumulation of p53 in the nuclei of these tumor cells (59, 186, 194) interpretation of pharmacokinetics data and its use in risk assessment
4868

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

will be complicated. The mutational spectrum of AFB (222, 223) has prevalent mutation is G:C—@'T:A transversion with a predominance of
been established in experimental systems; G:C—t@
T:A transversions guanine residues on the nontranscribed DNA strand (Fig. 2, H-K), and
are the most common base substitutions (224—226). The high fre the frequency of transitions at CpG sites (9%) is lower than in almost
quency of AGG—'AGT transversions on the nontranscribed strand at all other cancers. This spectrum is consistent with data for several
p53 codon 249 in HCCs from areas of China and Mozambique with different types of chemical carcinogens found in tobacco smoke. The
a high HCC incidence could be due to the high mutability of the third highly mutagenic metabolites of PAHs such as BP preferentially
base of codon 249 by AFB and/or a selective growth advantage of attack deoxyguanines and lead to mutations (Table 1), and one of the
hepatocyte clones carrying this specific 249@ mutant in liver quantitatively minor adducts from the tobacco-specific N-nitrosamine
chronically infected by HBV. 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone also leads to
The preferential mutability hypothesis has been tested by Aguilar et G:C—@
T:A transversions (233).
a!. (227), who found that in a human liver cell line exposed to AFB, Analysis of p53 mutations in the database in relation to smoking
the third base in codon 249 is preferentially but not exclusively history yields statistically significant conclusions consistent with the
mutated compared to immediately adjacent codons, suggesting that experimental data. The frequency of p53 mutation and of G:C—@ T:A
both preferential mutability and clonal selection are involved in hu transversion on the nontranscribed DNA strand are positively cone
man hepatocellular carcinogenesis. This highly sensitive and specific lated with lifetime cigarette consumption (234). Compared with non
genotypic mutation assay also can be used to determine the p53 smokers, smokers have significantly lower frequencies of all transi
mutation load in nontumorous liver tissue from donors living in tions (35 versus 69%; P < 0.05), G:C—*A:T transitions (21 versus
geographic areas of high HCC incidence in which urinary AFB and/or 69%; P < 0.001), and higher rates of G:C—*T:A transversions with a
macromolecular-AFB adducts have been detected. A pilot study using DNA nontranscribed strand bias (29 versus 0%; P < 0.01; Fig. 2, L-M;
a genotypic mutation assay has compared the load of codon 249scr Ref. 235). These observations are consistent with the model of pref
(AGG->AGT) mutant cells in nontumorous liver tissues from HCC erential repair of carcinogen-induced damage on the transcribed DNA
patients from Qidong, People's Republic of China, urban Thailand, strand (75, 236). Cigarette smoke and some of its components have
and the United States (228). The loads of codon 249@Tmutations were been shown in vitro (229, 237) to increase mutation frequency and
elevated in specimens from Qidong, slightly elevated in one biopsy cause defects in DNA repair, which may contribute to the p53 muta
from Thailand, but did not exceed background levels in specimens tion spectrum. These results also have implications in the current
from the United States. These results indicate a positive association controversies concerning the relative risk of lung cancer in nonsmok
between loads of codon 249ser mutation and dietary exposure to AFB. ers exposed to environmental (passive) tobacco smoke. Molecular
They also suggest that, in some populations, p53 mutation occurs analysis would contribute important data to the debate by determining
early in hepatocarcinogenesis, and clonal expansion of these mutant whether critical mutations in lung cancers from these individuals
cells occurs in the nonmalignant tissue. A number of hypotheses could show a spectrum similar to smokers (a dose-response increase in
account for a selection advantage for these mutant cells, including G:C— T:A transversions with exposure to environmental tobacco
altered responses to growth factors or resistance to programmed cell smoke), although a large sample set may be required to detect changes
death induced by hepatitis viral infection. These and other hypotheses can in the spectrum.
be tested by in vitro models using human hepatocytes (219) and clinico Evidence exists of potential differences in pathways of carcinogenesis
pathological studies, such as correlation of aflatoxin-DNA adducts with among histological types of lung cancer. In vitro studies of lung cancer
codon249G:C—*
T:Atransversionsin individualpatients.Thegenotypic differentiation support a pluripotent cell of origin common to all types, or
mutation assay also might be applied to other tumor-specific hotspots. multiple cells which can differentiate by variable, often overlapping
C) Tobacco:Lungand OtherCarcinomas.Tobaccosmokingis pathways (238—240).Sufficient data exist to analyze the mutational
the leading preventable cause of cancer mortality, accounting for 40% spectra of squamous cell, large cell, adenocarcinoma, and SCLC (Fig. 2,
of cancer deaths in men and 20% in women. Cigarette smoke is a H-K). p53 mutations are found in 70% of SCLC and 47% of NSCLC,
complex mixture with thousands of constituents including tumor including 65% of squamous and 60% of large cell, but only 33% of
initiators, promoters, complete carcinogens, and cocarcinogens (229). adenocarcinomas (140, 234, 241—246),a prevalence which more closely
The contributions of many of these agents to carcinogenesis in the approximates that of other adenocarcinomas than of other lung cancer
lung and other organs are not known. Some of the compounds found histologies. The mutational spectra are notable for an excess of
in tobacco smoke, most notably N-nitrosamines and PAHs, have been A:T—@
G:C mutations in SCLC (11 versus 6%) and an excess of deletions
purified, tested in model systems, and found to be in vitro carcinogens and insertions in NSCLC (14 versus 4%). Subdividing NSCLC types
which induce characteristic DNA abnormalities (section I.B). This reveals that the prevalence of G:C—@ T:A transversions in large cell,
finding has encouraged mutational spectrum analysis in tobacco squamous, and small cell carcinomas is similar (43—49%),but they are
associated cancers to test the hypothesis that patterns of mutation can less common in adenocarcinoma. This finding is consistent with the
implicate specific agents in tobacco responsible for inducing human weaker association of adenocarcinoma with cigarette smoking (247—
cancers. Compounds of interest include N-nitrosamines, PAHs (e.g., 249), since G:C—÷T:A transversions are thought to result from bulky
BP), active oxygen species, aldehydes, and metals. The p53 muta molecules found in tobacco smoke. In adenocarcinomas, higher propor
tional spectra in tobacco-associated cancers show surprising intertu tions of G:C—@A:T (non-CpG site) and G:C—@ C:G mutations are seen,
mor variation, suggesting that many different tobacco-related carcin suggesting the presence of other carcinogens. Since adenocarcinomas are
ogens may be germane to human cancers. the most common cell type in women and squamous cell is less frequent,
1) Lung Carcinoma. Cigarette smoking is now thought to be analysis of mutational spectrum by gender (controlling for cell type and
responsible for 90% of lung carcinomas in men and 78% in women smoking history) may provide clues to different carcinogenesis pathways,
(230). A large body of epidemiological evidence has confirmed a hormonal influences, or differing susceptibilities to tobacco-associated
dose-response relationship proportional to duration and amount of carcinogens.
smoking (231, 232). How does mutational spectrum analysis further 2) Other Tobacco-associated Cancers. Epidemiological studies
define this association? have linked cigarette smoking to other cancers with attributable risks
p53 mutations are common in lung cancer, with the highest prey ranging from 20 to 90% (250). Mutational spectrum analysis of some
alence (70%) in SCLC and the lowest (33%) in adenocarcinomas. The of these tumors should reflect the effects of relevant tobacco-associ
4869

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

ated carcinogens. Despite the similarity of risk factors, the mutational Table 1), PAHs, N-nitrosamines, and alkylating agents are among the
spectra of these tumors differ from each other and differ significantly compounds which cause these substitutions in experimental systems.
from the lung cancer spectrum. This may be due to different constit Risk factors for squamous cell carcinoma in addition to tobacco smoking
uents of tobacco or nontobacco-related carcinogens. are alcoholic beverage consumption, and in China, dietary N-nitrosamine
Head and Neck and Esophageal Carcinomas: The combination of ingestion (which could account for non-CpG site G:C—* A:T transitions;
tobacco use and drinking alcoholic beverages is responsible for ap Ref. 252). Future studies could address the carcinogenicity and muta
proximately 75% of esophageal and 90% of head and neck cancers, tional spectra of these suspected carcinogens in cultured human esopha
most of them squamous cell (250). Two lines of evidence derived geal and upper aerodigestive cells or in animal models.
from the database implicate environmental carcinogens in head and As in the lung, adenocarcinoma of the esophagus is increasing in
neck carcinogenesis: (a) over one-half of all mutations in head and frequency relative to squamous cell; the last decade has seen a 3-fold
neck cancers are substitutions at base pairs which usually demonstrate increase in the incidence of adenocarcinoma and a 23% decrease in
nontranscribed strand bias (Table 3; section II.B.1), and 80% of these squamous cell carcinoma (253, 254). Adenocarcinoma of the esoph
occur on the nontranscribed strand; and (b) CpG transitions (sugges agus resembles carcinoma of the gastric cardia and usually arises
tive of endogenous events; section II.D.5.F) are uncommon (13%). within the intestinal metaplasia of Barrett's esophagus. Chronic gas
Compared with lung cancers, G:C—*T:A transversions are less com tric reflux is considered a risk factor for this condition, but risk factors
mon (18 versus 40%), while A:T—*G:C (14%) and non-CpG for neoplastic transformation are unknown. Few studies of the muta
G:C—* A:T (18%) transitions are prominent, as are deletions and tions in esophageal adenocarcinomas or Barrett's esophagus have
insertions (19%). Of 18 deletions and insertions, 12 can be localized been performed, but they suggest differences in mutagens from squa
to repetitive DNA sequences, consistent with the hypothesis that mous carcinoma. Of the mutations reported, 6 of 9 (67%) have been
mispairing during replication causes frameshifts (section II.B.2), and G:C—* A:T transitions at CpG sites, compared with a 14% incidence
two are large deletions at splice site codons, suggesting that the actual of CpG site mutations in squamous cell esophageal cancer. Evaluation
DNA alteration is an intronic point mutation. Chemical carcinogens of more adenocarcinomas will be important to generate statistically
such as PAHs in tobacco smoke may contribute to these frameshift valid comparisons with squamous carcinomas of the esophagus and
mutations (section II.B.2). with gastric cardia cancers.
Subdivision of head and neck carcinomas by anatomic region of Bladder Carcinoma: One-half of bladder carcinomas in men and
origin shows differing patterns of p53 mutations among the various one-third in women are attributable to smoking (255). A significant
primary mucosal sites (Fig. 2, E-G). In laryngeal and pharyngeal association has been found in superficial bladder cancers between p53
primaries, the mutation prevalence is relatively low (34%), but the nuclear staining by IHC and the number of cigarettes smoked (256).
spectrum resembles that of lung cancer. G:C—@' T:A transversions, The p53 mutational spectrum in bladder cancer is unusual with a high
primarily on the nontranscribed strand, are the most common mutation frequency of G:C—@ C:G transversions (21 versus 7% for all tumors)
(34%), and non-CpG G:C—@ A:T transitions also are common. Pri with a nontranscribed DNA strand bias. G:C—@ A:T transitions at both
mary tumors in the oral cavity have a high mutation prevalence (81%) CpG and non-CpG sites are the most common mutation, with non
and exhibit a spectrum in which non-CpG site G:C—*A:T transitions, transcribed DNA strand bias at non-CpG sites. One study has found a
A:T—*G:C transitions, deletions, and insertions predominate. Thus, in hotspot G:C—@C:G (Tyr>Lys) mutation at codon 280, but this has not
this class of cancers, subgroups which are often grouped together been confirmed. Two reports have compared the spectra in a total of 34
when addressing carcinogenesis and therapy demonstrate mutational smokers and 23 nonsmokers (163, 257) with similar spectra found in the
differences, which could be due to differences in carcinogen exposure two groups. Further epidemiological studies, including quantitative
(e.g.,solubletobacco-specific
N-nitrosamines
in oralcancers
versus smoking histories, are warranted to evaluate the data for more subtle
tobacco combustion products in laryngeal primaries) or variation in patterns.
carcinogen activation or DNA repair between distinct mucosal sites. D) Radiation. 222Radon is a colorless, odorless, inert gas, a natu
These hypotheses can be tested in the laboratory and by molecular rally occurring decay product of @8uraniumwhose decay emits a-par
epidemiological studies of mutational spectra of well-characterized tide radiation. Conclusive epidemiological evidence implicates occu
tumor types in populations with well-defined exposures. pational exposure to radon in uranium miners as a cause of lung
The epidemiology of primary NPC differs from other head and cancer (reviewed in Refs. 258 and 259). Early epidemiological studies
neck sites; these tumors are associated with dietary N-nitrosamines, of domestic radon and lung cancer risk exposure suggest that the
EBV, and perhaps genetic factors (183). Clinical NPCS rarely contain dose-response and relative risk for residential radon exposure is the
mutated p53 (4%). One hypothesis for this low frequency of mutation same as for occupational exposure (260—262). Radon exposure and
could be the epigenetic inactivation of p53 by EBV proteins (251). cigarette smoking increase risk synergistically, and analysis of human
The spectrum of the few p53 mutations in NPC tumors and lines is tumors may reflect interactions between radon and tobacco carcino
unusual, reflecting a lack of mutations known to be associated with gens (258, 259, 262).
tobacco-related carcinogens or N-nitrosamines (G:C—*T:A, non-CpG Two studies report the molecular analysis of the p.53 gene in lung
G:C—*A:T, and A:T—@ G:C substitutions). G:C—*
C:G transversions, cancers from uranium miners (140, 263). Missense p53 mutations
which comprise only 3% of p53 mutations at other head and neck sites were found frequently in both studies. A mutational hotspot (16 of 30
and 7% in all cancers, are common (5 of 17; 29% of mutations; mutations) at codon 249, AGG—@A@ Arg—+Met, was observed in
@ P 0.00001 versus other head and neck sites), and four of five are on miners from the Colorado Plateau who were exposed to high amounts
the nontranscribed strand, suggesting the action of a chemical carcin of radon. Three of the codon 249met mutations occurred in lung
ogen. Further molecular epidemiological studies are indicated to gen cancers from never smokers, implicating a non-tobacco-associated
erate a statistically meaningful database. carcinogen. Since these miners also were heavily exposed to other
The spectrum in esophageal squamous cell carcinomas also shows carcinogens found in the mines, the hypothesis that radon was respon
less frequent G:C—*T:A transversions than in lung carcinomas (16 sible for the specific base substitution requires further testing.
versus 40%), with corresponding increases in G:C—@' A:T (non-CpG Other sources of ionizing radiation which have been associated
site 22 versus 16%) and A:T—@G:C (14 versus 7%) transitions, as well with the development of hematological and solid malignancies in
as A:T—*T:A transversions (15 versus 5%). As discussed (section I.B; dude therapeutic radiation, atomic bomb explosions, and the radio
4870

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

logical contrast agent Thorotrast. Mutational spectrum analysis of suggests that this is the usual mechanism by which G:C—'A:T tran
cancersin these patientscouldindicateradiation-inducedevents.p53 sitions arise at CpG sites in human tumors. Examination of the p53
mutations in lung cancers from nine nonsmoking atomic bomb sur database for associated features such as amino acid selection and
vivors from Hiroshima, Japan, were compared to eight nonexposed strand bias of CpG transitions may yield further insights into this
controls; no differences in prevalence or mutational spectrum were process in tumor cells.
noted in this small study (235). The study of larger numbers and a The p53 coding region contains 39 CpG dinucleotides (i.e., 78 base
greater variety of radiation-related cancers would contribute more pairs) which are potential sites for G:C—@A:T transition from deamina
statistically meaningful data to the issue of radiation-induced mu tion. Transitions at these sites account for 24% of all p53 mutations
tagenesis in human cancer. described, with large variations among tumors. Seven sites (codons 175,
E) Cervical Carcinoma and Human Papifioma Virus. Epidemi 196, 213, 245, 248, 273, and 282) account for 90% of CpG transitions, or
ological studies established the link between cervical carcinoma and 21% of all p53 mutations. These mutations occur equally on both DNA
HPV types 16 and 18, and the molecularbasis for this association strands; strand bias in recognition and repair of G:T mismatches is thus
emerging from laboratory research features the p53 pathway. HPV unlikely to contribute to the selection of mutants.
types 16 and 18 both produce E6 protein, which complexes with Some mechanisms by which cells with some CpG transitions ac
cellular protein AP6 and degrades p53 in the cytoplasm (264, 265), an quire a selective growth advantage have been studied. At codons 248
example of epigenetic p53 inactivation. Mutation data would support and 273, both C:G—@ T:A and G:C—*A:T transitions commonly occur,
this hypothesis if p53 mutations were found in HPV-negative more implying that both amino acid changes (Arg>Trp and >Gln at 248,
frequently than HPV-positive cancers, and IHC would be expected to
Arg>Cys and >His at 273) result in a selection advantage. At codons
demonstrate no p53 staining in HPV-positive cases and nuclear stain
175 and 282, only one of the two mutations is selected (175, Arg>His
ing in HPV-negative cases with missense mutations. Early reports
but not >Cys; 282, Arg>Trp but not >Gln). Soussi and colleagues
found that mutation frequency was 100% in HPV-negative and 0% in
have constructed all potential transition mutants at CpG hotspot
HPV-positive tumors and cell lines (179, 266, 267), but this dramatic
codons 175, 248, and 273 and assessed conformation and in vitro
difference has not been validated in further studies of clinical tumors
transcriptional activation in the sarcoma cell line SAOS, which con
(159, 178, 268—271). These later studies confirmed the rarity of
tains no endogenous p53 protein.5 Transactivation, which is thought
mutations in HPV-positive tumors (4%) and lines (7%); mutations
to contribute to p53 tumor suppressor function, was assessed by
have been found in 7 of 39 of HPV-negative carcinomas now reported
transcriptional assays using the reporter gene CAT linked to an up
(18%; P 0.0007 versus HPV-positive tumors).
stream p53 binding region. Wild-type and mutant p53 constructs
These data indicate that dysfunction of the p53 pathway contributes
to cervical carcinogenesis, but unless mutations or other alterations in cotransfected into target cells with the reporter constructs were com
the p53 pathway are demonstrated in all HPV-negative tumors, it pared in their transcriptional activating abilities. The favored Arg>His
cannot be considered essential. Future studies might address the low mutant at codon 175 assumes the mutant conformation and transac
rate of mutations in HPV-negative tumors by searching for other p53 tivation properties, whereas the rare Arg>Cys mutant retains wild
pathway abnormalities, such as occult mutations, downstream gene type conformation and activity. Interestingly, both mutants at codons
abnormalities, or other epigenetic mechanisms of inactivation. 248 (Arg>Trp and >Gln) and 273 (Arg>Cys and >His) maintain
F) Endogenous Sources of Mutation. As discussed in section I.A, apparent wild-type conformation by antibody analysis but acquire
not all mutations are caused by exogenous carcinogens. Normal mutant transactivation function. These two amino acids are in direct
cellular events which are mutagenic include deamination of 5-meth contact with DNA (97). This evidence supports the studies (section
ylcytosine at CpG dinucleotides, DNA polymerase infidelity due to II.C.1) which connect p53 transactivation activity with growth sup
mispairing at repeat sequences or other mechanisms, and oxygen pression, but they imply that conformational changes as detected by
radical damage (reviewed in section l.A. and Refs. 14 and 272). these methods are not necessary for modulation of this function (at
Studies of constitutive cellular genes to determine baseline mutation least in SAOS sarcoma cells). Future studies could address other
rates in human and other mammalian cells have found rates of features of CpG site mutants which could eliminate p53 suppressor
between 10 ‘° and i0@ mutations/gene/cell generation (10, 85). activity. It is possible that the more infrequent mutants are function
However, these may underestimate the true mutation rate because of ally silent, and their presence in cancer cells does not contribute to the
insensitivity of the assays to some classes of mutations (e.g., only cancer phenotype.
hemoglobin variants with point mutations were detectable) or failure Thus, observational and experimental analysis supports the hypoth
of an assay's cell culture conditions to select clones with mutations esis that selection advantage due to altered transactivation and perhaps
but no growth advantage (10, 85). This background level of mutations other functions is responsible for the hotspot mutations at CpG
implies that populations will always exhibit a baseline rate of cancer, dinucleotides. Evidence argues against strand bias in mismatch repair
even in the absence of carcinogen exposure. The rates of endogenous of CpG transitions.
mutations may also be affected by exogenous events; future studies on 2) Cancers with High Rates of CpG Mutations, Deletions, and
the effects of carcinogens and cocarcinogens on deamination and Insertions. The reasons discussed above argue strongly that deami
deletion are warranted. Mutational spectrum analysis may be able to nation of 5-methylcytosine, an endogenous mechanism of mutation, is
define these baseline mutational frequencies by identifying the pro primarily responsible for G:C—@ A:T transitions at CpG dinucleotides
portions of mutations in cancer-related genes which are caused by (273). Similarly, deletions and insertions may result from misalign
endogenous processes. ment due to polymerase infidelity during replication, another endog
1) CpG Deamination. It cannot be proven that an individual enous mechanism. Transitions and frameshifts also may be induced by
G:C—@ A:T transition found at a CpG dinucleotide in a tumor results carcinogen-DNA adducts and other mechanisms (section II.B.1.A).
from deamination of 5-methylcytosine (as opposed to mispairing of an We hypothesize, however, that the sum of CpG transitions, deletions,
O°-methylguanine adduct with thymine, for example). However, the and insertions, and potentially of other mutations which may eventu
in vitro rates of spontaneous deamination, the clustering of spontane ally be linked specifically to endogenous processes, may be used as a
ous G:C—+ A:T mutations in experimental systems at sites where
cytosine is methylated, and the large database of observed mutations 5 K. Ory and T. Soussi, personal communication.

4871

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONSAND CANCER

Fig. 3. The mutational spectrum in hepatocellular Legend:


carcinomas varies by geographic origin in Asia (Qi
dong, n = 34; other mainland China, n = 11; Tai
wan, n = 26; and Japan, n = 123).

@,J-
JAPAN

crude indicator of the proportion of mutations which are an inevitable prevalent in factor IX mutations than in p53 germline mutations.
consequence of biological processes. Expansion of the data sets on germline mutations in other genes such
Transitions at CpG sites, deletions, and insertions comprise 37% of all as HPRT, APC, DCC, hMLHJ, and hMSH2 may suggest patterns
reported p53 mutations but 45% of mutations of brain, colon (Fig. 2N), which are now inapparent.
stomach, endometrial, thyroid, and hematological cancers. This suggests 3) Mutational Spectrum Differences among Populations. Al
that endogenous cellular processes may contribute to a significant frac though the focus of mutational spectrum analysis in HCC has been on
tion of these tumors. The mutational spectrum of LFS implicates 5-meth the role of codon 249@ mutations and AFB exposure, deeper scrutiny
ylcytosine deamination as the primary event in the formation of p53 of the data produces additional hypotheses. Since the spectrum in
germline mutations; 53% of germline mutations are G:C—+ A:T transi HCC is particularly dependent on a population's environmental cx
tions at CpG sites (Fig. 20). The second most frequent class of mutations posure, subclassification of the data by geographic region may reveal
(13%) are deletions and insertions, mostly of a single base pair at repeat patterns hidden by the AFB-related tumors. Fig. 3 depicts the muta
sequences, a pattern which is consistent with the slipped mispairing tional spectra of tumors from Qidong, other regions of mainland
mechanism. Thus, two-thirds of LFS cases can be attributed to endoge China, Taiwan, and Japan. While tumors from Qidong contain an
nous-type mutational events, compared with slightly more than one-third overwhelming preponderance of G:C—@T:A transversions (95%) due
of p53 somatic mutations found in human cancers. Mutations known or to association with AFB, Japanese and Taiwanese tumors harbor this
suspected to be associated with carcinogen exposure (A:T—t'G:C and type in only 26% of cases each. That both of these latter groups
non-CpG site G:C—* A:T transitions, G:C—@
T:A transversions, and tan exhibit some codon 249@1mutations suggests that AFB may be a risk
dem mutations) represent only 23% of LFS mutations versus 46% of factor even in populations not normally considered to be at high risk
somatic mutations (Fig. 2, A and 0). for dietary contamination. The remainder of the spectrum differs
Comparison to germline mutations of other genes associated with between the groups, however, and suggests other potential carcino
cancer or other diseases supports the concept that these putative gens. In Taiwanese patients, the most frequent base change for all
mechanisms of endogenous mutations are important sources of germ HCCs is A:T—@ T:A transversion (41%); the strand bias (all 11 mu
line mutation, but mutation patterns are not consistent and probably tations are A—@ T) suggests that an exogenous carcinogen is respon
reflect specific functional characteristics of the gene and protein. For sible. For Japanese patients, G:C—*A:T (at CpG and non-CpG sites)
example, germline mutations in the APC gene cause familial adenom and A:T—'G:C transitions (also with nontranscribed strand bias) are
atous polyposis, a syndrome in which multiple colon polyps, adeno both common, suggesting environmental carcinogens; deletions and
mas, and eventually carcinomas develop in all affected persons. insertions, linked to both endogenous and exogenous mutational
Germline and somatic mutations in APC result in truncation or com mechanisms, also are frequent. The identification of relevant carcin
plete loss of the protein in 96% of cases and in an altered protein in ogens with these spectra will be important. Only nine mutations have
only 4% (274). Thus, frameshift mutations are prominent (48% of been reported in HCC from Western countries; five are G:C—+ A:T
mutations), and CpG site G:C—+ A:T transitions, although the most transitions, three of which are at CpG sites. Thus, no pattern suggesting
frequent base substitution, comprise only 14% of all mutations. Sim carcinogen exposure is yet discernible.
ilar to p53 germline mutations, however, three-quarters ofAPC germ Environmental exposures or inherited characteristics which vary
line mutations are consistent with the endogenous processes of deami among populations may be important determinants of mutational
nation and slipped mispairing. The spectrum of mutations in the factor spectra in other tumors as well. Sufficient data are available to
IX gene, which causes hemophilia B, contains similarities and differ evaluate lung, gastric, and breast cancers for differences in spectrum
ences from the spectra in the tumor suppressor genes (273, 275). between Japanese and Western patients. Compared to Western pa
Approximately two-thirds are missense substitutions, less than 10% tients, Japanese lung tumors show a higher frequency of G:C—@ A:T
are deletions and insertions, and the remaining one-quarter are splice transitions (29 versus 20%, P 0.13; most are at non-CpG sites) and
site alterations and promoter mutations. Transitions at CpG sites are a lower frequency of G:C—+ T:A transversions (30 versus 43%,
common (approximately 36%), but other base substitutions are more P = 0.01), both with a nontranscribed strand bias. These differences
4872

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

are not found in SCLC but are seen in both adenocarcinomas markers. This can be accomplished by performing prospective studies
(G:C—s A:T transitions: 48 versus 20%, P = 0.02; G:C—+T:A trans in conjunction with large clinical trials to ensure statistical power,
versions: 14 versus 27%, P = 0.21) and non-adeno NSCLC (24 versus facilitate multivariate analyses to control for other established prog
20%, P = 0.004; 29 versus 55%, P 0.0003, respectively). Deletions nostic variables, and assess the responses to therapies in relation to
and insertions are more frequent in Japanese tumors (18 versus 10%, p53 status. The ability to perform IHC and PCR-based diagnostic
P = 0.22). These results suggest differences between Western and studies from archived tissue means that some of these studies could be
Japanese individuals, either in exogenous carcinogens or in cancer accomplished on materials from completed clinical trials which may
susceptibility factors such as germline polymorphisms in genes already have reached their end points, expediting the answers to some
involved in carcinogen metabolism (49). questions. Comparison of multiple molecular markers in the same
Most of the gastric tumors analyzed have been from Japan, where tumors can help to determine the relative importance to carcinogen
incidence rates are much higher than in Western countries. Epidemi esis of abnormalities in various cellular pathways.
ological studies in Japan have found an association between gastric Such studies can address both specific and general hypotheses regard
cancer and dietary salt and N-nitrosamines (276). Comparison of the ing carcinogenesis, for example: that tumors with p53 mutations might be
mutational spectra reveals that endogenous-type mutations are seen in more resistant to therapy-induced apoptosis; that tumors with mutations
72% of Western and 55% of Japanese tumors. More Western tumors are more likely to be associated with amplification ofother cancer-related
must be analyzed to provide a more statistically valid spectrum. The genes which influence therapy; whether all cancers have abnormalities of
major type of p53 mutation seen in Japanese but not Western tumors the p53 pathway, or of a specific combination of pathways; the relation
is A:T—@ G:C transition (20 of 69 versus 1 of 18, P = 0.03), with ship of response to therapies to molecularly determined tumor pheno
strand bias favoring T—@ C on the nontranscribed strand, the opposite types and genotypes; identification of subsets of patients with favorable
of the usual A:T—*G:C strand bias. Perhaps this represents the con and unfavorable prognoses that should be analyzed separately and cx
sequences of alkylthymine adducts from N-nitrosamines (277—279)in cluded from standard treatment protocols; and whether innovative ther
the Japanese diet. apies (e.g., antisense oligonucleotides and gene therapy) can be tailored
No specific exogenous carcinogens have been conclusively linked to specific molecular features of a tumor to improve the probability of
with development of human breast carcinoma. Although only 25% of successful outcome.
breast cancers contain p53 mutations, 15% of these are G:C—@' T:A
transversions, 16% are G:C—'A:T transitions at non-CpG sites, and
ifi. Conclusions
11% are A:T—@' G:C transitions, with nontranscribed strand bias of 89,
63, and 75%, respectively, suggesting that exogenous bulky carcino p53 mutational spectrum analysis contributes to a variety of mech
gens may play a role. The responsible carcinogens may vary among anistic questions in carcinogenesis, including integration of molecular
populations. Two reports of breast cancers from Japanese women and cellular hypotheses. Examples which are of ongoing interest
(135, 280) indicate a mutational spectrum different from tumors in include: what is the timing and sequence of the genetic changes
Western women. G:C—*T:A transversions are uncommon in Japanese contributing to neoplastic transformation, progression, and metasta
cases (1 of 48 versus 25 of 175, P = 0.02), while A:T—@ G:C sis? Does cancer always arise from a single transformed clone, or can
transitions are more common (8 of 48 versus 10 of 175, P = 0.01). multiple transforming events occur in some tissues? How do carcin
Thus, A:T—*G:C transitions with nontranscribed strand bias are ogen-host interactions, such as individual differences in carcinogen
prominent in the spectra of three different types of tumors in Japanese metabolism and DNA repair, influence an individual's risk of devel
patients (HCC, gastric, and breast). Whether this represents the effect oping cancer? Do different histological types of cancer contain char
of a regional environmental agent(s) or population differences in acteristic mutations reflecting a pattern of mutagen exposure, and do
carcinogen metabolism or DNA repair is an area for future investigation. cancers of similar histological types in different organs share these
Transitions at CpG sites, deletions, and insertions are common in both patterns? Are abnormalities of the p53 pathway critical for all tumors?
populations, suggesting a similar contribution of endogenous mutations. In cancers with low rates of p53 mutation, is p53 inactivated by
These analyses suggest that molecular studies with careful identi epigenetic events, or is its function interrupted by changes in other
fication of geographic and ethnic features of populations can make a genes which participate in p53-mediated pathways?
significant contribution to carcinogenesis research. Future research The emerging field of molecular epidemiology promises important
should evaluate statistically valid sample sizes in order to address contributions to cancer research, but full realization of its potential
specific hypotheses of differences in carcinogenesis among popula will require careful adherence to the tenets of both laboratory science
tions. For example, studies of Japanese populations in different geo and epidemiology. This will require increased interaction between
graphic regions (Japan, Hawaii, and mainland United States) have laboratory researchers, clinicians, and epidemiologists in order to
demonstrated differences in cancer rates. Molecular studies on these design studies which can adequately address important hypotheses.
cancers should be done to search for changes in mutational spectra, For example, published reports of the molecular features of cancers
which would suggest that environmental carcinogens differ among (and other diseases) should include important epidemiological data
geographic regions for several tumor types. such as gender, age, ethnicity, geographic origin, and, when appro
4)p53 Mutation and Prognosis. Since tumors with p53 mutations priate, associated medical conditions, tobacco smoking, and occupa
often behave more aggressively, the value of p53 as a prognostic factor tional or other exposures. Large scale clinical trials which include data
has been addressed. Studies have implicated p53 protein expression as an on the relevant variables can provide tumor tissue for molecular study.
independent prognostic factor in carcinomas of the breast, stomach, This may be accomplished using archived materials from past studies;
colon/rectum, bladder, and NSCLC (reviewed in Ref. 129). future trials could collect and preserve tissue specifically for molec
Future studies might integrate hypotheses of carcinogenesis into ular analyses. Attention to specimen handling at all times would
studies evaluating clinical outcomes. Since p53 mutation may corre minimize tissue degradation and maximize the reliability of laboratory
late with other indicators of poor prognosis, such as aneuploidy, gene findings.
amplification, and high proliferative fraction (section II.D.3), it will Mutational spectrum analysis and molecular epidemiology can be
be important to adhere to careful epidemiological methodologies to applied in innovative ways to both generate and address basic and
assure that p53 status adds information to established prognostic practical hypotheses including the areas of chemical, physical, and bio
4873

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

logical carcinogens, risk assessment, host factors, control of cell and carcinogenesis. Nature (Land.), 363: 558—561, 1993.
30. Yuspa, S. H., and Poirier, M. C. Chemical carcinogenesis: from animal models to
tissue behaviors, mechanisms of subcellular communication, protein
molecular models in one decade. Adv. Cancer Res., 50: 25—70,1988.
function, DNA replication, transcription, translation, and evolution. 31. Horsfall, M. J., Gordon, A. J., Bums, P. A., Zielenska, M., van der Vliet, G. M., and
Glickman, B. W. Mutational specificity of alkylating agents and the influence of
Acknowledgments DNA repair. Environ. Mol. Mutagen., 15: 107—122,
1990.
32. Strauss, B. S. The “A rule―of mutagen specificity: a consequence of DNA polym
erase bypass of non-instructional lesions? Bioessays, 13: 79—84, 1991.
We appreciate discussion and comments from our colleagues, including
33. Basu, A. K., and Essigmann, J. M. Site specifically modified oligodeoxynucleotides
Ettore Appella, Mark Boguski, Kathleen Forrester, Stephen Friend, S. Perwez as probes for the structural and biological effects of DNA-damaging agents. Chem.
Hussain, David Lane, W. Edward Mercer, Ruggero Montesano, Moshe Rca. Toxicol., 1: 1—18,1988.
Oren, Jennifer Pietenpol, Varda Rotter, Thierry Soussi, Glen Trivers, Bert 34. Skandalis, A., and Glickman, B. W. Endogenous gene systems for the study of
Vogelstein, Xin Wang, and Stuart Yuspa. The expert graphic and editorial mutational specificity in mammalian cells. Cancer Cells, 2: 79—83, 1990.
35. Vogelstein, B., and Kinzler, K. W. Carcinogens leave fingerprints. Nature (Land.),
assistance of Dorothea Dudek and the assistance of Peter Shields with 355: 209—210,1992.
statistical analyses are also appreciated. 36. Sarasin, A. Use of shuttle vectors for the analysis of DNA damage processing in
mammalian cells. Mutat. Res., 220: 101—106,
1989.
37. Miller, J. H. Carcinogens induce targeted mutations in Escherichia co!i. Cell, 31: 5—7,
References 1982.
38. Mawr, M., Gordon, A. J. F., Bernelot-Moens, C., and Glickman, B. W. Comparison
1. Hollstein, M., Sidransky, D., Vogelstein, B., and Harris, C. C. p53 mutations in of the mutational specificities exhibited by BPDE in Escherichia co!i and CHO cells.
human cancers. Science (Washington DC), 253: 49—53,1991. in: M. W. Lambert and J. Laval (eds.), DNA Repair Mechanisms and their Biological
2. Levine, A. J., Momand, J., and Finlay, C. A. The p53 tumour suppressor gene. Nature Implications in Mammalian Cells, pp 205—231.New York: Plenum Publishing Corp.,
(Land.), 351: 453—456, 1991. 1989.
3. Harris, C. C. The p53 tumor suppressor gene: at the crossroads of molecular 39. Grosovsky, A. J., de Boer, J. G., de Jong, P. J., Drobetsky, E. A., and Glickman, B. W.
carcinogenesis, molecular epidemiology and cancer risk assessment. Science (Wash Base substitutions, frameshifts, and small deletions constitute ionizing radiation
ington DC), 262: 1980—1981, 1993. induced point mutations in mammalian cells. Proc. NatI. Acad. Sci. USA, 85:
4. Harris, C. C., and Hollstein, M. Clinical implications of the p53 tumor-suppressor 185—188, 1988.
gene. N. EngI. J. Med., 329: 1318—1327, 1993. 40. Liber, H. L, Call, K. M., and Little, J. B. Molecular and biochemical analyses of
5. Vogelstein, B., and Kinzler, K. W. p53 function and dysfunction. Cell, 70: 523—526, spontaneous and X-ray-induced mutants in human lymphoblastoid cells. Mutat. Res.,
1992. 178: 143—153,1987.
6. Montenarh, M. Functional implications of the growth-suppressor/oncoprotein p53. 41. Yang, J. L., Chen, R. H., Maher, V. M., and McCormick, J. J. Kinds and location of
Int. J. Oncol., 1: 37—45,1992. mutations induced by (±)-7@,8a-dihydroxy-9a,10a-epoxy-7,8,9,10-tetrahydro-ben
7. Lindahl, T. Instability and decay of the primary structure of DNA. Nature (Land.), zo[a]pyrene in the coding region of the hypoxanthine (guanine) phosphoribosyltrans
362:709—715, 1993. ferase gene in diploid human fibroblasts. Carcinogenesis (Land.), 12: 71—75,1991.
8. Kunkel, T. A. Nucleotide repeats. Slippery DNA and diseases. Nature (Land.), 365: 42. Albertini, R. J., Sullivan, L. M., Berman, J. K., Greene, C. J., Stewart, J. A., Silveira,
207—208,1993. J. M., and O'Neill, J. P. Mutagenicity monitoring in humans by autoradiographic
9. Holliday, R., and Grigg, G. W. DNA methylation and mutation. Mutat. Res., 285: assay for mutant T-lymphocytes. Mutat. Res., 204: 481—492,1988.
61—67,1993. 43. Cariello, N. F., Craft, T. R., Vrieling, H., Van Zeeland, A. A., Adams, T., and Skopek,
10. Strauss, B. The origin of point mutations in human tumor cells. Cancer Res., 52: T. R. Human HPRT mutant database: software for data entry and retrieval. Environ.
249—253, 1992. Mol. Mutagen., 20: 81—83,1992.
11. Ehrlich, M., Zhang, X. Y., and Inamdar, N. M. Spontaneous deamination of cytosine 44. Kress, S., Sutter, C., Strickland, P. T., Mukhtar, H., Schweizer, J., and Schwarz, M.
and 5-methylcytosine residues in DNA and replacement of 5-methylcytosine residues Carcinogen-specific mutational pattern in the p53 gene in ultraviolet B radiation
with cytosine residues. Mutat. Res., 238: 277—286,1990. induced squamous cell carcinomas of mouse skin. Cancer Res., 52: 6400—6403,
12. Loeb, L. A., and Cheng, K. C. Errors in DNA synthesis: a source of spontaneous 1992.
mutations. Mutat. Res., 238: 297—304,1990. 45. Kanjilal, S., Pierceall, W. E., Cummings, K. K., Kripke, M. L., and Ananthaswamy,
13. Loeb, L. A., and Preston, B. D. Mutagenesis by apurinic/apyrimidinic sites. Annu. H. N. High frequency of p53 mutations in ultraviolet radiation-induced murine
Rev. Genet., 20: 201—230,
1986. skin tumors: evidence for strand bias and tumor heterogeneity. Cancer Res., 53:
14. Lutz, W. K. Endogenous genotoxic agents and processes as a basis of spontaneous 2961—2964,
1993.
carcinogenesis. Mutat. Res., 238: 287—295,1990. 46. Ruggeri, B., DiRado, M., Zhang, S. Y., Bauer, B., Goodrow, T., and Klein-Szanto,
15. Breimer, L. H. Molecular mechanisms of oxygen radical carcinogenesis and mu A. J. P. Benzo(a)pyrene-induced murine skin tumors exhibit frequent and character
tagenesis: the role of DNA base damage. Mol. Carcinog., 3: 188—197,1990. istic G to T mutations in the p53 gene. Proc. Natl. Acad. Sci. USA, 90: 1013—1017,
16. Rideout, W. M., III, Coetzee, G. A., Olumi, A. F., and Jones, P. A. 5-Methylcytosine 1993.
as an endogenous mutagen in the human LDL receptor and p53 genes. Science 47. Ruggeri, B., Caamano, J., Goodrow, T., DiRado, M., Bianchi, A., Trono, D., Conti,
(Washington DC), 249: 1288—1290, 1990. C. J., and Klein-Szanto, A. J. Alterations of the p53 tumor suppressor gene during
17. Kunkel, T. A. Misalignment-mediated DNA synthesis errors. Biochemistry, 29: mouse skin tumor progression. Cancer Res., 51: 6615—6621, 1991.
8003-8011, 1990. 48. Burns, P. A., Kemp, C. J., Gannon, J. V., Lane, D. P., Bremner, R., and Balmain, A.
18. Phear, G., Nalbantoglu, J., and Meuth, M. Next-nucleotide effects in mutations driven Loss of heterozygosity and mutational alterations of the p53 gene in skin tumours of
by DNA precursor pool imbalances at the apri locus of Chinese hamster ovary cells. interspecific hybrid mice. Oncogene, 6: 2363—2369,
1991.
Proc. Nati. Acad. Sci. USA, 84: 4450—4454,1987. 49. Shields, P. G., and Harris, C. C. Molecular epidemiology and the genetics of
19. Liu, P. K., Chang, C. C., Trosko, J. E., Dube, D. K., Martin, G. M., and Loeb, L A. environmental cancer. JAMA, 266: 681—687, 1991.
Mammalian mutator mutant with an aphidicolin-resistant DNA polymerase a. Proc. 50. Hollstein, M., Rice, K., Greenblatt, M. S., Soussi, T., Fuchs, R., Sortie, T., Hovig, E.,
Natl. Acad. Sci. USA, 80: 797—801, 1983. Smith-Sorensen, B., Montesano, R., and Harris, C. C. Database of p53 gene somatic
20. Loeb, L. A., Springgate, C. F., and Battula, N. Errors in DNA replication as a basis mutations in human tumors and cell lines. Nucleic Acids Res., 22(Supp.): in press,
of malignant changes. Cancer Res., 34: 2311—2321,1974. 1994.
21. Nowell, P. C. The clonal evolution of tumor cell populations. Science (Washington 51. Soussi, T., Caron de Fromentel, C., and May, P. Structural aspects of the p53 protein
DC), 194: 23—28,1976. in relation to gene evolution. Oncogene, 5: 945—952,1990.
22. Foulds, L. The experimental study of tumor progression: a review. Cancer Res., 14: 52. Park, M., and Vande Woude, G. F. Principles of molecular cell biology of cancer:
317—339, 1954. oncogenes. In: V. T. DeVita, Jr., S. Heilman, and S. A. Rosenberg (ads.), Cancer.
23. Chorazy, M. Sequence rearrangements and genome instability. A possible step in Principles and Practice of Oncology, Ed. 3, pp. 45—66.New York: J. B. Lippincott
carcinogenesis. J. Cancer Res. Clin. Oncol., 109: 159—172,1985. Co., 1989.
24. German, J. Bloom's syndrome. X. The cancer proneness points to chromosome 53. Macsaws, C., Tamura, G., Suzuki, Y., Ishida, K., Saito, K., and Satodate, R. Sensitive
mutation as a crucial event in human neoplasia. in: J. German (ed), Chromosome detection of p53 gene mutations in esophageal endoscopic biopsy specimens by cell
Mutation and Neoplasia, pp. 347—357.New York: Alan R. lAss, 1983. sorting combined with polymerase chain reaction single-strand conformation poly
25. Fishel, R., Lescoe, M. K., Rao, M. R., Copeland, N. G., Jenkins, N. A., Gather, J., morphism analysis. Jpn. J. Cancer Res., 83: 1253—1256,1992.
Kane, M., and Kolodner, R. The human mutator gene homolog MSH2 and its 54. Tamura, G., Kihana, T., Nomura, K., Terada, M., Sugimura, T., and Hirohashi, S.
association with hereditary nonpolyposis colon cancer. Cell, 75: 1027—1038,
1993. Detection of frequent p53 gene mutations in primary gastric cancer by cell sorting and
26. Leach, F. S., Nicolaides, N. C., Papadopoulos, N., et a!. Mutations of a mutS homolog polymerase chain reaction single-strand conformation polymorphism analysis. Cancer
in hereditary nonpolyposis colorectal cancer. Cell, 75: 1215—1235,1993. Res., 51: 3056—3058, 1991.
27. Bronner, C. E., Baker, S. M., Morrison, P. T., et a!. Mutation in the DNA mismatch 55. Nigro, J. M., Baker, S. J., Preisinger, A. C., et a!. Mutations in the p53 gene occur in
repair gene homologue hMLH1 is associated with hereditary non-polyposis colon diverse human tumor types. Nature (Land.), 342: 705—708,1989.
cancer. Nature (Land.), 368: 258—261, 1994. 56. Hsu, 1. C., Metcalf, R. A., Sun, T., Welsh, J., Wang, N. J., and Harris, C. C. p53 gene
28. Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., et a!. Mutation of a mutL homolog mutational hotspot in human hepatocellular carcinomas from Qidong, China. Nature
in hereditary colon cancer. Science (Washington DC), 263: 1625—1629,1994. (Land.), 350: 427—428,1991.
29. Ionov, Y., Peinado, M. A., Malkhosyan, S., Shibata, D., and Perucho, M. Ubiquitous 57. Hinds, P. W., Finlay, C. A., Quartin, R. S., Baker, S. J., Fearon, E. R., Vogelstein, B.,
somatic mutations in simple repeated sequences reveal a new mechanism for colonic and Levine, A. J. Mutant p53 DNA clones from human colon carcinomas cooperate
4874

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

with ras in transforming primary rat cells: a comparison of the “hot spot: mutant 87. Fraumeni, J. F., Hoover, R. N., Devesa, S. S., and Kinlen, L. J. Epidemiology of
phenotypes―. Cell Growth & Differ., I: 571—580,1990. cancer. in: V. T. DeVita, S. Hetlman, and S. A. Rosenberg (eds.), Cancer: Principle
58. Hall, P. A., and Lane, D. P. p53 in tumour pathology: can we trust and Practice of Oncology, pp. 150—181.Philadelphia: J. B. Lippincott, 1993.
immunohistochemistry?—revisited. J. Pathol., 172: 1—4,1994. 88. Jego, N., Thomas, G., and Hamelin, R. Short direct repeats flanking deletions, and
59. Castresana, J. S., Rubio, M. P., Vazquez, J. J., Idoate, M., Sober, A. J., Seizinger, duplicating insertions in p53 gene in human cancers. Oncogene, 8: 209—213, 1993.
B. R., and Barnhill, R. L. Lack of allelic deletion and point mutation as mechanisms 89. Krawczak, M., and Cooper, D. N. Gene deletions causing human genetic disease:
of p53 activation in human malignant melanoma. tnt. J. Cancer, 55: 562—565,1993. mechanisms of mutagenesis and the role of the local DNA sequence environment.
60. Weiss, J., Schwechheimer, K., Cavenee, W. K., Herlyn, M., and Arden, K. C. Hum. Genet., 86: 425—441,1991.
Mutation and expression of the p53 gene in malignant melanoma cell lines. tnt. J. 90. Ripley, L. S. Frameshift mutation: determinants of specificity. Annu. Rev. Genet., 24:
Cancer, 54: 693—699,1993. 189—213,1990.
61. Heimdal, K., Lathe, R. A., Lystad, S., HoIm, R., Fossa, S. D., and Borresen, A. L No 91. Lambert, I. B., Napolitano, R. L., and Fuchs, R. P. Carcinogen-induced frameshift
germline TP53 mutations detected in familial and bilateral testicular cancer. Genes mutagenesis in repetitive sequences. Proc. Natl. Acad. Sci. USA, 89: 1310—1314,
Chromosom. Cancer (Phila.), 6: 92—97,1993. 1992.
62. Peng, H. Q., Hogg, D., Malkin, D., Bailey, D., Gallie, B. L., Bulbul, M., Jewett, M., 92. Aaltonen, L. A., Peltomaki, P., Leach, F. S., et a!. Clues to the pathogenesis of
Buchanan, J., and Goss, P. E. Mutations of the p53 gene do not occur in testis cancer. familial colorectal cancer. Science (Washington DC), 260: 812—815, 1993.
Cancer Res., 53: 3574—3578, 1993. 93. Thibodeau, S. N., Bren, G., and Schaid, D. Microsatellite instability in cancer of the
63. El-Deity, W. S., Tokino, T., Velculescu, V. E., Levy, D. B., Parsons, R., Trent, J. M., proximal colon. Science (Washington DC), 260: 816—819, 1993.
Lin, D., Mercer, W. E., Kinzler, K. W., and Vogelstein, B. WAF1, a potential 94. Dunn, J. M., Phillips, R. A., Becker, A. J., and Gallie, B. L. Identification of germline
mediator of p53 tumor suppression. Cell, 75: 817—825, 1993. and somatic mutations affecting the retinoblastoma gene. Science (Washington DC),
64. Harper, J. W., Adami, G. R., Wei, N., Keyomarsi, K., and Elledge, S. J. The p21 241: 1797—1800, 1988.
talk-interacting protein Cipi is a potent inhibitor of G, cyclin-dependent kinases. Cell, 95. Harbour, J. W., Lxi, S. L., Whang-Peng, J., Gazdar, A. F., Minna, J. D., and Kaye,
75: 805—816,1993. F. J. Abnormalities in structure and expression of the human retinoblastoma gene in
65. Orita, M., Iwahana, H., Kanazawa, H., Hayashi, K., and Sekiya, T. Detection of SCLC. Science (Washington DC), 241: 353—357,1988.
polymorphisms of human DNA by gel electrophoresis as single-strand conformation 96. Clore, G. M., Omichinski, J. G., Sakaguchi, K., Zambrano, N., Sakamoto, H.,
polymorphisms. Proc. Natl. Acad. Sci USA, 86: 2766—2770, 1989. Appella, E., and Gronenborn, A. M. High-resolution solution structure of the oh
66. Murakami, Y., Hayashi, K., and Sekiya, T. Detection of aberrations of the p53 alleles gomerization domain of p53 by multi-dimensional NMR. Science (Washington DC),
and the gene transcript in human tumor cell lines by single-strand conformation 265:386—391, 1994.
polymorphism analysis. Cancer Res., 51: 3356—3361, 1991. 97. Cho, Y., Gorina, S., Jeffrey, P., and Pavletich, N. P. Crystal structure of a p53 tumor
67. Mitsudomi, T., Steinberg, S. M., Nau, M. M., et a!. p53 gene mutations in non-small suppressor-DNA complex: a framework for understanding how mutations inactivate
cell lung cancer cell lines and their correlation with the presence of ras mutations and p53. Science (Washington DC), 265: 346—355,1994.
clinical features. Oncogene, 7: 171—180,1992. 98. Unger, T., Nau, M. M., Segal, S., and Minna, J. D. p53: a transdominant regulator of
68. Borresen, A. L., Hovig, E., Smith-Sorensen, B., Malkin, D., Lystad, S., Andersen, transcription whose function is ablated by mutations occurring in human cancer.
T. I., Nesland, J. M., Isselbacher, K. J., and Friend, S. H. Constant denaturant gel EMBO J., 11: 1383—1390, 1992.
electrophoresis as a rapid screening technique for p53 mutations. Proc. Nail. Acad. 99. Momand, J., Zambetti, G. P., Olson, D. C., George, D., and Levine, A. J. The mdm-2
Sci. USA, 88: 8405—8409, 1991. oncogene product forms a complex with the p53 protein and inhibits p53-mediated
69. Smith-Sorensen, B., Gebhardt, M. C., Kloen, P., McIntyre, J., Aguilar, F., Cerutti, P., transactivation. Cell, 69: 1237—1245,1992.
and Borresen, A. L. Screening for TPS3 mutations in osteosarcomas using constant 100. Ohiner, J. D., Pietenpol, J. A., Thiagahingam, S., Gyuris, J., Kinzler, K. W., and
denaturant gel electrophoresis (CDGE). Hum. Mutat., 2: 274—285,1993. Vogelstein, B. Oncoprotein MDM2 conceals the activation domain of tumour
70. Condie, A., Eeles, R., Borresen, A. L., Coles, C., Cooper, C., and Prosser, J. Detection suppressor p53. Nature (Land.), 362: 857—862, 1993.
of point mutations in the p53 gene: comparison of single-strand conformation poly 101. Chen, J., Marechal, V., and Levine, A. J. Mapping of the p53 and mdm-2 interaction
morphism, constant denaturant gel electrophoresis, and hydroxylamine and osmium domains. Mol. Cell. Biol., 13: 4107—41 14, 1993.
tetroxide techniques. Hum. Mutat., 2: 58—66,1993. 102. Brown, D. R., Deb, S., Munoz, R. M., Subler, M. A., and Deb, S. P. The tumor
71. Matlashewski, G. J., Tuck, S., Pim, D., Lamb, P., Schneider, J., and Crawford, L. V. suppressor p53 and the oncoprotein simian virus 40 T-antigen bind to overlapping
Primary structure polymorphism at amino acid residue 72 of human p53. Mol. Cell. domains on the MDM2 protein. Mol. Cell. Biol., 13: 6849—6857, 1993.
Biol., 7: 961—963,1987. 103. Srinivasan, R., Roth, J. A., and Maxwell, S. A. Sequence-specific interaction of a
72. Carbone, D., Chiba, I., and Mitsudomi, T. Polymorphism at codon 213 within the p53 conformational domain of p53 with DNA. Cancer Res., 53: 5361—5364,1993.
gene. Oncogene, 6: 1691—1692,1991. 104. Ruppert, J. M., and Stillman, B. Analysis of a protein-binding domain of p53. Mol.
73. Eckert, K. A., and Kunkel, T. A. The fidelity of DNA polymerases used in the Cell. Biol., 13: 3811—3820,1993.
polymerase chain reactions. in: M. J. McPherson, P. Quirke, and G. R. Taylor (eds.), 105. Sturzbecher, H. W., Brain, R., Addison, C., Rudge, K., Remm, M., Grimahdi, M.,
PCR: A Practical Approach, pp. 225—245.
New York: Oxford University Press, 1991. Keenan, E., and Jenkins, J. R. A C-terminal a-helix plus basic region motif is the
74. Jackson, D. P., Hayden, J. D., and Quirke, P. Extraction of nucleic acid from fresh and major structural determinant ofp53 tetramerization. Oncogene, 7: 1513—1523,1992.
archival material. in: M. J. McPherson, P. Quirke, and G. R. Taylor (eds.), PCR: A 106. Milner, J., and Medcalf, E. A. Cotranslation of activated mutant p53 with wild type
Practical Approach, pp. 29—50.New York: Oxford University Press, 1991. drives the wild-type p53 protein into the mutant conformation. Cell, 65: 765—774,
75. Bohr, V. A., Phillips, D. H., and Hanawalt, P. C. Heterogeneous DNA damage and 1991.
repair in the mammalian genome. Cancer Res., 47: 6426—6436, 1987. 107. Tarunina, M., and Jenkins, J. R. Human p53 binds DNA as a protein homodimer but
76. Xanthoudakis, S., Miao, G., Wang, F., Pan, Y. C., and Curran, T. Redox activation of monomeric variants retain full transcription transactivation activity. Oncogene, 8:
Fos-Jun DNA binding activity is mediated by a DNA repair enzyme. EMBO J., Ii: 3165—3173, 1993.
3323—3335, 1992. 108. Pietenpol, J. A., Tokino, T., Thiagalingam, S., El-Deity, W. S., Kinzler, K. W., and
77. Schaeffer, L., Roy, R., Humbert, S., Moncollin, V., Vermeulen, W., Hoeijmakers, Vogelstein, B. Sequence-specific transcriptional activation is essential for growth
J. H., Chambon, P., and Egly, J. M. DNA repair helicase: a component of BTF2 suppression by p53. Proc. Nath. Acad. Sci. USA, 91: 1998—2002, 1994.
(TFIIH) basic transcription factor. Science (Washington DC), 260: 58—63, 1993. 109. Hainaut, P., and Milner, J. Redox modulation of p53 conformation and sequence
78. Evans, M. K., Taffe, B. G., Harris, C. C., and Bohr, V. A. DNA strand bias in the specific DNA binding in vitro. Cancer Res., 53: 4469—4473, 1993.
repair of the p53 gene in normal human and xeroderma pigmentosum group C 110. Martinez, J., Georgoff, I., and Levine, A. J. Cellular localization and cell cycle
fibroblasts. Cancer Res., 53: 5377—5381, 1993. regulation by a temperature-sensitive p53 protein. Genes 0ev., 5: 151—159,1991.
79. Bohr, V. A. Gene specific DNA repair. Carcinogenesis (Land.), 12: 1983—1992, 111. Michalovitz, D., Halevy, 0., and Oren, M. Conditional inhibition of transformation
1991. and of cell proliferation by a temperature-sensitive mutant of p53. Cell, 62: 671—
80. Sato, M., Nishigori, C., Zghal, M., Yagi, T., and Takebe, H. Ultraviolet-specific 680, 1990.
mutations in p53 gene in skin tumors in xeroderma pigmentosum patients. Cancer 112. Reed, M., Wang, Y., Mayr, G., Anderson, M. E., Schwedes, J. F., and Tegtmeyer,
Res., 53: 2944—2946, 1993. P. p53 domains: suppression, transformation, and transactivation. Gene Exp., 3:
81. Dumaz, N., Drougard, C., Sarasin, A., and Daya-Grosjean, L. Specific UV-induced 95—107,1993.
mutation spectrum in the p53 gene of skin tumors from DNA-repair-deficient xero 113. Davidoff, A. M., Iglehart, J. D., and Marks, J. R. Immune response to p53 is
derma pigmentosum patients. Proc. Natl. Acad. Sci. USA, 90: 10529—10533,1993. dependent upon p53IHSP7O complexes in breast cancers. Proc. NatI. Acad. Sci.
82. Gupta, P. K., Johnson, D. L., Reid, T. M., Lee, M. S., Romano, L. J., and King, C. M. USA, 89: 3439—3442, 1992.
Mutagenesis by single site-specific arylamine-DNA adducts. Induction of mutations 114. Schhichtholz, B., Legros, Y., Gillet, D., Gaillard, C., Marty, M., Lane, D., Calvo, F.,
at multiple sites. J. Biol. Chem., 264: 20120—20130, 1989. and Soussi, T. The immune response to p53 in breast cancer patients is directed
83. Klein, J. C., Bleeker, M. J., Lutgerink, J. T., et a!. Use of shuttle vectors to study the against immunodominant epitopes unrelated to the mutational hot spot. Cancer Res.,
molecular processing of defined carcinogen-induced DNA damage: mutagenicity of 52: 6380—6384, 1992.
single O―-ethylthymine adducts in HeLa cells. Nucleic Acids Res., 18: 4131—4137, I 15. Crawford, L. V., Pim, D. C., and Bulbrook, R. D. Detection of antibodies against the
1990. cellular protein p53 in sera from patients with breast cancer. Int. J. Cancer, 30:
84. Moriya, M. Single-stranded shuttle phagemid for mutagenesis studies in mammalian 403—408, 1982.
cells: 8-oxoguanine in DNA induces targeted G. C—p T.A transversions in simian 116. Caron de Fromentel, C., May-Levin, F., Mouriesse, H., Lemerle, J., Chandrasekaran,
kidney cells. Proc. NatI. Acad. Sci USA, 90: 1122—1 126, 1993. K., and May, P. Presence of circulating antibodies against cellular protein p53 in a
85. Harris, C. C. Chemical and physical carcinogenesis: advances and perspectives. notable proportion of children with B-cell lymphoma. Int. J. Cancer, 39: 185—189,
Cancer Res., 51: 5023s—5044s,1991. 1987.
86. Hussain, S. P., Aguilar, F., and Cerutti, P. Mutagenesis of codon 248 of the human 117. Lubin, R., Schlichtholz, B., Bengoufa, D., et a!. Analysis of p53 antibodies in
p53 tumor suppressor gene by N-ethyl-N-nitrosourea. Oncogene, 9: 13—18,1994. patients with various cancers define B-cell epitopes of human p53: distribution on
4875

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

primary structure and exposure on protein surface. Cancer Res., 53: 5872—5876, 146. Dolcetti, R., Doglioni, C., Maestro, R., Gasparouo, D., Barzan, L, Pastore, A.,
I 993. Romanelli, M., and Boiocchi, M. p53 over-expression is an early event in the
118. Vohkmann, M., Muller, M., Hofmann, W. J., Meyer, M., Hagelstein, J., Rath, U., development of human squamous-cell carcinoma of the larynx: genetic and prog
Kommerell, B., Zentgraf, H., and Galle, P. R. The humorahimmune response to p53 nostic implications. Int. J. Cancer, 52: 178—182,1992.
in patients with hepatocellular carcinoma is specific for malignancy and independent 147. Coltrera, M. D., Zarbo, R. J., Sakr, W. A., and Gown, A. M. Markers for dysphasia
of the a-fetoprotein status. Hepatology, 18: 559—565, 1993. of the upper aerodigestive tract. Suprabasal expression of PCNA, p53, and CK19 in
119. Schuler, G. D., Altschul, S. F., and Lipman, D. J. A workbench for multiple alcohol-fixed, embedded tissue. Am. J Pathol., 141: 817—825, 1992.
alignment construction and analysis. Proteins, 9: 180—190,1991. 148. Ogden, G. R., Lane, D. P., and Chishohm, D. M. p53 expression in dyskeratosis
120. Milne, D. M., Palmer, R. H., and Meek, D. W. Mutation of the casein kinase II congenita: a marker for oral premahignancy? J. Chin. Pathol., 46: 169—170,1993.
phosphorylation site abolishes the anti-proliferative activity of p53. Nucleic Acids 149. Boyle, J. 0., Hakim, J., Koch, W., van der Riet, P., Hruban, R. H., Roa, R. A.,
Res., 20: 5565—5570, 1992. Correo, R., Eby, Y. J., Ruppert, J. M., and Sidransky, D. The incidence of p53
121. Fiscella, M., Ullrich, S. J., Zambrano, N., Shields, M. T., Lin, D., Lees-Miller, S. P., mutations increases with progression of head and neck cancer. Cancer Res., 53:
Anderson, C. W., Mercer, W. E., and Appella, E. Mutation of the serine 15 4477—4480, 1993.
phosphoryhation site of human p53 reduces the ability of p53 to inhibit cell cycle 150. Bennett, W. P., Hohlstein, M. C., Metcalf, R. A., Welsh, J. A., He, A., Thu. S.,
progression. Oncogene, 8: 1519—1528, 1993. Kusters, I., Resau, J. H., Trump, B. F., Lane, D. P., and Harris, C. C. p53 mutation
122. Fiscella, M., Zambrano, N., Uhlrich, S. J., Unger, T., Lin, D., Coo, B., Mercer, W. E., and protein accumulation during multistage human esophageal carcinogenesis. Can
Anderson, C. W., and Appella, E. The carboxy-terminal serine 392 phosphorylation cer Res., 52: 6092—6097, 1992.
site of human p53 is not required for wild-type activities. Oncogene, in press, 1994. 151. Sasano, H., Goukon, Y., Nishihira, T., and Nagura, H. In situ hybridization and
123. Lehman, T. A., Bennett, W. P., Metcalf, R. A., Reddel, R., Welsh, J. A., Ecker, J., immunohistochemistry of p53 tumor suppressor gene in human esophageal card
Modahi,R. V., Ullrich, S., Romano, J. W., Appella, E., Testa, J. R., Gerwin, B. I., noma. Am. J. Pathol., 141: 545—550,1992.
and Harris, C. C. p53 mutations, ras mutations and p53-heat shock 70 protein 152. Wang, L. D., Hong, J. Y., Qiu, S. L., Gao, H., and Yang, C. S. Accumulation of p53
complexes in human hung cell lines. Cancer Res., Si: 4090—4096, 1991. protein in human esophageal precancerous lesions: a possible early biomarker for
124. Enomoto, T., Fujita, M., Inoue, M., Rice, J. M., Nakajima, R., Tanizawa, 0., and carcinogenesis. Cancer Res., 53: 1783—1787,1993.
Nomura, T. Alterations of the p53 tumor suppressor gene and its association with 153. Fearon, E. R., and Vogehstein, B. A genetic model for cohorectal tumorigenesis. Cell,
activation of the c-K-ras-2 protooncogene in premahignant and malignant lesions of 61: 759—767,1990.
the human uterine endometrium. Cancer Res., 53: 1883—1888,1993. 154. Kikuchi-Yanoshita, R., Konishi, M., Ito, S., a' a!. Genetic changes of both p53
125. Shaw, P., Tardy, S., Benito, E., Obrador, A., and Costa, J. Occurrence of Ki-ras and alleles associated with the conversion from cohorectal adenoma to early carcinoma
p53 mutations in primary colorectal tumors. Oncogene, 6: 2121—2128,1991. in familial adenomatous polyposis and non-familial adenomatous pohyposis patients.
126. Kalthoff, H., Schmiegeh, W., Roeder, C., et a!. p53 and K-RAS alterations in Cancer Res., 52: 3965—3971,
1992.
pancreatic epithehial cell lesions. Oncogene, 8: 289—298, 1993. 155. Burmer, G. C., Crispin, D. A., Kolli, V. R., Haggitt, R. C., Kulander, B. G., Rubin,
127. Nikolaidou, A., Liloglou, T., Malliri, A., Ergazaki, M., Tiniakos, G., Tiniakos, D., C. E., and Rabinovitch, P. S. Frequent loss of a p53 allele in carcinomas and their
and Spandidos, D. A. Detection of hepatitis B virus DNA and mutations in K-ras precursors in ulcerative colitis. Cancer Commun., 3: 167—172,1991.
and p53 genes in human hepatocellular carcinomas. Int. J. Oncol., 3: 593—601,1993. 156. Burmer, G. C., Rabinovitch, P. S., Haggitt, R. C., Crispin, D. A., Brentnahh, T. A.,
128. Rodenhuis, S., and Shebos, R. J. Clinical significance of ras oncogene activation in Kohhi, V. R., Stevens, A. C., and Rubin, C. E. Neophastic progression in ulcerative
human lung cancer. Cancer Res., 52: 2665s—2669s, 1992. colitis: histology, DNA content, and loss of a p53 allele. Gastroenterology, 103:
129. Dowell, S. P., and Halt, P. A. The clinical relevance of the p53 tumour suppressor 1602—1610,1992.
gene. Cytopathol., 5: 133—145,1994. 157. Pignatelhi, M., Stamp, G. W., Kafiri, G., Lane, D., and Bodmer, W. F. Over
130. Slaughter, T. P., Southwick, H. W., and Smejkel, W. Field cancerization in oral expression of p53 nuclear oncoprotein in colorectal adenomas. Int. J. Cancer, 50:
stratified epithehium. Cancer (Phila.), 6: 963-968, 1953. 683—688, 1992.
131. Yin, J., Harpaz, N., Tong, Y., Huang, Y., Laurin, J., Greenwald, B. D., Hontanosas, 158. van den Berg, F. M., Tigges, A. J., Schipper, M. E., den Hartog-Jager, F. C., Kroes,
M., Newkirk, C., and Mehtzer,S. J. p53 point mutations in dysplastic and cancerous w. G., and Walboomers,J. M. Expressionof the nuclearoncogenep53 in colon
ulcerative colitis lesions. Gastroenterohogy, 104: 1633—1639,1993. tumours. J. Pathol., 157: 193—199,1989.
132. Nees, M., Homann, N., Discher, H., Andl, T., Enders, C., Herold-Mende, C., 159. Tsuda, H., and Hirohashi, S. Frequent occurrence of p53 gene mutations in uterine
Schuhmann, A., and Bosch, F. X. Expression of mutated p53 occurs in tumor-distant cancers at advanced clinical stage and with aggressive histological phenotypes. Jpn.
epithehia of head and neck cancer patients: a possible molecular basis for the J. Cancer Res., 83: 1184—1191,1992.
development of multiple tumors. Cancer Res., 53: 4189—4196, 1993. 160. Kohler, M. F., Marks, J. R., Wiseman, R. W., Jacobs, I. J., Davidoff, A. M.,
133. Chung, K. Y., Mukhopadhyay, T., Kim, J., Casson, A., Ro, J. Y., Goepfert, H., Clarke-Pearson, D. L, Soper, J. T., Bast, R. C., Jr., and Berchuck, A. Spectrum of
Hong, W. K., and Roth, J. A. Discordant p53 gene mutations in primary head and mutation and frequency of allehic deletion ofthe p53 gene in ovarian cancer. J. Natl.
neck cancers and corresponding second primary cancers of the upper aerodigestive Cancer Inst., 85: 1513—1519,
1993.
tract. Cancer Rca, 53: 1676—1683,
1993. 161. Navone, N. M., Troncoso, P., Pisters, L L., Goodrow, T. L., Palmer, J. L., Nichols,
134. Thor, A. D., Moore, D. H., II, Edgerton, S. M., et a!. Accumulation of p53 tumor W. W., Von Eschenbach, A. C., and Conti, C. J. p53 protein accumulation and gene
suppressor gene protein: an independent marker of prognosis in breast cancers. J. mutation in the progression of human prostate carcinoma. J. Nath. Cancer Inst., 85:
NatI. Cancer Inst., 84: 845—855, 1992. 1657—1669, 1993.
135. Tsuda, H., Iwaya, K., Fukutomi, T., and Hirohashi, S. p53 mutations and c-erbB-2 162. Fujimoto, K., Yamada, Y., Okijima, E., Kakizoe, T., Sasaki, H., Sugimura, T., and
amplification in intraductal and invasive breast carcinomas of high histologic grade. Terada, M. Frequent association of p53 gene mutation in invasive bladder cancer.
Jpn. J Cancer Res., 84: 394—401, 1993. Cancer Res., 52: 1393—1398,1992.
136. Murakami, Y., Hayashi, K., Hirohashi, S., and Sekiya, T. Aberrations of the tumor 163. Habuchi, T., Takahashi, R., Yamada, H., et a!. Influence of cigarette smoking
suppressor p53 and retinoblastoma genes in human hepatocellular carcinomas. and schistosomiasis on p53 gene mutation in urothehial cancer. Cancer Res., 53:
Cancer Rca., 51: 5520—5525, 1991. 3795—3799,1993.
137. Nishida, N., Fukuda, Y., Kokuryu, H., Toguchida, J., Yandell, D. W., Ikenega, M., 164. Chiba, I., Takahashi, T., Nau, M. M., D'Amico, D., Curiel, D. T., Mitsudomi, T.,
Imura, H., and Ishizaki, K. Role and mutational heterogeneity of the p53 gene in Buchhagen, D. L., Carbone, D., Piantadosi, S., Koga, H., Reissman, P. T.,
hepatocellular carcinoma. Cancer Res., 53: 368—372, 1993. Slamon, D. J., Holmes, E. C., and Minna, J. D. Mutations in the p53 gene are
138. Kohler, M. F., Kerns, B. J., Humphrey, P. A., Marks, J. R., Brat, R. C., Jr., and frequent in primary, resected non-small cell hung cancer. Oncogene, 5:
Berchuck, A. Mutation and overexpression of p53 in early-stage epithehial ovarian 1603—1610, 1990.
cancer. Obstet. Gynecol., 81: 643—650,1993. 165. Lahmann, D., Puts, B., Reich, U., Bohm, J., PraOer, H., and Höfler,H. Mutational
139. Teneriello, M. G., Ebina, M., Linnoila, R. I., Henry, M., Nash, J. D., Park, R. C., and spectrum of the p53 gene in human small-cell hung cancer and relationship to
Birrer, M. J. p53 and Ki-ras gene mutations in epithehial ovarian neoplasms. Cancer chinicopathohogicaldata. Am. J Pathol., 142: 907—915,
1993.
Res., 53: 3103-3108, 1993. 166. Lee, N. K., Ye, Y. W., Chen, J., Li, X., Waber, P. G., and Nisen, P. D. p53,
140. Vahakangas, K. H., Samet, J. M., Metcalf, R. A., Welsh, J. A., Bennett, W. P., Lane, retinoblastoma, and human papihlomavirus in squamous cell carcinoma and adjacent
D. P., and Harris, C. C. Mutations of p53 and ras genes in radon-associated lung normal mucosa of the upper aerodigestive tract. Arch. Otoharyngol. Head Neck
cancer from uranium miners. Lancet, 339: 576—580, 1992. Surg., 119: 1125—1131,1993.
141. Sundaresan, V., Ganly, P., Hasleton, P., Rudd, R., Sinha, G., Bleehen, N. M., and 167. Sidransky, D., Mikkelsen, T., Schwechheimer, K., Rosenblum, M. L., Cavanee, W.,
Rabbitts, P. p53 and chromosome 3 abnormalities, characteristics of malignant and Vogehstein, B. Clonal expansion of p53 mutant cells is associated with brain
lung tumours, are detectable in preinvasive lesions of the bronchus. Oncogene, 7: tumour progression. Nature (Land.), 355: 846—847, 1992.
1989—1997, 1992. 168. Hayashi, Y., Yamashita, J., and Yamaguchi, K. Timing and role of p53 gene
142. Sozzi, G., Miozzo, M., Donghi, R., Pilotti, S., Cariani, C. T., Pastorino, U., Della mutation in the recurrence of ghioma. Biochem. Biophys. Res. Commun., 180:
Porta, G., and Pierotti, M. A. Deletions of l7p and p53 mutations in preneoplastic 1145—1150,1991.
lesions of the hung. Cancer Rca., 52: 6079—6082, 1992. 169. Gasparini, G., Weidner, N., Mahuta, S., Pozza, F., Boracchi, P., Mezzetti, M.,
143. Nuorva, K., Soini, Y., Kamel, D., Autio-Harmainen, H., Ristehi, L., Ristehi, J., Testohin, A., and Bevihacqua, P. Intratumoral microvessel density and p53 protein:
Vahakangas, K., and Paakko, P. Concurrent p53 expression in bronchial dysphasias correlation with metastasis in head-and-neck squamous-cell carcinoma. Int. J.
and squamous cell lung carcinomas. Am. J. Pathol., 142: 725—732,1993. Cancer, 55: 739—744,1993.
144. Bennett, W. P., Colby, T. V., Vahakangas, K. H., et a!. p53 protein accumulates 170. Chin, K. V., Ueda, K., Pastan, 1., and Gottesman, M. M. Modulation of activity of
frequently in early bronchial neophasia. Cancer Res., 53: 4817—4822, 1993. the promoter of the human MDRJ gene by Ras and p53. Science (Washington DC),
145. Gusterson, B. A., Anbazhagan, R., Warren, W., Midgehy, C., Lane, D. P., O'Hare, 255: 459—462,1992.
M., Stamps, A., Carter, R., and Jayatilake, H. Expression of p53 in premalignant and 171. Dameron, K. M., Volpert, 0. V., and Bouck, N. Regulation of an angiogenesis
malignant squamous epithehium.Oncogene, 6: 1785—1789, 1991. inhibitor in human fibrobhasts by the p53 tumor suppressor gene. J. Cell Biochem.,
4876

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

18C: 187, 1994. p53 are associated in vitro and in liver tissues from patients with primary hepato
172. Kihana, T., Tsuda, H., Teshima, S., Okada, S., Matsuura, S., and Hirohashi, S. High cellular carcinoma. Oncogene, 8: 1109—1 117, 1993.
incidence of pS3 gene mutation in human ovarian cancer and its association with 202. Wang, X. W., Forrester, K., Yeh, H., Feitelson, M. A., Gu, J. R., and Harris, C. C.
nuclear accumulation of p53 protein and tumor DNA aneuploidy. Jpn. J. Cancer Hepatitis B virus X protein inhibits p53 sequence-specific DNA binding, transcrip
Res., 83: 978—984,1992. tional activity, and association with transcription factor ERCC3. Proc. Nail. Acad.
173. Remvikos, Y., Tominaga, 0., Hammel, P., Laurent-Puig, P., Salmon, R. J., Sci. USA, 91: 2230—2234,1994.
Dutrihhaux,B., and Thomas, G. Increased p53 protein content of colorectal tumours 203. Wogan, G. N. Aflatoxins as risk factors for hepatocellular carcinoma in humans.
correlates with poor survival. Br. J. Cancer, 66: 758—764, 1992. Cancer Res., 52: 21 14s—21 18s, 1992.
174. Jung, M., Notario, V., and Dritschiho, A. Mutations in the p.5.3 gene in radiation 204. Buhatao-Jayme, J., Almero, E. M., Castro, M. C., Jardeheza, M. T., and Sahamat,
sensitive and -resistant human squamous carcinoma cells. Cancer Res., 52: L. A. A case-control dietary study of primary liver cancer risk from aflatoxin
6390—6393, 1992. exposure. Int. J. Epidemiol., 11: 112—119,1982.
175. Brachman, D. G., Beckett, M., Graves, D., Haraf, D., Vokes, E., and Weichsehbaum, 205. Peers, F., Bosch, X., Kaldor, J., Linsell, A., and Phuijmen, M. Aflatoxin exposure,
R. R. p53 mutation does not correlate with radiosensitivity in 24 head and neck hepatitis B virus infection andhiver cancer in Swaziland. Int. J. Cancer, 39:545-553,
cancer cell lines. Cancer Res., 53: 3667—3669,1993. 1987.
176. O'Connor, P. M., Jackman, J., Jondle, D., Bhatia, K., Magrath, I., and Kohn, K. W. 206. Van Rensburg, S. J., Cook-Mozaffari, P., Van Schalkwyk, D. J., Van der Watt, J. J.,
Role of the p53 tumor suppressor gene in cell cycle arrest and radiosensitivity of Vincent, T. J., and Purchase, I. F. Hepatocelluhar carcinoma and dietary aflatoxin in
Burkitt's lymphoma cell lines. Cancer Rca., 53: 4776—4780, 1993. Mozambique and Transkei. Br. J. Cancer, 51: 713—726,1985.
177. Harris, C. C. Human tissues and cells in carcinogenesis research. Cancer Rca., 47: 207. Yeh, F. S., Yu, M. C., Mo, C. C., Lao, S., Tong, M. J., and Henderson, B. E.
1—10,1987. Hepatitis B virus, aflatoxins, and hepatocehiular carcinoma in southern Guangxi,
178. Chen, T. M., Chen, C. A., Hsieh, C. Y., Chang, D. Y., Chen, Y. H., and Defendi, China.CancerRes.,49: 2506-2509,1989.
V. The state of p53 in primary human cervical carcinomas and its effects in 208. Bressac, B., Kew, M., Wands, J., and Ozturk, M. Selective G to T mutations of
human papillomavirus-immortahized human cervical cells. Oncogene, 8: 1511— p53 gene in hepatocelluhar carcinoma from southern Africa. Nature (Land.), 350:
1518,1993. 429—431, 1991.
179. Crook, T., Wrede, D., and Vousden, K. H. p53 point mutation in HPV negative 209. Chahlen, C., Lunec, J., Warren, W., Collier, J., and Bassendine, M. F. Analysis of the
human cervical carcinoma cell lines. Oncogene, 6: 873—875,1991. p53 tumor-suppressor gene in hepatocelluhar carcinomas from Britain. Hepatohogy,
180. Scheffner, M., Munger, K., Byrne, J. C., and Howley, P. M. The state ofthe p53 and 16: 1362—1366,1992.
retinoblastoma genes in human cervical carcinoma cell lines. Proc. Nath. Acad. Sci. 210. Hohhstein, M. C., Wild, C. P., Bleicher, F., Chutimataewin, S., Harris, C. C.,
USA, 88: 5523—5527,1991. Srivatanakuh, P., and Montesano, R. p53 mutations and aflatoxin B, exposure in
181. Fagin, J. A., Matsuo, K., Karmakar, A., Chen, D. L., Tang, S. H., and Koeffler, H. P. hepatocehlular carcinoma patients from Thailand. Int. J. Cancer, 53: 51—55,1993.
High prevalence of mutations of the p53 gene in poorly differentiated human thyroid 21 1. Li, D., Ceo, Y., He, L., Wang, N. J., and Gu, J. Aberrations of p53 gene in human
carcinomas. J. Chin. Invest., 91: 179—184,1993. hepatocellular carcinoma from China. Carcinogenesis (Land.), 14: 169—173,1993.
182. Wright, P. A., Lemoine, N. R., Goretzki, P. E., Wylhie, F. S., Bond, J., Hughes, C., 212. Oda, T., Tsuda, H., Scarpa, A., Sakamoto, M., and Hirohashi, S. p53 gene mutation
Roher, H. D., Williams, E. D., and Wynford-Thomas, D. Mutation of the p53 gene spectrum in hepatocelhular carcinoma. Cancer Res., 52: 6358—6364, 1992.
in a differentiated human thyroid carcinoma cell line, but not in primary thyroid 213. Debuire, B., Paterhini, P., Pontisso, P., Basso, G., and May, E. Analysis of the p.53
tumours. Oncogene, 6: 1693—1697,1991. gene in European hepatocellular carcinomas and hepatoblastomas. Oncogene, 8:
183. Effert, P., McCoy, R., Abdeh-Hamid, M., Flynn, K., Zhang, Q., Busson, P., Tursz, 2303—2306, 1993.
T., Liu, E., and Raab-Traub, N. Alterations of the p53 gene in nasopharyngeal 214. Buetow, K. H., Sheffield, V. C., Zhu, M., Thou, T., Shen, F., Hino, 0., Smith, M.,
carcinoma. J. Virol., 66: 3768—3775, 1992. McMahon, B. J., Lanier, A. P., Landon, W. T., Redeker, A. G., and Govindarajan,
184. Spruck, C. H., Thai, Y. C., Huang@D. P., Yan& A. S., RideOut,W. M., Gonzalez S. Law frequencyof p53 mutationsobservedin a diversecollectionof primary
Zulueta, M., Choi, P., La, K. W., Yu, M. C., and Jones, P. A. Absence of p53 gene hepatocehiuharcarcinomas. Proc. Nail. Acad. Sci. USA, 89: 9622—9626,1992.
mutations in primary nasopharyngeal carcinomas. Cancer Res@52: 4787-4790, 1992. 215. Teramoto, T., Satonaka, K., Kitazawa, S., Fujimori, T., Hayashi, K., and Maeda, S.
185. Somers, K. D., Merrick, M. A., Lopez, M. E., Incognito, L. S., Schechter, G. L., and p53 gene abnormalities are closely related to hepatoviral infections and occur at a
Casey, G. Frequent p53 mutations in head and neck cancer. Cancer Res., 52: late stage of hepatocarcinogenesis. Cancer Res., 54: 231—235,1994.
5997—6000,1992. 216. Fujimoto, Y., Hampton, L. L., Wirth, P. J., Wang, N. J., Xie, J. P., and Thorgeirsson,
186. Stretch, J. R., Gatter, K. C., Rahfkiaer,E., Lane, D. P., and Harris, A. L. Expression S. S. Alterations of tumor suppressor genes and allehiclosses in human hepatocel
of mutant p53 in melanoma. Cancer Rca., 51: 5976—5979,1991. hular carcinomas in China. Cancer Res., 54: 281-285, 1994.
187. Council on Scientific Affairs, Harmful effects of ultraviolet radiation. JAMA, 262: 217. Ross, R. K., Yuan, J. M., Yu, M. C., Wogan, G. N., Qian, G. S., Tu, J. T., Groopman,
380—384, 1989. J. D., Gao, Y. T., and Henderson, B. E. Urinary aflatoxin biomarkers and risk of
188. Reid, T. M., and Laeb, L. A. Mutagenic specificity of oxygen radicals produced by hepatocellular carcinoma. Lancet, 339: 943—946,1992.
human leukemia cells. Cancer Rca., 52: 1082—1086,1992. 218. Qian, G. S., Ross, R. K., Yu, M. C., Yuan, J. M., Gao, Y. T., Henderson, B. E.,
189. Reid, T. M., and Loeb, L. A. Tandem double CC—.iT mutations are produced by Wogan, G. N., and Groopman, J. D. A follow-up study of urinary markers of
reactive oxygen species. Proc. NatI. Acad. Sci. USA, 90: 3904—3907, 1993. aflatoxin exposure and liver cancer risk in Shanghai, People's Republic of China.
190. Brash, D. E., Rudolph, J. A., Simon, J. A., Lin, A., McKenna, G. J., Baden, H. P., Cancer Epidemiol. Biomarkers. Prey., 3: 3—10,1994.
Halperin, A. J., and Ponten, J. A role for sunlight in skin cancer: UV-induced 219. Pfeifer, A. M. A., Cole, K. E., Smoot, D. T., Weston, A., Groopman, J. D., Shields,
p53 mutations in squamous cell carcinoma. Proc. NatI. Acad. Sci. USA, 88: P. G., Vignaud, J-M., Juillerat, M., Lipsky, M. M., Trump, B. F., Lechner, J. F., and
10124—10128, 1991. Harris, C. C. SV4O T-antigen immortalized normal human liver epithehial cells
191. Nakazawa, H., English, D., Randell, P. L, Nakazawa, K., Martel, N., Armstrong, express hepatocyte characteristics and metabolize chemical carcinogens. Proc. Nath.
B. K., and Yamasaki, H. UV and skin cancer: specific p53 gene mutation in normal Acad. Sci. USA, 90: 5123—5127,
1993.
skin as a biologically relevant exposure measurement. Proc. Nath. Acad. Sci. USA, 220. Autrup, H., Harris, C. C., Wu, S. M., Bao, L Y., Pci, X. F., Lu, S., Sun, T. T., and
91: 360—364,1994. Hsia, C. C. Activation of chemical carcinogens by cultured human fetal liver,
192. Tornaletti, S., and Pfeifer, G. P. Slow repair of pyrimidine dimers at p53 mutation esophagus and stomach. Chem. Bioh. Interact., 50: 15—25,1984.
hotspots in skin cancer. Science (Washington DC), 263: 1436—1439, 1994. 221. Aoyama, T., Yamano, S., Guzehian, P. 5., Gelboin, H. V., and Gonzalez, F. J. Five
193. Balch, C. M., Houghton, A. N., and Peters, L. J. Cutaneous melanoma. in: V. T. of 12 forms of vaccinia virus-expressed human hepatic cytochrome P450 metabol
DeVita, S. Hehhman, and S. A. Rosenberg (eds.), Cancer: Principles and Practice of ically activate aflatoxin B,. Proc. Nath. Acad. Sci. USA, 87: 4790—4793, 1990.
Oncology, pp. 1612—1662.Philadelphia: J. B. Lippincott Co., 1993. 222. Lin, J. K., Miller, I. A., and Miller, E. C. 2,3-Dihydro-2-(guan-7-yl)-3-hydroxy
194. McGregor, J. M., Yu, C. C., Dublin, E. A., Barnes, D. M., Levison, D. A., and aflatoxin B,, a major acid hydrolysis product of aflatoxin B,-DNA or -ribosomal
MacDonald, D. M. p53 immunoreactivity in human malignant melanoma and RNA adducts formed in hepatic microsome-mediated reactions and in rat liver in
dysplastic naevi. Br. J. Dermatol., 128: 606—611, 1993. vivo. Cancer Res., 37: 4430—4438, 1977.
195. Kamb, A., Gruis, N. A., Weaver-Fehdhaus, J., Liu, Q., Harshman, K., Tavtigian, 223. Essigmann, J. M., Croy, R. G., Nadzan, A. M., Busby, W. F., Jr., Reinhold, V. N.,
S. V., Stockert, E., Day, R. S., Johnson, B. E., and Skolnick, M. H. A cell cycle Buchi, G., and Wogan, G. N. Structural identification of the major DNA adduct
regulator potentially involved in genesis of many tumor types. Science (Washington formed by aflatoxin B, in vitro. Proc. Nath. Acad. Sci. USA, 74: 1870—1874,1977.
DC), 264: 436—440, 1994. 224. McMahon, G., Davis, E. F., Huber, L J., Kim, Y., and Wogan, G. N. Character
196. Nebert, T., Miura, K., Wu, D. J., Lais, A., Takabayashi, K., and Carson, D. A. ization of c-Ki-ras and N-ms oncogenes in aflatoxin B,-induced rat liver tumors.
Deletions of the cychin-dependent kinase-4 inhibitor gene in multiple human can Proc. NaIl. Acad. Sci. USA, 87: 1104-1108, 1990.
cers. Nature (Land.), 368: 753—756,1994. 225. Levy, D. D., Groopman, J. D., Lim, S. E., Seidman, M. M., and Kraemer, K. H.
197. Beashey, R. P. Hepatitis B virus. The major etiology of hepatocellular carcinoma. Sequence specificity of aflatoxin B,-induced mutations in a phasmid replicated in
Cancer (Phila.), 61: 1942—1956,1988. xeroderma pigmentosum and DNA repair proficient human cells. Cancer Res., 52:
198. Beasley, R. P., Hwang, L. Y., Un, C. C., and Chien, C. S. Hepatocellular carcinoma 5668—5673, 1992.
and hepatitis B virus. A prospective study of 22,707 men in Taiwan. Lancet, 2: 226. Trottier, Y., Waithe, W. I., and Anderson, A. Kinds of mutations induced by
1129—1133,
1981. aflatoxin B, in a shuttle vector replicating in human cells transiently expressing
199. Koshy, R., and Hofschneider, P. H. Transactivation by hepatitis B virus may cytochrome P450IA2 cDNA. Mol. Carcinog., 6: 140—147,1992.
contribute to hepatocarcinogenesis. Curr. Top. Microbiol. Immunoh., 144: 265—281, 227. Aguilar, F., Hussain, S. P., and Cerutti, P. Aflatoxin B, induces the transversion of
1989. G to T in codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proc.
200. Kim, C. M., Koike, K., Saito, I., Miyamura, T., and Jay, G. HBx gene of hepatitis NatI. Acad. Sci. USA, 90: 8586—8590, 1993.
B virus induces liver cancer in transgenic mice. Nature (Land.), 351: 317—320,
1991. 228. Aguilar, F., Harris, C. C., Sun, T., Hollstein, M., and Cerutti, P. Geographic
201. Feitelson, M. A., Zhu, M., Duan, L-X., and Landon, W. T. Hepatitis B X antigen and variation of p53 mutational profile in nonmalignant human liver. Science (Wash.
4877

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research
p53 MUTATIONS AND CANCER

ingtonDC), 264: 1317—1319, 1994. M. C., and Jones, P. A. Distinct pattem of p53 mutations in bladder cancer:
229. DeMarini, D. M. Genotoxicity of tobacco smoke and tobacco smoke condensate. relationship to tobacco usage. Cancer Res., 53: 1162—1
166, 1993.
Mutat. Res., 114: 59—89, 1983. 258. Samet, J. M. Radon and lung cancer. J. Nail. Cancer Inst., 81: 745—757,
1989.
230. Shophand, D. R., Eyre, H. J., and Pechacek, T. F. Smoking-attributable cancer 259. Moolgavkar, S. H., Luebeck, E. G., Krewski, D., and Ziehinski, J. M. Radon,
mortality in 1991 : is lung cancer now the leading cause of death among smokers in cigarette smoke, and lung cancer: a re-analysis of the colorado plateau uranium
the United States? J. Nath. Cancer Inst., 83: 1142—1148, 1991. miners' data. Epidemiology, 4: 204—217,1993.
231 . Doll, R., and Peto, R. Cigarette smoking and bronchial carcinoma: dose and time 260. Neuberger, J. S. Residential radon exposure and hung cancer: an overview of
relationships among regular smokers and lifelong non-smokers. J. Epidemiol. Cam ongoing studies. Health Phys., 63: 503—509,1992.
mun. Health, 32: 303—313,1978. 261. Samet, J. M., and Homung, R. W. Review of radon and lung cancer risk. Risk Anal.,
232. U.S. Department of Health, Education and Welfare Smoking and Health. in: A 10: 65—75,1990.
Report of the Surgeon General. DHEW Publication No. (PHS) 79—50066,pp. 5-11, 262. Pershagen, G., Akerbiom, G., Axelson, 0., et a!. Residential radon exposure and
1988. lung cancer in Sweden. N. Engl. J. Med., 330: 159—164,1994.
233. Ronai, Z. A., Gradia, S., Peterson, L. A., and Hecht, S. S. G to A transitions and G
263. Taylor, J. A., Watson, M. A., Devereux, T. R., Michels, R. Y., Saccomanno, G., and
to T transversions in codon 12 of the Ki-ras oncogene isolated from mouse lung
Anderson, M. Mutational hotspot in the p53 gene in radon-associated lung tumors
tumors induced by 4-(methylnitrosamino)-l-(3-pyridyh)-l-butanone (NNK) and re
from uranium miners. Lancet, 343: 86—87,1993.
hated DNA methyhating and pyridyloxobutyhating agents. Carcinogenesis (Land.),
264. Scheffner, M., Wemess, B. A., Huibregtse, J. M., Levine, A. J., and Howley, P. M.
14:2419—2422, 1993.
The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the
234. Suzuki, H., Takahashi, T., Kuroishi,T., Suyama, M., Ariyoshi, Y., and Ueda, R. p53
degradation of p53. Cell, 63: 1129—1136, 1990.
mutations in non-small cell hung cancer in Japan: association between mutations and
smoking. Cancer Res., 52: 734—736, 1992. 265. Liang, X. H., Volkmann, M., Klein, R., Herman, B., and Lackett, S. J. Ca
235. Takeshima, Y., Scyama, T., Bennett, W. P., Metcalf, R. A., Welsh, J. A., Akiba, S., localization of the tumor-suppressor protein p53 and human papillomavirus E6
Fujihara, M., Hayashi, Y., Yonehara, S., Ito, T., Mizuno, T., hnai, K., Yamakido, M., protein in human cervical carcinoma cell lines. Oncogene, 8: 2645—2652,1993.
Nakamura, N., Akiyama, M., Tokuoka, S., Mabuchi, K., Land, C. E., and Harris, 266. Srivastava, S., Tong, Y. A., Devadas, K., Thu. Z. Q., Chen, Y., Pirollo, K. F., and
C. C. p53 gene mutations in lung cancers from Japanese non-smokers and atomic Chang, E. H. The status of the p53 gene in human papilloma virus positive or negative
bomb survivors. Lancet, 342: 1520—1521,1993. cervical carcinomacell lines. Carcinogenesis(Land.), 13: 1273—1275,
1992.
236. Hanawalt, P., and Mellon, I. Stranded in an active gene. Curr. Biol., 3: 67—69,1993. 267. Crook, T., Wrede, D., Tidy, J. A., Mason, W. P., Evans, D. J., and Vousden, K. H.
237. Grafstrom, R. C., Fornace, A. J., Jr., Autrup, H., Lechner, J. F., and Harris, C. C. Chonahp53 mutation in primary cervical cancer: association with human-papilloma
Formaldehyde damage to DNA and inhibition of DNA repair in human bronchial virus-negative tumours. Lancet, 339: 1070—1073, 1992.
cells. Science (Washington DC), 220: 216—218, 1983. 268. Borresen, A. L, Helland, A., Nesland, J., HoIm, R., Trope, C., and Kaern, J.
238. McDowell, E. M., and Trump, B. F. Histogenesis of preneoplastic and neophastic Papillomaviruses, p53, and cervical cancer. Lancet, 339: 1350—1351, 1992.
lesions in tracheobronchial epithehium. Sun'. Synth. Pathol. Res., 2: 235—279,1983. 269. Fujita, M., Inoue, M., Tanizawa, 0., Iwamoto, S., and Enomoto, T. Alterations of the
239. Berendsen, H. H., de Leij, L., Poppema, S., Postmus, P. E., Boes, A., Sluiter, H. J., p53 gene in human primary cervical carcinoma with and without human papilho
and The, H. Clinical characterization of non-small-cell hung cancer tumors showing mavirus infection. Cancer Res., 52: 5323—5328, 1992.
neurocndocrine differentiation features. J. Chin. Oncoh., 7: 1614—1620, 1989. 270. Busby-Earle, R. M., Steel, C. M., Williams, A. R., Cohen, B., and Bird, C. C.
240. Graziano, S. L, Mazid, R., Newman, N., Tatum, A., 01cr, A., Mortimer, J. A., Papilhomaviruses, PS3, and cervical cancer. Lancet, 339: 1350, 1992.
Gulho, J. J., DiFino, S. M., and Scahzo, A. J. The use of neuroendocrine immunoper 271. Naito, M., Satake, M., Sakai, E., Hirano, Y., Tsuchida, N., Kanzaki, H., Ito, Y., and
oxidase markers to predict chemotherapy response in patients with non-small-cell Mori, T. Detection ofp53 gene mutations in human ovarian and endometrial cancers
hung cancer. J. Chin. Oncol., 7: 1398—1406, 1989. by pohymerase chain reaction-single strand conformation polymorphism analysis.
241. Takahashi, T., Suzuki, H., Hida, T., Sekido, Y., Ariyoshi, Y., and Ueda, R. The p53 Jpn. J. Cancer Rca., 83: 1030—1036,1992.
gene is very frequently mutated in small-cell hung cancer with a distinct nucleotide 272. Willey, J. C., and Harris, C. C. Cellular and molecular biological aspects of human
substitution pattem. Oncogene, 6: 1775—1778,1991. bronchogemc carcinogenesis. CRC Crit. Rev. Hematol. Oncol., 10: 181—209,
1990.
242. Miller, C. W., Simon, K., Asho, A., Kok, K., Yokota, J., Buys, C. H. C. M., Terada, 273. Jones, P. A., Buckley, J. D., Henderson, B. E., Ross, R. K., and Pike, M. C. From
M., and Koeffler, H. P. p.53 mutations in human hung tumors. Cancer Rca., 52: gene to carcinogen: a rapidly evolving field in molecular epidemiology. Cancer Res.,
1695—1698,1992. 51: 3617—3620,1991.
243. D'Amico, D., Carbone, D., and Mitsudomi, 1. High frequency of somatically 274. Nagase, H., and Nakamura, Y. Mutations of the APC gene. Hum. Mutat., 2:
acquired p53 mutations in small cell lung cancer cell lines and tumors. Oncogene, 425-434, 1993.
7: 339—346,1992. 275. Giannehhi,F., Green, P. M., High, K. A., et aL HaemophihiaB: database of point
244. Sameshima, Y., Matsuno, Y., Hirohashi, S., Shimosato, Y., Mizoguchi, H., Sug mutations and short additions and deletions—fourthedition, 1993. Nucleic ACIdS
imura, T., Terada, M., and Yokota, J. Alterations of the p53 gene are common and Res., 21: 3075—3087,1993.
critical events for the maintenance of malignant phenotypes in small-cell lung 276. Sugimura, T. Studies on environmental chemical carcinogenesis in Japan. Science
carcinoma. Oncogene, 7: 451—457, 1992. (Washington DC), 233: 312—318,1986.
245. Kishimoto, Y., Murakami, Y., Shiraishi, M., Hayashi, K., and Sekiya, T. Aberrations 277. Ziehenska, M., Beranek, D., and Guttenphan, J. B. Different mutational profiles
of the p53 tumor suppressor gene in human non-small cell carcinomas of the hung. induced by N-nitroso-N-ethylurea: effects of dose and error-prone DNA repair and
Cancer Res., 52: 4799—4804,1992. correlations with DNA adducts. Environ. Mol. Mutagen., 11: 473—485, 1988.
246. Lohmann, D., Putz, B., Reich, U., Bohm, J., Prauer, H., and Hofler, H. Mutational 278. Singer, B. in vivo formation and persistence of modified nucheosides resulting from
spectrum of the p53 gene in human small-cell hung cancer and relationship to alkylating agents. Environ. Health Perspect., 62: 41—48,1985.
chinico-pathohogicah data. Am. J. Pathol., 142: 907—915,1993. 279. Pe@, A. E. Methyhation of the O@position of guanine in DNA is the most likely
247. Osann, K. E., Anton-Culver, H., Kurosaki, T., and Taylor, 1. Sex differences in initiating event in carcinogenesis by methyhating agents. Cancer Invest., 2: 223—231,
hung-cancer risk associated with cigarette smoking. Int. J. Cancer, 54: 44—48,1993. 1984.
248. Jedrychowski, W., Becher, H., Wahrendorf, J., Basa-Cierpialek, Z., and Gomola, K. 280. Sasa, M., Kondo, K., Komaki, K., Uyama, T., Morimoto, T., and Monden, Y.
Effect of tobacco smoking on various histological types of lung cancer. J. Cancer Frequency of spontaneous p53 mutations (CpG site) in breast cancer in Japan.
Res. Chin. Oncol., 118: 276—282, 1992. Breast. Cancer Rca. Treat., 27: 247—252,
1993.
249. Stockwell, H. G., Goldman. A. L, Lyman, G. H., Noss, C. I., Armstrong, A. W., 281. de Jong, P. J., Grosovsky, A. J., and Ghickman, B. W. Spectrum of spontaneous
Pinkham,P. A., Candelora,E. C., and Brusa, M. R. Environmentaltobaccosmoke and mutation at the APRT locus of Chinese hamster ovary cells: an analysis at the DNA
hung cancer risk in nonsmoking women. J Nati. Cancer Inst., 84: 1417—1422,1992. sequence level. Proc. Nati. Acad. Sci. USA, 85: 3499—3503, 1988.
250. Blum, A. Curtailing the tobacco pandemic. in: V. T. DeVita, S. Hehhman,and S. A. 282. Drobetsky, E. A., Grosovsky, A. J., and Ghickman, B. W. The specificity of
Rosenberg (eds.), Cancer: Principle and Practice of Oncology, pp. 480—491.Phil UV-induced mutations at an endogenous locus in mammalian cells. Proc. NatI.
adehphia: J. B. Lippincott, 1993. Acad. Sci. USA, 84: 9103—9107,
1987.
251. Szekehy, L., Sehivanova, G., Magnusson, K. P., Klein, G., and Wiman, K. G. 283. Mazur, M., and Ghickman, B. W. Sequence specificity of mutations induced by
EBNA-5, an Epstein-Barr virus-encoded nuclear antigen, binds to the retinobhastoma benzo[a]pyrene-7,8-dioh-9,10-epoxide at endogenous aprt gene in CHO cells.
and p53 proteins. Proc. Nath.Acad. Sci. USA, 90: 5455—5459,
1993. Somat. Cell Mol. Genet., 14: 393—400,1988.
252. Nishihira, T., Hashimoto, Y., Katayama, M., Mon. S., and Kuroki, T. Molecular and 284. Halevy, 0., Rodeh, J., Pehed, A., and Oren, M. Frequentp53 mutations in chemically
cellular features of esophageal cancer cells. J. Cancer Res. Clin. Oncol., 119: induced murine fibrosarcoma. Oncogene, 6: 1593—1600,1991.
441—449,1993. 285. Lozano, J. C., Nakazawa, H., Cros, M. P., Cabral, R., and Yamasaki, H. G—@ A
253. Blot, W. J., Devesa, S. S., Kneller,R. W., and Fraumeni,J. F., Jr. Rising incidenceof mutations in p53 and Ha-ras genes in esophageal papillomas induced by N-
adenocarcinomaof the esophagusand gastric cardia. JAMA, 265: 1287—1289, 1991. nitrosomethyhbenzyhamine in two strains of rats. Mol. Carcinog., 9: 33—39,
254. Blot, W. J., Devesa, S. S., and Fraumeni, J. F., Jr. Continuing climb in rates of 1994.
esophageal adenocarcinoma: an update. JAMA, 270: 1320, 1993. 286. Smith, M. L, Yeheswarapu, L, Scalamogna, P., Locker, J., and Lambardi, B. p53
255. Hartge, P., Silverman, D., Hoover, R., el a!. Changing cigarette habits and bladder mutations in hepatocelluharcarcinomas induced by a choline-devoid diet in male
cancer risk: a case-control study. J. Nath.Cancer Inst., 78: 1119—1125, 1987. Fischer-344 rats. Carcinogenesis (Land.), 14: 503—510,1993.
256. Zhang, Z. F., Sarkis, A. S., Cordon-Cardo, C., Dalbagni, G., Mehamed,J., Aprikian, 287. Hegi, M. E., Soderkuist, P., Foley, J. F., et a!. Characterization of p53 mutations in
A., Pohiack,D., Sheinfeld, J., Herr, H. W., Fair, W. R., Reuter, V. E., and Begg, C. methyhene chloride-induced lung tumors from B6C3F1 mice. Carcinogenesis
Tobacco smoking, occupation, and p53 nuclear overexpression in early stage bladder (Land.), 14: 803—810,1993.
cancer. Cancer Epidemiol. Biomarkers Prey., 3: 19—24,1994. 288. Goodrow, T. L., Storer, R. D., Leander, K. R., et a!. Murine p53 intron sequence 5—8
257. Spruck, C. H., III, Rideout, W. M., III, Olumi, A. F., Ohneseit, P. F., Yang, A. S., and their use in pohymerase chain reaction/direct sequencing analysis of p53 muta
Tsai, Y. C., Nichols, P. W., Horn, T., Hermann, G. G., Steven, K., Ross, R. K., Yu, tions in CD-i mouse liver and hung tumors. Mol. Carcinog., 5: 9—15,1992.

4878

Downloaded from cancerres.aacrjournals.org on March 3, 2011


Copyright © 1994 American Association for Cancer Research

You might also like