You are on page 1of 43

3

Mechanisms of tumour development


The phenotypic changes which a cell undergoes in the
process of malignant transformation is a reflection of the
sequential acquisition of genetic alterations. This multi-step
process is not an abrupt transition from normal to malignant
growth, but may take place over 20 years or more. The muta-
tion of critical genes, including suppressor genes, oncogenes
and genes involved in DNA repair, leads to genetic instability
and progressive loss of differentiation. Tumours enlarge
because cancer cells lack the ability to balance cell division
by cell death (apoptosis) and by forming their own vascular
system (angiogenesis). The transformed cells lose their abili-
ty to interact with each other and exhibit uncontrolled
growth, invade neighbouring tissues and eventually spread
through the blood stream or the lymphatic system to distant
organs.
MULTISTAGE CARCINOGENESIS

SUMMARY During this process the cell develops :


- Defects in terminal differentiation
> Tumours consist of cells whose growth - Defects in growth control
- Resistance to cytotoxicity
and morphological characteristics are - Defects in programmed cell death
markedly different from those of normal
cells. Criteria for malignancy include
increased cell proliferation, loss of differ- Selective
entiation, infiltrative growth and metasta- CHEMICALS Genetic clonal Genetic Genetic Genetic
change expansion change change change
sis to other organs.

> Malignant transformation is a multistage VIRUS


process, typically a progression from
PRE-
benign lesions (e.g. adenoma) to malig- NORMAL INITIATED NEOPLASTIC MALIGNANT CLINICAL CANCER
nant tumours (e.g. carcinoma). This evo- RADIATION CELL CELL LESION TUMOUR CANCER METASTASIS
lution of malignant cells is caused by the
sequential accumulation of alterations
in genes responsible for the control of
cellular proliferation, cell death and the These steps are caused by :
- Activation of proto-oncogenes
maintenance of genetic integrity.
- Inactivation of tumour suppressor genes
- Inactivation of genomic stability genes
> The development of cancer may be initi-
ated by environmental agents (chemical
carcinogens, radiation, viruses) and
inherited genetic factors (germline Fig. 3.1 Carcinogenesis is a multistage process involving multiple genetic and epigenetic events in proto-
mutations). oncogenes, tumour suppressor genes and anti-metastasis genes.

Cancer arises from a single cell


Malignant tumours (or cancers) are specific cell populations may be identified cells or tissues exposed to them. DNA-
described as monoclonal, meaning that as marking a commitment towards malig- damaging activity may be identified on the
each tumour arises from a single cell. The nancy, and these may be exploited as an basis of defined protocols (sometimes
development of a malignant tumour from early indicator in the context of carcino- called short-term tests, to emphasize
a normal cell usually occurs over a con- gen testing [3]. Thus, wholly on morpho- their difference from chronic lifetime
siderable fraction of our lifetime. Such a logical grounds, cancer may be perceived bioassay in rodents). Chemicals which
long period is reflected, for example, by as the outcome of a complex biological exhibit mutagenic activity in short-term
the difference between the age at which process. tests, which typically involve sensitive
people start smoking and the age at bacterial strains and cell-free extracts to
which diagnosis of lung cancer most Multiple steps are required for a can- catalyse metabolism of the test com-
often occurs. The long latent period in cer to arise pound, are characterized as genotoxic
lung cancer and almost all other malig- Animal models of cancer development, [5]. Genotoxic agents may be complete
nancies is not explicable on the basis of a most commonly involving treatment of carcinogens, but can also act as initiating
single-step transition from a normal cell rodents with carcinogenic chemicals or agents. After a single treatment with an
to malignant one. Rather, the tumour is other cancer-inducing agents, have pro- initiating agent, tumour growth may be
the outcome of an evolutionary process vided clear evidence that specific stages facilitated by chemicals (or treatments)
involving successive generations of cells, in malignant transformation can occur dis- which stimulate cell proliferation, some-
which are progressively further advanced cretely [4]. Chemicals which cause cancer times by inducing mild toxic damage in
towards cancerous growth [1]. in animals without the need for other exposed tissue. These agents are termed
Human histopathological observations treatment are sometimes called com- promoters (Table 3.1). As well as these
support this scenario, and a range of pre- plete carcinogens (although carcino- genotoxic chemicals, a range of non-geno-
malignant lesions have been identified gens would be appropriate). Most such toxic agents can cause cancer in humans
[2]. Likewise, in experimental animals, carcinogens cause damage to DNA of and/or experimental animals [6].

84 Mechanisms of tumour development


The stages in tumorigenesis have been
designated initiation, which encom-
passes damage to, and then division of Factor Cancer site/cancer
exposed cells such that their growth
potential is changed irreversibly, and Hormones Estrogens, progesterone Uterus, mammary gland
progression, denoting multiple rounds Gonadotrophins Ovary, testis, pituitary
Testosterone Prostate gland
of cell replication mediating the gradual
transition of an initiated cell towards Pharmaceutical Oral contraceptives Liver
autonomous, cancerous, growth. Ultimate products Anabolic steroids Liver
spread of malignant cells resulting in mul- Analgesics Renal pelvis
tiple tumour sites has been termed
metastasis. The unequivocal identifica- Miscellaneous Bile acids Small intestine
tion by the mid-1970s of these various substances Saturated fatty acids Colon
phases was one indication that carcino- Salt Stomach
genesis is a multistage process. Arguably, Tobacco Oral cavity, lung, bladder etc.
the greatest achievement of cancer Saccharin, uracil, melamine, Urinary bladder
research during the last decades of the tetraphthalic acid and other
20th century has been the elucidation of xenobiotics causing urinary stones
Dichlorobenzene, trimethylpentane Kidney
multistage carcinogenesis at the molecu-
(lead-free gasoline), perchloroethyl-
lar genetic level.
ene
Butylated hydroxyanisole, propionic Stomach
The molecular basis of tumour acid
pathology Nitrilotriacetate Kidney
In a seminal publication, Vogelstein and
colleagues [7] provided evidence that Table 3.1 Promoting agents: non-genotoxic agents that facilitate carcinogenesis by stimulating cell division.
the different stages in the cellular evo- Tobacco smoke also contains genotoxic carcinogens.
lution of colon cancer in humans, histo-
logically identified as hyperplasia, CHROMOSOME: 5q 12p 18q 17p
early-stage adenoma, late-stage adeno- ALTERATION: Mutation Mutation Loss Loss
GENE: FAP KRAS DCC? p53
ma etc., could be identified with specif-
ic successive genetic changes (Fig.
3.2). The genetic changes included
oncogene activation by mutation at DNA Other
specific sites and loss of chromosomal hypomethylation alterations
regions (necessarily involving multiple
genes) which were subsequently shown
to be the location of tumour suppressor Normal Hyperproliferative Early Intermediate Late
Carcinoma Metastasis
genes. Since that initial description, epithelium epithelium adenoma adenoma adenoma
knowledge of the molecular genetic
basis for human colon cancer has been Fig. 3.2 The original Vogelstein model for the genetic and histological evolution of colon cancer.
massively extended (Colorectal cancer, (Colorectal cancer, p198).
p198). For most tumours, the genetic
changes are not inherited from our par-
ents but arise in a previously normal Commonality and heterogeneity amplification) are common to a number
cell. The progeny of this cell after cell The molecular biological basis of multi- of tumour types. However, each tumour
division carry the same genetic change stage carcinogenesis initially described type is associated with a distinctive set
but the surrounding cells remain nor- for colon cancer appears to have appli- of gene alterations. The genes in ques-
mal. Because these genetic changes cation to all tumour types, although tion are discussed under the subhead-
affect only the cancer cells, they are there is marked variation in the extent ing Pathology and genetics for each of
not passed on to the children of cancer to which genes relevant to particular the tumour types included in Chapter 5.
patients. However, in a minority of tumours have been identified [8]. Some Such enumeration of relevant genes
cases some critical changes are inherit- genes, and the corresponding change necessitates a degree of simplification.
ed, giving a familial predisposition to associated with tumorigenesis (muta- There is clear heterogeneity between
colon or other cancers. tion, overexpression, deletion and/or individual tumours of the same type. In

Multistage carcinogenesis 85
Peutz-Jeghers polyp

RER+ cancer
(Replication
Dysplasia in hamartoma air Error Positive)
rep
atch
ism
s of m
Los
Juvenile polyp
Normal Early adenoma Intermediate adenoma Late adenoma Cancer

Flat dysplasia Ulcerative colitis-associated


colorectal carcinoma

MHAP/Serrated adenoma Cancer in mixed hyperplastic adenomatous


polyps (MHAP)

Flat adenoma Flat cancer

Fig. 3.3 Histological representation of the pathogenesis of colorectal cancer. Phenotypic changes in the morphology of the colonic mucosa reflect the sequen-
tial acquisition of genetic alterations.

other words, not every tumour will nec- and tumour suppressor genes, p96) have esis are being progressively identified
essarily exhibit all the genetic changes been identified in terms of their biological [11]. As noted earlier, members of some
established for the tumour type in ques- function [9]. Such genes are among those cancer-susceptible families inherit muta-
tion. Moreover, there is often marked that facilitate transmission of growth con- tions in particular genes that contribute to
heterogeneity within an individual trol signals from the cell membrane to the cancer development, and hence to their
tumour: adjacent cells differ. Mapping nucleus (that is, signal transduction), that individual risk of disease. However, with
and identification of genes involved in mediate cell division, differentiation or cell most cancers, the genetic change critical
malignant transformation has been a death and, perhaps most critical of all, to carcinogenesis results from damage to
major component of the study of the that maintain the integrity of genetic infor- DNA by chemicals, radiation and viruses
molecular mechanisms of carcinogene- mation by DNA repair and similar process- (Fig. 3.1). This damage is not entirely and
sis. es (Carcinogen activation and DNA repair, perhaps not predominantly produced by
p89). Since mutations are normally infre- exogenous agents but by natural process-
Multiple genetic changes required quent events, it seems unlikely that in the es, such as the production of reactive oxy-
The emergence of a malignant cell popu- course of a human lifetime a cell would gen species or the spontaneous deamina-
lation is understood to be the cumulative acquire all the mutations necessary for tion of the 5-methylcytosine naturally
effect of multiple (perhaps five, ten or cancer to develop, unless at some point present in DNA [13]. Furthermore, as
more) genetic changes, such changes the developing cell lost its ability to pro- shown as the second step in Fig. 3.2, bio-
being accumulated in the course of the tect itself against mutation and gained logical change that is heritable may result
evolution of the cell from normal to malig- what is called a mutator phenotype [10]. from non-genetic processes including the
nant. The genes designated as oncogenes Thus, alterations in gene structure and modulation of gene expression by hyper-
and tumour suppressor genes (Oncogenes expression which bring about carcinogen- methylation [12].

86 Mechanisms of tumour development


PRECURSOR LESIONS IN Engl J Med, 328: 901-906, 1993). Several
CHEMOPREVENTION TRIALS epidemiological studies have shown that
regular use of aspirin or related drugs is
Trials of agents for chemopreventive associated with a reduced adenoma inci-
activity which are based on assessment dence (IARC Handbooks of Cancer
of malignant disease are almost unman- Prevention. Vol. 1, Lyon, 1997). This pro-
ageable because of the long period of vides further confirmation that adenomas
time (perhaps decades) potentially are precursor lesions for colon cancer,
involved. Attention has therefore been since aspirin is known to reduce the inci-
focused on lesions, either cellular or dence of malignant colon cancer.
molecular, demonstrated to be valid indi- Fig. 3.6 Tubular adenoma of the colon is a precur-
cators of the subsequent development of Potential precursor lesions of carcinogene- sor lesion for colorectal cancer.
malignancy. A trial may then evaluate the sis include both phenotypic and genotypic
effect of the putative chemopreventive markers (Miller AB et al. Biomarkers in
agent on such precursor lesions. Cancer Chemoprevention, IARC Scientific cific gene and general chromosomal dam-
Publications 154, Lyon, 2001). Thus oral age, cell growth regulatory molecules,
The best-validated precursor lesions are leukoplakia is a recognized precursor for and biochemical activities (e.g. enzyme
benign tumours, such as colorectal ade- cancer of the oral cavity. Histological mod- inhibition). Serum proteins are of special
nomas. It is established that adenoma ulation of a precancer (often called intraep- interest because of their availability. Thus
number, size, and severity of dysplasia are ithelial neoplasia) has been used as a pre- prostate-specific antigen (PSA) is being
predictive factors for colorectal cancer cursor lesion in prevention trials (Kelloff GJ used as a surrogate marker for prostate
incidence. It has been estimated that 2- et al., Cancer Epidemiol Biomarkers Prev, 9: cancer. It is expected that the number and
5% of all colorectal adenomas progress to 127-137, 2000). Additionally, genetic variety of biomarkers for precursor
adenocarcinomas if not removed or treat- lesions such as progressive genomic insta- lesions will continue to expand In parallel
ed. The risk is greater for large and bility as measured by loss of heterozygosi- with the advances in understanding of the
severely dysplastic polyps. Cancer risk is ty or amplification at specific microsatellite genetic and cellular basis of carcinogene-
decreased by polyp removal, and a strong loci, have been considered (Califano J et al. sis.
correlation exists between the relative Cancer Res, 56: 2488-2492, 1996). Other
prevalence of adenomas and cancers potential precursor endpoints include pro-
across populations (Winawer SJ et al., N liferation and differentiation markers, spe-

Ageing also at the cellular level. In humans, as well when further maintained in culture, once-
Apart from multistage development, cer- as in other mammals, the incidence of normal cells acquire the same characteris-
tain other processes are fundamental to cancer rises dramatically with age. An tics as cells cultured from malignant
malignant disease. Principal amongst exponential increase occurs from mid-life tumours. These and various other alter-
these is ageing, which can be considered [14]. Passage of time is also critical to cell ations in growth characteristics are recog-
both in relation to the whole individual, and biology. Normal cells do not divide indefi- nized as the experimental counterpart of
nitely due to senescence (Box: Telomeres multistage carcinogenesis through which
and Telomerase, p108). Senescent cells tumours develop in intact animals or
cannot be stimulated to divide further, humans. The genetic basis for senescence,
become resistant to apoptotic cell death and its relationship to malignancy, is a sub-
and acquire differentiated functions. ject of intense investigation [15].
Senescence may be an anti-cancer mech-
anism that limits accumulation of muta- Preventing cancer
tions. However, when maintained in cul- The significance of multistage carcino-
ture, cells treated with carcinogenic chem- genesis extends beyond facilitating
icals or infected with oncogenic viruses understanding of how a transition from
Fig. 3.4 Severe intraepithelial neoplasia (dyspla-
may avoid senescence and proliferate normal to malignant cell growth occurs.
sia) in the epithelium of an intrahepatic large bile indefinitely. Such cell populations are The fundamental cellular studies outlined
duct, a condition caused by hepatolithiasis. described as being transformed and earlier provide a basis for preventing can-

Multistage carcinogenesis 87
cer (see chapter 4). The fact that partic- mechanisms now known to operate in
ular patterns of cell morphology and the proliferation of cancer cells provide a
growth precede emergence of an basis for the development of new, more
unequivocally malignant cell population efficient therapies without the side-
is the basis of secondary prevention of effects that currently often afflict cancer
cancer. patients [17].
Examples include detection of polyps in
the large bowel (Fig. 3.5) and of morpho-
logical change which is the basis of the
Papanicolaou smear test for early detec-
tion of cervical cancer. Moreover, dietary
or pharmaceutical interventions calculat-
ed to prevent or reverse such lesions are
the basis of chemoprevention [16]. Most
importantly, knowledge of the genetic
basis underlying tumour growth should
provide new criteria for individual deter-
Fig. 3.5 Pedunculated hyperplastic polyp of the mination of diagnosis and prognosis. The
colon.

REFERENCES
1. Foulds L, ed. (1969) Neoplastic Development, Vol. 1, mechanisms: a European project. Mutat Res, 353: 47-63. 13. Marnett LJ, Plastaras JP (2001) Endogenous DNA
London, Academic Press. damage and mutation. Trends Genet, 17: 214-221.
7. Vogelstein B, Fearon ER, Hamilton SR, Kern SE,
2. Correa P (1996) Morphology and natural history of can- Preisinger AC, Leppert M, Nakamura Y, White R, Smits AM, 14. Armitage P, Doll R (1954) The age distribution of can-
cer precursors. In: Schottenfeld D, Fraumeni JF, eds, Bos JL (1988) Genetic alterations during colorectal-tumor cer and a multistage theory of carcinogenesis. Br J Cancer,
Cancer Epidemiology and Prevention, New York, Oxford development. N Engl J Med, 319: 525-532. 8: 1-12.
University Press, 45-64. 15. Wynford-Thomas D (1999) Cellular senescence and
8. Balmain A, Harris CC (2000) Carcinogenesis in mouse
3. Ito N, Imaida K, Asamoto M, Shirai T (2000) Early and human cells: parallels and paradoxes. Carcinogenesis, cancer. J Pathol, 187: 100-111.
detection of carcinogenic substances and modifiers in rats. 21: 371-377. 16. Bartsch H (2000) Studies on biomarkers in cancer
Mutat Res, 462 : 209-217. etiology and prevention: a summary and challenge of 20
9. Evan GI, Vousden KH (2001) Proliferation, cell cycle
4. Weinstein IB (1982) Carcinogenesis as a multistage and apoptosis in cancer. Nature, 411: 342-348. years of interdisciplinary research. Mutat Res, 462: 255-
processexperimental evidence. In: Bartsch H, Armstong 279.
B, eds, Host Factors in Human Carcinogenesis (IARC 10. Loeb LA (2001) A mutator phenotype in cancer.
17. Kallioniemi OP, Wagner U, Kononen J, Sauter G
Scientific Publications No. 39) Lyon, IARCPress, 9-25. Cancer Res, 61: 3230-3239.
(2001) Tissue microarray technology for high-throughput
5. Vainio H, Magee PN, McGregor DB, McMichael AJ, eds 11. Hahn WC, Counter CM, Lundberg AS, Beijersbergen molecular profiling of cancer. Hum Mol Genet, 10: 657-
(1992) Mechanisms of Carcinogenesis in Risk Identification RL, Brooks MW, Weinberg RA (1999) Creation of human 662.
(IARC Scientific Publications No. 116), Lyon, IARCPress. tumour cells with defined genetic elements. Nature, 400:
464-468.
6. Yamasaki H, Ashby J, Bignami M, Jongen W, Linnainmaa
K, Newbold RF, Nguyen-Ba G, Parodi S, Rivedal E, 12. Esteller M, Corn PG, Baylin SB, Herman JG (2001) A
Schiffmann D, Simons JW, Vasseur P (1996) Nongenotoxic gene hypermethylation profile of human cancer. Cancer
carcinogens: development of detection methods based on Res, 61: 3225-3229.

88 Mechanisms of tumour development


CARCINOGEN ACTIVATION AND DNA REPAIR

Carcinogen activation carbons, aromatic amines, N-nitro-


SUMMARY The first indication that certain cancers samines, aflatoxins and vinyl halides,
were associated with exposure to chemi- which yield electrophilic species through
>Many chemical carcinogens require cals arose from observations by clini- phase I activation [3]. Other metabolic
spontaneous or enzymatic activation to cians in the 18th and 19th centuries. The pathways are known. For example,
produce reactive intermediates which
field of experimental chemical carcino- dihaloalkanes are activated to carcino-
bind to DNA. The resulting carcinogen-
DNA adducts may be eliminated from genesis started in 1915 with the experi- genic metabolites by glutathione trans-
DNA by various enzyme-mediated repair ments of Yamagiwa and Ichikawa, who ferases.
processes. showed that application of tar to the ears Understanding of carcinogen-DNA inter-
of rabbits induced skin tumours. In the actions (Fig. 3.9) has resulted largely
> In cells and tissues with deficient DNA 1940s, experiments on mouse skin from the development of sensitive and
repair, replication of carcinogen-dam- demonstrated the stepwise evolution of specific methods for determining DNA
aged DNA may result in the mutation of cancer and allowed the characterization adducts [4]. The most frequently used
genes that regulate cell growth and dif- of two classes of agents, initiators and methods include immunoassays using
ferentiation in target cell populations. promoters [1]. Most chemical carcino- adduct-specific anti-sera or antibodies,
Such genetic alterations typically lead
gens are subject to metabolism that 32 P-postlabelling, fluorescence spec-
to progressive genetic instability result-
ing in uncontrolled growth, loss of dif- results in their elimination, but in the troscopy, electrochemical detection and
ferentiation, invasion and metastasis. course of which reactive intermediates mass spectrometry. Measurement of car-
are generated. Such metabolic activation cinogen-DNA adducts in rodents has
results in the modification of cellular revealed correlations between the con-
macromolecules (nucleic acids and pro- centration of the carcinogen in the envi-
teins) [2]. Accordingly, mutagenicity ronment, DNA adduct levels in tissues
tests using bacteria and mammalian cells where tumours may arise and cancer
Experimental studies in rodents and in in culture were developed and are exten- incidence. It is therefore accepted that
cultured cells have led to the classifica- sively used to identify potential carcino- DNA adducts may be used as indicators
tion of chemical carcinogens into two gens. Not all chemicals known to cause
broad classes: genotoxic and non-geno- cancer, however, can be demonstrated to
toxic. Genotoxic carcinogens alter the bind to DNA and hence be classified as
structure of DNA, mostly by covalent genotoxic. PROCARCINOGEN
binding to nucleophilic sites. These Activation of chemical carcinogens in mam-
lesions, that is, the chemical entity of malian tissue mostly occurs through oxida- Detoxification Metabolic activation
carcinogen bound to DNA, are called tion by microsomal mono-oxygenases
DNA adducts. The replication of DNA (cytochromes P450, phase I enzymes). Ultimate carcinogen
containing unrepaired adducts may Cytochromes P450 are located in the
result either in the generation of endoplasmic reticulum (internal mem- Covalent binding to
DNA, RNA, proteins
sequence changes (mutations) in the branes of the cell) and constitute a super-
newly synthesized daughter strands of family of proteins; about 50 are now
Promutagenic DNA
DNA or in DNA rearrangements evident known in humans. The oxidation products adducts
as chromosome aberrations. are substrates for other families of
This critical, irreversible genetic event enzymes (transferases, phase II enzymes) No or error-prone
can thus result in fixation of the original which link the carcinogen residues to a DNA repair DNA repair
structural change in DNA as permanent, glutathione, acetyl, glucuronide or sulfate
transmissible, genetic damage, or in t he group; the resulting conjugates are Cell replication Cell replication with
loss of ge netic information through hydrophilic and thus can be easily excret- with no DNA sequence changes
alterations in chromosomes. Such heri- ed. Carcinogenic electrophilic metabo- DNA sequence changes (gene mutations)

tab le change has the po tential to per- lites arise as by-products of these meta-
turb gro wth control in the affected cell, bolic reactions. The metabolic pathways NORMAL CELLS INITIATED CELLS
and is sometimes referred to as the ini- are well characterized for the major
tiation step of the tumorigenic process classes of chemical carcinogens (Fig. Fig. 3.7 Critical stages in the process of initiation
(Fig. 3.7). 3.8), including polycyclic aromatic hydro- by genotoxic chemicals.

Carcinogen activation and DNA repair 89


of the effective biological exposure, and adducts, together with other genetic polymorphisms involved, large-scale stud-
hence of carcinogenic risk in humans [5]. determinants that regulate DNA repair or ies and high throughput assays (based on
However, analysis of DNA adducts in cell cycle control, for example, and thus DNA microchips, for example) will be
human cells and tissues remains diffi- affect the outcome of exposure to DNA- required to fully elucidate the complex
cult, due to the very low levels of adducts damaging agents and influence cancer nature of such gene-environment interac-
present in DNA (typically, one adduct per risk in different individuals [6]. Many stud- tions.
107-108 parent nucleotides). ies have sought to correlate genetic poly-
Activities of the enzymes involved in car- morphisms, adduct levels and cancer risk Mutational spectra
cinogen metabolism vary greatly between in human populations (Genetic suscepti- Adducts of DNA and proteins can be used
individuals due to induction and inhibition bility, p71). These studies have hitherto as early markers of exposure to carcino-
processes or to gene polymorphisms that provided some correlations for risk pre- gens as indicated. However, because
can affect activity. These variations can diction at the population level. However, adducts only persist for a short time (typ-
affect the formation of carcinogen-DNA due to the great number of enzymes and ically, for a few hours or days for DNA

Fig. 3.8 Carcinogen activation by mammalian enzymes: reactions catalysed during metabolism of benzo[a]pyrene and NNK (4-(methylnitrosamino)-1-(3-
pyridyl)-1-butanone), both contained in tobacco, and of aflatoxin B1, produce reactive intermediates (ultimate carcinogens, in box), which bind to DNA. Other
reaction pathways leading to the formation of glucuronides and other esters, which are excreted, are not shown. 1. Benzo{a}pyrene-7, 8-diol-9, 10-epoxide; 2. 4-(methyl-
nitrosamino)-1-(3-pyridyl)-1-butanol; 3. Diazohydroxide; 4. Diazohydroxide; 5. Aflatoxin B1-8,9-oxide; 6. 2,3-Dihydro-2-(N7- guanyl)-3-hydroxyaflatoxin B1.

90 Mechanisms of tumour development


adducts, a few weeks or months for albu- ribosyl-transferase gene HPRT, when inac- of almost all tumour types. A large data-
min or haemoglobin adducts), their use- tivated by mutation, renders cells resist- base of p53 mutations has been generat-
fulness as exposure markers is limited. ant to growth inhibition by 6-thioguanine; ed. Mutational spectra have been identi-
Mutations in specific genes can be used such mutant cells can therefore be isolat- fied that provide evidence for the direct
as longer-term biomarkers of early bio- ed by culture in the presence of this action of environmental carcinogens in
logical effects or of disease [7]. Indeed, agent. Studies in humans have associated the development of certain cancers (i.e. in
gene mutation patterns are probably the increases in the frequency of HPRT muta- these cases, cancer can be linked causal-
only biological marker that can be char- tions (measured in circulating lympho- ly to past exposure to a defined carcino-
acteristic of a past exposure to a carcino- cytes) with exposure to environmental genic agent). These mutations, which
genic agent or mixture. Study of such genotoxic agents. However, in contrast to could in principle be used to identify expo-
mutations will increasingly assist in the observations made in rodents, in which sure to particular agents, have been
identification of etiologic agents, in risk mutation profiles often reflect the rela- termed signature mutations. They result
prediction and in cancer prevention stud- tively extreme DNA damage that induced from the formation of specific DNA
ies. Mutation spectra can be analysed them, characteristic HPRT mutation spec- adducts. For example, p53 mutations
either in normal tissues (including blood tra (i.e. the types and positions of the characteristic of the known or suspected
cells) or in tumour tissues. Analysis of base changes within the DNA sequence of etiological agent occur in lung cancer
mutations in normal tissues remains diffi- the HPRT gene) are more difficult to (attributable to benzo[a]pyrene in tobacco
cult, because the mutant cell or DNA observe in humans. smoke) and hepatocellular carcinomas
must be identified against a background The identification of oncogenes and (due to aflatoxin B1 in contaminated food)
of a very large excess of non-mutant cells tumour suppressor genes (Oncogenes and (Box: Geographic variation in mutation
or DNA, and a selection or an enrichment tumour suppressor genes, p96) has led to patterns, p102). In general, however, it is
step is required. In contrast, mutations in the characterization of gene mutations often not practical to obtain DNA from
tumour cells often favour growth and are which are more directly associated with healthy tissue to analyse for potentially
amplified due to clonal expansion of the carcinogenesis. The RAS family of onco- tumorigenic mutations, as invasive meth-
tumour cell population. genes was among the first that was rec- ods of sampling are required. Fortunately,
A few genes are suitable markers ognized as being mutated in a wide variety the protein products of the mutated genes
(reporters) of mutation induction in of human cancers. p53 is the most com- and, even the mutated DNA itself, can be
experimental animals and in humans. monly altered tumour suppressor gene in detected and measured in body fluids or
Thus the hypoxanthine-guanine phospho- human cancer, being mutated in over 50% secretions, such as blood plasma, that
have been in contact with the malignant
tissue.
DAMAGING AGENT Presumed signature mutations have also
been identified in normal tissues (non-
X-rays UV light X-rays Replication pathological but probably containing initi-
Oxygen radicals Polycyclic aromatic Anti-tumour agents errors
Alkylating agents hydrocarbons (cisplatin, mitomycin C) ated cells) from exposed individuals. For
Spontaneous reactions
example, the p53 mutation associated
with exposure to aflatoxin B1 has been
found in liver tissue and in plasma DNA
G G from healthy subjects (without cancer)
U T A
G G G C
T C T who have consumed food contaminated
T
with aflatoxins. Therefore, mutations in
cancer genes could be used, in certain
cases, as early indicators of risk before
disease diagnosis.
Uracil 6-4 Photoproduct Interstrand cross-link A-G Mismatch
Abasic site Bulky adduct Double-strand break T-C Mismatch DNA repair
8-Oxoguanine Cyclobutane pyrimidine- Insertion
Single-strand break dimer Deletion The 3 x 109 nucleotides of the DNA within
each human cell are constantly exposed
Base-excision Nucleotide-excision Recombinational Mismatch repair to an array of damaging agents of both
repair repair repair (homologous environmental origin, exemplified by sun-
or end-joining)
light and tobacco smoke, and of endoge-
REPAIR PROCESS
nous origin, including water and oxygen
Fig. 3.9 Common DNA damaging agents, examples of DNA lesions induced by these agents and the most [8] (Table 3.2). This scenario necessitates
important DNA repair mechanism responsible for the removal of these lesions. constant surveillance so that damaged

Carcinogen activation and DNA repair 91


nucleotides may be removed and
replaced before their presence in a DNA
strand at the time of replication leads to
the generation of mutations [9].
Restoration of normal DNA structure is
achieved in human cells by one of several
DNA repair enzymes that cut out the
damaged or inappropriate bases and
replace them with the normal nucleotide
sequence. This type of cellular response
is referred to as excision repair and
there are two major repair pathways
which function in this manner: base exci-
sion repair which works mainly on modi-
fications caused by endogenous agents
and nucleotide excision repair which
removes lesions caused by environmental
mutagens. UV light is probably the most
common exogenous mutagen to which
human cells are exposed and the impor-
tance of the nucleotide excision repair
pathway in protecting against UV-induced
carcinogenesis is clearly demonstrated in
the inherited disorder xeroderma pigmen-
tosum. Individuals who have this disease
lack one of the enzymes involved in
nucleotide excision repair and have a
1,000 times greater risk of developing skin
cancer following exposure to sunlight than
normal individuals. The genes in question
have been named XPA, XPB, etc. [10].
One of the great achievements of the last
two decades has been the isolation and
characterization of the genes, and their
protein products, involved in base excision
repair and nucleotide excision repair. It
has become apparent that certain pro-
teins so identified are not exclusively
involved in DNA repair but play an integral
part in other cellular processes such as Fig. 3.10 Nucleotide excision repair (NER). Two NER pathways are predominant for removal of UV light-
and carcinogen-damaged DNA. In global genome NER, the lesion is recognized by the proteins XPC and
DNA replication and recombination. hHR23B while in transcription-coupled NER of protein-coding genes, the lesion is recognized when it
stalls RNA polymerase II. Following recognition, both pathways are similar. The XPB and XPD helicases
Excision repair of the multi-subunit transcription factor TFIIH unwind DNA around the lesion (II). Single-stranded binding
The first step in both base excision repair protein RPA stabilizes the intermediate structure (III). XPG and ERCC1-XPF cleave the borders of the dam-
and nucleotide excision repair is the aged strand, generating a 24-32 base oligonucleotide containing the lesion (IV). The DNA replication
machinery then fills in the gap (V).
recognition of a modification in DNA by
enzymes that detect either specific forms
of damage or a distortion in the DNA
helix. Recognition of damage is followed Nucleotide excision repair may occur in the lesion in a reaction that uses the ATP-
by an excision step in which DNA con- the non-transcribed (non-protein-coding) dependent helicase activities of XPB and
taining the modified nucleotide is regions of DNA (Fig. 3.10, steps I to V). A XPD (two of the subunits of TFIIH) and
removed. Gap-filling DNA synthesis and distortion in DNA is recognized, probably also involves XPA and RPA (II-III). The
ligation of the free ends complete the by the XPC-hHR23B protein (I). An open XPG and ERCC1-XPF nucleases excise
repair process. bubble structure is then formed around and release a 24- to 32-residue oligonu-

92 Mechanisms of tumour development


Reactive oxygen species X-rays
Methylation, deamination (single-stranded break)

P OH

Spontaneous hydrolysis
(abasic site) PARP
DNA
glycosylase XRCC1

I
PNK
APE1

II P

III
DNA pol
PCNA

XRCC1 DNA pol /


+dNTPs
+dGTP

IV VII Fig. 3.12 In the human genome there are


numerous places where short sequences of DNA
FEN1 are repeated many times. These are called
microsatellites. In DNA from a patient with hered-
itary nonpolyposis colorectal cancer, there are
changes in the number of repeats in the
VIII microsatellites. Note the difference in the
V microsatellite pattern between normal (N) and
DNA DNA tumour tissue (T) from the same patient. This
ligase 3 ligase 1
microsatellite instability is caused by errors in
post-replicative DNA mismatch repair.

VI IX AP1) [12]. Gap-filling may proceed by


SHORT-PATCH BASE EXCISION REPAIR LONG-PATCH BASE EXCISION REPAIR replacement of a single base or by resyn-
(Main pathway) (Minor pathway) thesis of several bases in the damaged
strand (depending on the pathway
Fig. 3.11 Stages of base excision repair. Many glycosylases, each of which deals with a relatively narrow employed).
spectrum of lesions, are involved. The glycosylase compresses the DNA backbone to flip the suspect More complex and unusual forms of dam-
base out of the DNA helix. Inside the glycosylase, the damaged base is cleaved, producing an abasic age to DNA, such as double strand breaks,
site (I). APE1 endonuclease cleaves the DNA strand at the abasic site (II). In the repair of single-strand-
ed breaks, poly(ADP-ribose)polymerase (PARP) and polynucleotide kinase (PNK) may be involved. In the
clustered sites of base damage and non-
short-patch pathway, DNA polymerase fills the single nucleotide gap and the remaining nick is sealed coding lesions that block the normal repli-
by DNA ligase 3. The long-patch pathway requires the proliferating cell nuclear antigen (PCNA) and cation machinery are dealt with by alter-
polymerases , and fill the gap of 2-10 nucleotides. Flap endonuclease (FEN-1) is required to remove native mechanisms. Inherited human dis-
the flap of DNA containing the damage and the strand is sealed by DNA ligase 3. eases in which the patient shows extreme
sensitivity to ionizing radiation and altered
processing of strand breaks, such as atax-
cleotide (IV) and the gap is filled in by DNA base excision repair (Fig. 3.11, steps ia telangiectasia and Nijmegen breakage
PCNA-dependent polymerases (POL) I to VI or steps III to IX) involves the syndrome, constitute useful models to
epsilon and delta and sealed by a DNA lig- removal of a single base by cleavage of study the repair enzymes involved in these
ase, presumed to be LIG1 (V). Nucleotide the sugar-base bond by a damage-specific processes. Indeed, if elucidation of base
excision repair in regions which are tran- DNA glycosylase (e.g. hNth1 or uracil DNA excision repair and nucleotide excision
scribed (and hence code for proteins) glycosylase) and incision by an repair was the great achievement of the
requires the action of TFIIH [11]. apurinic/apyrimidinic nuclease (human late 1990s, then understanding strand

Carcinogen activation and DNA repair 93


Other repair pathways
SINGLE BASE MISPAIRS INSERTION OR DELETION LOOPS Human cells, in common with other
hMutS hMutL hMutS hMutL
eukaryotic and prokaryotic cells, can also
or hMutS perform one very specific form of damage
hMSH6 hPMS2 hMSH6 hPMS2 reversal, the conversion of the methylated
adduct, O6-methylguanine, in DNA back to
hMSH2 hMLH1 hMLH1
hMSH2 the normal base (Fig. 3.14). O6-Methylgua-
nine is a miscoding lesion: both RNA and
DNA polymerases read it incorrectly
CA when they transcribe or replicate a DNA
C TAG G T TA C ACACACA template containing it. As this modified
G ATC C G AT GTGTGTG T
base can pair with both the base cytosine
(its correct partner) and the base thymine
hMSH6
(an incorrect partner), its presence in DNA
hPMS2 hPMS2 can give rise to transition mutations by
hMS H2
mispairing of relevant bases. A specific
hMSH2
hMLH1 hMLH1 protein, O6-alkylguanine-DNA-alkyltrans-
ferase, catalyses transfer of the methyl
group from the guanine base to a cysteine
C TAG G C TA C ACACACA
G ATC C G AT G TG TG TG T amino acid residue located at the active
site of the protein [13]. This error-free
Fig. 3.13 Mismatch repair pathways: after DNA synthesis, base pairing mistakes that have escaped the process restores the DNA to its original
editing function of DNA polymerase are recognized by mismatch repair proteins.
state but results in the inactivation of the
repair protein. Consequently, repair can be
saturated when cells are exposed to high
break repair will probably be the great reduced to try to avoid adverse reactions. doses of alkylating agents and synthesis of
achievement of the next decade. This will A better understanding of the possible the transferase protein is required before
have important consequences. Certain causes of this radiosensitivity, including repair can continue.
cancers are often treated with radiother- characterization of the enzymes involved Mismatched bases in DNA arising from
apy (Radiotherapy, p277) and a small per- in the repair of DNA damage produced by errors in DNA replication, for instance gua-
centage of patients show considerable ionizing radiation, may lead to better tai- nine paired with thymine rather than cyto-
sensitivity to their treatment, with the loring of radiotherapy doses to individual sine, are repaired by several pathways
result that treatment schedules are patients. involving either specific glycosylases,

Agent Mutation hotspot Type of mutation Tumours associated


(> = changes to)

Benzo[a]pyrene Codons 157, 158, 248, 273 G>T transversions Lung, larynx
(tobacco smoke)

4-Aminobiphenyl Codons 280, 285 G>C transversions Bladder


(aromatic dyes, tobacco smoke) G>A transitions

Aflatoxin B1 Codon 249 AGG>AGT Hepatocellular carcinoma


(arginine > serine)

Ultraviolet (UV) Codons 177-179, 278 C>T transitions Skin cancer


CC>TT transitions (not melanoma)

Vinyl chloride Several codons A>T transversions Angiosarcoma of the liver

Endogenous mechanism Codons 175, 248, 273, 282 C>T transitions Colon, stomach
(enhanced by nitric oxide) at CpG dinucleotides Brain cancers

Table 3.2 Spectra of p53 mutations caused by environmental carcinogens or endogenous mechanisms.

94 Mechanisms of tumour development


which remove the mismatched bases, or scans the DNA for a lesion or structure
long-patch mismatch repair involving that is not a normal constituent of the
homologues of the bacterial genes MUTS DNA. Defects in at least four of the genes
and MUTL (Fig. 3.13). Insertion or deletion whose products are involved in mismatch
loops at microsatellite sequences can be repair, namely hMSH2, hMLH1, hPMS1
recognized by hMutS (a heterodimer of and hPMS2, have been associated with Me
MGMT
hMSH2 and hMSH6) or hMutS (a het- hereditary nonpolyposis colorectal cancer. Cys
erodimer of hMSH2 and hMSH3). This is one of the most common genetic
Subsequent recruitment of hMutL (a het- diseases and affects as many as 1 in 200 MGMT
Cys
erodimer of hMLH1 and hPMS2) to the individuals and may account for 4-13% of Me
altered DNA targets the area for repair, all colorectal cancers (Colorectal cancer,
which requires excision, resynthesis, and p198). Affected individuals also develop Fig. 3.14 The repair of O6-methylguanine by
ligation. Single nucleotide mispairing tumours of the endometrium, ovary and O6-alkylguanine-DNA-alkyltransferase.
events require hMutS function for recog- other organs. The DNA of hereditary non-
nition. One important requirement of such polyposis colorectal cancer tumours is
repair processes is that they are able to characterized by instabilities in simple directly from alterations in the proteins
distinguish the correct base from the mono-, di- and trinucleotide repeats which involved in mismatch repair [14]. Generally
incorrect one in the mispair. Since both are common in the human genome (Fig. speaking, genomic instability is considered
bases are normal constituents of DNA, this 3.12). This instability is also seen in certain an indicator of, and fundamental to the
cannot be achieved by an enzyme that sporadic colorectal tumour cells and arises nature of, malignant cell growth.

REFERENCES WEBSITES
1. Miller EC, Miller JA (1979) Milestones in chemical car- 8. Friedberg EC, Walker GC, Siede W, eds (1995) DNA A comprehensive listing of human DNA repair genes:
cinogenesis. Semin Oncol, 6: 445-460. Repair and Mutagenesis, Washington DC, ASM Press. http://www.sciencemag.org/cgi/content/abstract/291/
2. Miller JA, Miller EC (1977) Ultimate chemical carcino- 9. Lindahl T (2000) Suppression of spontaneous mutage- 5507/1284
gens as reactive mutagenic electrophiles. In: Hiatt HH, nesis in human cells by DNA base excision-repair. Mutat DNA Repair Interest Group (NCI):
Watson, JD, Winsten, JA eds, Origins of Human Cancer Res, 462: 129-135. http://www.nih.gov:80/sigs/dna-rep/
(Book B), Cold Spring Harbor, Cold Spring Harbor 10. de Boer J, Hoeijmakers JH (2000) Nucleotide excision
Laboratory, 605-627. repair and human syndromes. Carcinogenesis, 21: 453-
3. Guengerich FP (2000) Metabolism of chemical car- 460.
cinogens. Carcinogenesis, 21: 345-351. 11. Benhamou S, Sarasin A (2000) Variability in
4. Hemminki K, Dipple A, Shuker DEG, Kadlubar FF, nucleotide excision repair and cancer risk: a review. Mutat
Segerbck D, Bartsch H, eds (1994) DNA Adducts. Res, 462: 149-158.
Identification and Biological Significance (IARC Scientific 12. Cadet J, Bourdat AG, D'Ham C, Duarte V, Gasparutto
Publications No. 125), Lyon, IARCPress. D, Romieu A, Ravanat JL (2000) Oxidative base damage to
5. Toniolo P, Boffetta P, Shuker DEG, Rothman N, Hulka B, DNA: specificity of base excision repair enzymes. Mutat
Pearce N, eds (1997) Application of Biomarkers in Cancer Res, 462: 121-128.
Epidemiology (IARC Scientific Publications No. 142), Lyon, 13. Pegg AE (2000) Repair of O6-alkylguanine by alkyl-
IARCPress. transferases. Mutat Res, 462: 83-100.
6. Vineis P, Malats N, Lang M, d'Errico A, Caporaso N, 14. Pedroni M, Sala E, Scarselli A, Borghi F , Menigatti M,
Cuzick J, Boffetta P, eds (1999) Metabolic Polymorphisms Benatti P, Percesepe A, Rossi G, Foroni M, Losi L, Di
and Susceptibility to Cancer (IARC Scientific Publications Gregorio C, De Pol A, Nascimbeni R, Di Betta E, Salerni B,
No. 148), Lyon, IARCPress. de Leon MP, Roncucci L (2001) Microsatellite instability
7. McGregor DB, Rice JM, Venitt S, eds (1999) The Use of and mismatch-repair protein expression in hereditary and
Short- and Medium-Term Tests for Carcinogens and Data sporadic colorectal carcinogenesis. Cancer Res, 61: 896-
on Genetic Effects in Carcinogenic Hazard Evaluation 899.
(IARC Scientific Publications No. 146), Lyon, IARCPress.

Carcinogen activation and DNA repair 95


ONCOGENES AND TUMOUR SUPPRESSOR GENES

mechanisms, including point mutations control the speed or timing of cell division
SUMMARY that constitutively activate an enzyme, but rather its accuracy. Caretaker genes
deletions that remove negative regulatory are usually involved in DNA repair and in
> Human cells become malignant through regions from proteins, or increased the control of genomic stability. Their
the activation of oncogenes and expression resulting from promoter dereg- inactivation does not enhance cell prolif-
inactivation of tumour suppressor
ulation or from multiplication of the num- eration per se but primes the cell for rapid
genes. The pattern of genes involved
varies markedly at different organ sites. ber of copies of the gene (a phenomenon acquisition of further genetic changes [4].
called amplification [3]). Activation of an The combined activation of oncogenes
> Oncogenes stimulate cell proliferation oncogene is a dominant mechanism, since and inactivation of tumour suppressor
and may be overexpressed by gene alteration of a single allele is sufficient to genes drive the progression of cancer. The
amplification (e.g. MYC). In addition, confer a gain of function for cancer onset most evident biological consequences of
oncogenes may be activated by muta- or progression. The non-activated coun- these alterations are autonomous cell pro-
tions (e.g. the RAS gene family). terpart of an oncogene is sometimes liferation, increased ability to acquire
called a proto-oncogene. A proto-onco- genetic alterations due to deregulated
> Tumour suppressor genes are typically gene is in fact a normal gene in all DNA repair, ability to grow in adverse con-
inactivated by gene mutations in one
respects, often with important functions ditions due to decreased apoptosis,
allele (gene copy), followed by loss of
the intact allele during cell replication in the control of the signalling of cell pro- (Apoptosis, p113) capacity to invade tis-
(two-hit mechanism). This leads to loss liferation, differentiation, motility or sur- sues locally and to form distant metas-
of expression and abolition of the sup- vival. tases, and ability to activate the formation
pressor function, which is particularly A tumour suppressor gene is a gene of new blood vessels (a process called
important in cell cycle control. whose alteration during carcinogenesis angiogenesis). Together, these five biolog-
results in the loss of a functional property ical phenomena may be caricatured as
> Mutational inactivation of suppressor essential for the maintenance of normal pieces of the cancer jigsaw [5] (Fig.
genes in germ cells is the underlying cell proliferation. Loss of function of a 3.15). None alone is sufficient in itself, but
cause of most inherited tumour tumour suppressor gene is typically a cancer arises when they interact together
syndromes. The same type of mutation
recessive mechanism. Indeed, in many into a chain of coordinated events that
may arise through mutations occurring
during an individuals lifetime. instances both copies of the gene need to profoundly modifies the normal cellular
be inactivated in order to switch off the pattern of growth and development.
corresponding function. Inactivation of
tumour suppressor genes proceeds by
loss of alleles (most often through the loss
Definitions of entire chromosomal sections encom-
The multi-step nature of carcinogenesis passing several dozen genes), small dele-
has long been recognized (Multistage car- tions or insertions that scramble the read-
Genetic Invasiveness
cinogenesis, p84). Over the past 20 years, ing frame of the gene, transcriptional instability
experimental studies in animals and silencing by alteration of the promoter
molecular pathological studies have con- region, or point mutations that change the
verged to establish the notion that each nature of residues that are crucial for the
step in malignant transformation is deter- activity of the corresponding protein. Autonomous
mined by a limited number of alterations Recently, it has emerged that tumour sup- growth
in a small subset of the several thousands pressor genes can be conveniently sub-
of cellular genes [1]. The terms onco- classified into two major groups. The
gene and tumour suppressor gene are genes of the first group are nicknamed
commonly used to identify the sets of gatekeepers. Their products control the
genes involved in such sequences of gates on the pathways of cell proliferation. Unlimited
Angiogenesis
events [2]. Both groups of genes are Typically, gatekeeper genes are negative replicative potential
extremely diverse in terms of nature and regulators of the cell cycle, acting as
function. An oncogene is a gene whose brakes to control cell division. The genes
function is activated in cancer. This can be of the second group are called caretak- Fig. 3.15 The cancer jigsaw: multiple functions
achieved by a number of simple molecular ers, as their primary function is not to must be altered for tumorigenesis to occur.

96 Mechanisms of tumour development


Common human oncogenes
Many common proto-oncogenes encode
components of the molecular cascades
that regulate the cellular response to
mitogenic signals [6]. They include growth
factors (e.g. TGFA), growth factor recep-
tors (e.g. the receptors for epidermal
growth factor, EGF and its close homo-
logue, ERBB2), receptor-coupled signal
transduction molecules (in particular, sev- A B
eral small guanosine triphosphate (GTP)- Fig. 3.16 Analysis of the status of the ERBB2 oncogene by fluorescent in situ hybridization (FISH) with a
binding proteins located on the inner face rhodamine-labelled ERBB2 probe (pink). In breast tumour cells without amplification of the gene, each
of the cell membrane, such as the various nucleus possesses two copies of ERBB2 (A). In tumour cells with high-level amplification of the gene,
members of the RAS family), kinases numerous signals are evident in each nucleus (B).
(SRC, ABL, RAF1), regulatory subunits of
cell cycle kinases (CCND1 and CCNA), gene is located within a region of the with activated tyrosine kinases and act as
phosphatases (CDC25B), anti-apoptotic genome which is amplified in about 27% of amplifiers to increase the strength of the
molecules (BCL2) and transcription fac- advanced breast cancers, leading to a signal generated by the activation of cell-
tors (MYC, MYB, FOS, JUN). The cumber- spectacular increase in the density of the surface receptors [8]. In their active form,
some nomenclature of these genes (Box: molecule at the cell surface. ERBB2 ras proteins bind guanosine triphosphate
Naming genes and proteins, p101) owes encodes a transmembrane protein with (GTP) and catalyse its hydrolysis into
much to the way they were discovered and the structure of a cell-surface receptor, the guanosine diphosphate (GDP) returning to
identified. The SRC gene, for example, was intracellular portion of which carries a their inactive form. Oncogenic forms of
the first oncogene identified, in 1976, as a tyrosine kinase activity. Overexpression of activated RAS genes often carry missense
modified version of a cellular gene incor- ERBB2 leads to constitutive activation of mutations at a limited number of codons
porated in the genome of a highly trans- the growth-promoting tyrosine phosphory- within the GTP-binding site of the enzyme,
formant chicken retrovirus, the Rous sar- lation signal. The elucidation of this mech- making it unable to hydrolyse GTP and thus
coma virus. The MYC gene was also origi- anism has led to the development of neu- trapping it in the active form. Activation of
nally identified in the genome of an avian tralizing antibodies and specific chemical RAS genes thus induces the cell to behave
retrovirus inducing promyelocytic inhibitors of tyrosine kinase activity as as if the upstream, Ras-coupled receptors
leukaemia. The RAS genes were first iden- therapeutic approaches to the blocking of were being constantly stimulated.
tified as activated genes capable of induc- ERBB2 action.
ing the formation of rat sarcomas, and MYC
various members of the family were found RAS The MYC oncogene may be seen as a pro-
in different murine retroviruses, such as The RAS genes are located one step down- totype of the family of molecules which lies
the Harvey sarcoma virus (HRAS) and the stream of ERBB2 in growth signalling cas- at the receiving end of the signal transduc-
Kirsten sarcoma virus (KRAS). cades. The protein products of the RAS tion cascades. MYC encodes a transcrip-
The most commonly activated oncogenes genes are small proteins anchored at the tion factor which is rapidly activated after
in human cancers are ERBB2 (in breast cytoplasmic side of the plasma membrane growth stimulation and which is required
and ovarian cancers), members of the RAS by a lipidic moiety. They indirectly interact for the cell to enter into cycle [9].
family (in particular KRAS in lung, colorec-
tal and pancreatic cancers, and MYC (in a
large variety of tumours such as cancers of
the breast and oesophagus and in some
forms of acute and chronic leukaemia).
These three examples give an excellent
illustration of the diversity of the mecha-
nisms of oncogene activation and of their
consequences for cell growth and division.

ERBB2 A B
In the case of ERBB2, oncogenic activation Fig. 3.17 In cell cultures, activation of a single oncogene may result in a changed morphology from
is almost always the result of amplification normal (A) to transformed (B) and this often corresponds to a change in growth properties. Malignant
of the normal gene [7] (Fig. 3.16). This transformation appears to require the co-operation of at least three genes.

Oncogenes and tumour suppressor genes 97


Normal Myc transactivates a number of other cel- Tumour suppressor genes: history of a
chromosome 13 lular genes and has a wide spectrum of concept
Parent Normal
heterozygous Parent molecular effects (a phenomenon that Whereas the study of retroviruses and
for RB1 RB1 may explain why Myc is activated in many gene transfection experiments were the
different types of cancer cells). keys to the discovery of oncogenes,
Chromosome 13
with deletion
Activation of Myc often proceeds through tumour suppressor genes were identified
of RB1 amplification of the region containing the through the study of large DNA viruses
Child
heterozygous gene on chromosome 8, but Myc is also and the analysis of familial tumour syn-
for RB1 commonly activated by chromosomal dromes.
translocation in some forms of B-cell
Somatic mutation with high frequency
in retinal cell with loss of leukaemia (Leukaemia, p242). Retinoblastoma
normal chromosome In 1971, Knudsen proposed the now popu-
BCL2 lar two hits hypothesis to explain the
The BCL2 gene (activated in B cell lym- inheritance of retinoblastoma, a rare
phomas) exemplifies another kind of childhood tumour type [11,12] (Genetic
oncogene. Initially identified as a gene susceptibility, p71). He postulated that, in
located within a chromosomal breakpoint a familial setting, individuals may inherit
Proliferation in some forms of leukaemia, BCL2 was only one normal copy of the gene (local-
found to encode a protein capable of ized by linkage studies to chromosome
extending the life span of a cell by pre- 13q14), the other being either lost, par-
venting the onset of programmed cell tially deleted or otherwise inactivated.
death, or apoptosis [10] ( Apoptosis , Consequently, these individuals would just
p113). Biochemical studies have revealed need one additional mutagenic step to
Nonmalignant
Proliferating retinoblastoma cells cells that BCL2 encodes a regulator of the per- switch off the remaining copy of the gene,
meability of the mitochondrial mem- thus totally losing the corresponding func-
brane. Mitochondrial damage and cyto- tion (Fig. 3.18). The very same type of can-
Fig. 3.18 The retinoblastoma gene is a paradigm
plasmic leakage of mitochondrial compo- cer may also occur in a sporadic manner,
for tumour suppressor genes: if a child inherits a
mutation or deletion of one copy (allele) of the nents is one of the important signals that but in this case it would require two con-
retinoblastoma gene, the remaining normal copy lead a cell to apoptosis. By helping to secutive hits (mutagenic events) to inac-
tends to be lost at a high frequency in cells of the keep the mitochondrial permeability tivate the two copies of the gene in the
retina, resulting in loss of function and in the for- pores closed, Bcl-2 protein prevents this same cell. This theory paved the way for
mation of tumours. The diagram shows loss of the
whole normal chromosome but the normal allele leakage and thus allows the survival of the modern concept of recessive tumour
can also be lost by mutation, deletion, gene con- cells that would otherwise have been suppressor genes. In 1988, the gene
version or mitotic recombination. eliminated by a physiological process. responsible for familial retinoblastoma
was identified [13]. The RB1 gene encodes
a protein that binds and inactivates tran-
Cytotoxic scription factors that are essential for the
drugs progression of the cell cycle, thus fulfilling
UV Cytokines the functions of a molecular brake on
X-rays Ribonucleotide
-rays depletion cell division.
Hypoxia Microtubule
depletion Large DNA viruses
Redox changes Growth factor In parallel with events previously outlined,
depletion it became evident that many DNA viruses
Temperature ? Senescence associated with cancer encode complex
viral proteins that are capable of seques-
Activation or accumulation of p53 protein
tering and inactivating cellular proteins
[14]. This is the case of a tumorigenic
Induction of p53 target genes
Binding to p53-interacting proteins simian virus, SV40, of several adenoma
and polyoma viruses and of oncogenic
forms of human papillomaviruses. In the
case of SV40, the virus encodes a large
protein (called LT for Large Tumour anti-
Fig. 3.19 Many types of biological stress lead to a p53-mediated response. gen) which binds two cellular proteins, the

98 Mechanisms of tumour development


product of the RB1 gene (pRb) and an molecules and transcription factors that Others
ubiquitous protein that was conservatively regulate cell proliferation. Loss of APC In the case of hereditary Wilms tumours, a
called p53. In the case of oncogenic function sets these transcription factors rare type of kidney cancer, the gene identi-
human papillomaviruses, the viruses free, an event that favours not only the fied encodes a protein essential for the cor-
encode two distinct proteins, E7 (which formation of polyps but also their trans- rect differentiation of the nephron. This very
neutralizes pRb) and E6 (which neutralizes formation into adenomas and carcino- specific role may explain why the hereditary
p53). Thus it was suggested that pRb and mas. loss of this gene does not seem to be asso-
p53 might have similar, complementary ciated with cancers at any other site.
functions, operating jointly in the control Breast cancer
of cell division. Two genes have been identified as This short overview gives only a few exam-
The missing link in this conceptual edi- involved in familial breast cancer risk, ples of the diversity of tumour suppressor
fice was the discovery of alterations in the BRCA1 and BRCA2 [18]. These genes genes, and there is little doubt that many
gene encoding p53. This was achieved in encode large proteins with complex still remain to be identified. Given the
1989, when it emerged that the p53 gene functions in many aspects of cell regu- breadth of the concept of tumour suppres-
was often mutated and/or deleted in lation, such as cell cycle control and sors, many genes encoding components of
many forms of cancers [15]. In 1991, DNA repair. However, how their inacti- stress response pathways have the poten-
inherited loss of p53 was found to be vation contributes to the onset or tial to behave in this fashion (as their alter-
associated with a rare familial syndrome development of breast cancer is still ation may prevent cells from mounting an
of multiple cancers, the Li-Fraumeni syn- largely unknown. adequate response to genotoxic, potentially
drome, in which afflicted family members
suffer vastly increased incidence of many
tumour types [16]. Today, about 215 fami-
lies worldwide affected by this syndrome
have been described and the p53 muta-
tions they exhibit are compiled in a data-
base maintained at IARC.

Tumour suppressor genes and familial


cancer syndromes
Most familial cancer syndromes are
inherited as a recessive trait, and cor- A B
respond to the constitutive inactivation Fig. 3.20 Accumulation of p53 in human epidermis after exposure to sunlight. Unexposed skin shows no
immunostaining against p53 protein (A). Exposed skin (B) shows a dense dark nuclear coloration of epi-
of an important tumour suppressor dermal cells due to positive immunostaining for p53 protein.
gene, as described above in the case of
familial retinoblastoma. Over the past
15 years, many loci containing tumour
suppressor genes have been identified Transcriptional activation
by linkage studies in cancer-prone fam- Transcriptional repression
ilies. Protein interactions

Colorectal cancer
In colorectal cancers, two different famil- Bcl-2
ial cancer syndromes have been found to 14 - 3 -3 p21 p53R2 Bax Unknown
be associated with the constitutive alter- RPA IGF/BP3 proteins?
TFIIH Killer/DR5
ation of two distinct sets of tumour sup- PAG608
pressor genes (Colorectal cancer, p198). PIG3
Patients with familial adenomatous poly- cdc25 Cdk PCNA
posis, a disease that predisposes to the G2 G1 G1/S
Replication/
early occurrence of colon cancer, often Transcription/
Cell cycle arrest Apoptosis
carry alterations in one copy of the ade- Repair
nomatous polyposis coli (APC) gene [17].
This gene plays a central role in a sig-
nalling cascade that couples cell-surface
receptors, calcium-dependent adhesion Fig. 3.21 Multiple response pathways are triggered by the accumulation of p53 in the cell nucleus.

Oncogenes and tumour suppressor genes 99


expression of several dozen genes
involved in cell cycle control, in the induc-
tion of apoptosis, in DNA repair and in dif-
ferentiation control. Together these genes
exert complex, anti-proliferative effects
(Fig. 3.21). Essentially, when cells are sub-
jected to tolerable levels of DNA-damag-
ing agents, activation of p53 will result in
cell cycle arrest, temporarily removing the
cells from the proliferative pool or mediat-
ing differentiation. However, when faced
with highly damaging levels of genotoxic
stress, p53 will induce apoptosis, a pro-
grammed form of suicide that eliminates
cells with potentially oncogenic alter-
ations. This complex role in the protection
of the cell from DNA damage has resulted
in p53 being described as the guardian of
the genome [20]. Loss of this function by
mutation, as often occurs during carcino-
Fig. 3.22 Molecular modelling of part of the p53 protein (DNA-binding domain), showing its interaction genesis, will allow cells with damaged
with DNA. The amino acids labelled (arginine 175, 248, 273) are important for maintaining biological
activity and are among the hotspots for mutations in cancer. The zinc atom is required for stabilizing DNA to remain in the proliferative popula-
the complex three-dimensional structure of the p53 oligomer. tion, a situation that is essential for the
expansion of a clone of cancer cells.
The p53 gene differs from most other
CDKN2A/ tumour suppressors in its mode of inacti-
INK4A vation in human cancers. Whereas most
Exon 1 Exon 1 Exon 2 Exon 3 gene
tumour suppressors are altered by loss of
alleles or inactivating deletions or inser-
tions, p53 is commonly the target of
p16INK4A point mutations within the portion of the
Inhibitor of
cyclin D/CDK4
gene that encodes the DNA-binding
complexes domain of the protein (Fig. 3.22). These
mutations prevent the correct folding of
p14ARF this protein domain, and therefore dis-
Inhibitor of p53-
Mdm2 complex rupt the interactions of p53 with its spe-
formation
cific DNA targets. However, the mutant
Fig. 3.23 Generally, a single segment of DNA codes for a single protein. However, the p16 and p14ARF proteins are often extremely stable and
proteins are both encoded by a single region of DNA. P = promoter. therefore accumulate to high levels with-
in the nucleus of cancer cells. This accu-
oncogenic forms of stress). The genes are also mutated at variable rates in many mulated protein can often be detected by
responsible for complex inherited diseases forms of sporadic cancer. However, two of immunohistochemistry in primary tumours
such as ataxia telangiectasia or xeroderma them, p53 and CDKN2A, are very com- as well as in distant metastases. Although
pigmentosum (Carcinogen activation and monly altered in almost every kind of not all mutations induce accumulation of
DNA repair, p89) belong to this category human cancer. the protein, p53 accumulation provides a
[19]. Alteration of such genes results in convenient tool for pathologists to assess
many defects, including hypersensitivity to p53, the guardian of the genome the possibility of a p53 dysfunction in
radiation and therefore to the development The p53 gene encodes a phosphoprotein cancer specimens [21].
of cancers such as skin tumours. of molecular weight 53,000 daltons, Mutation is not the only way to alter p53
which accumulates in the nucleus in protein in cancer. In cervical cancers,
Tumour suppressor genes and spo- response to various forms of stress, in p53 gene mutations are infrequent, but
radic cancers particular, DNA damage (Fig. 3.20). In this the protein is inactivated by binding of
Many of the tumour suppressor genes context, p53 acts as a transcriptional reg- the viral protein E6 which is produced by
associated with familial cancer syndromes ulator, increasing or decreasing the human papillomavirus. This protein cre-

100 Mechanisms of tumour development


ates a molecular bridge between p53 and restore the function of mutant p53 have reading frame (often called by the same
the protein degradation machinery, shown promising results in experimental name as its mouse homologue, p19ARF), is
resulting in the rapid degradation and systems. As knowledge of the p53 path- synthesized from a different portion of the
effective elimination of p53 protein. This way improves, it is anticipated that this CDKN2A locus but shares one exon (exon
interaction plays an important role in cer- central molecular event in human cancer 2) with p16. However, although the DNA
vical cancer (Cancers of the female will provide a basis for developing new sequence encoding the two products is
reproductive tract, p215). In normal cells, forms of therapy. identical, p16 and p14ARF use different
the degradation of p53 is regulated by reading frames of exon 2, such that their
the Mdm2 protein. Mdm2 (murine dou- CDKN2A: one locus, two genes amino acid sequences are completely dif-
ble minute gene 2) was originally identi- CDKN2, or cyclin dependent kinase ferent. p14ARF is a protein that controls
fied in the mouse as the product of a inhibitor 2, is known under several names, Mdm2, which in turn regulates p53 protein
gene amplified in aberrant chromosome including INK4A (inhibitor of kinase 4A) stability. Activation of p14ARF blocks Mdm2
fragments called double minute chromo- and MTS1 (multiple tumour suppressor 1). and therefore results in p53 protein accu-
somes. Amplification of MDM2 is com- The CDKN2A locus is located at the mulation and activation. Thus the CDKN2A
mon in osteosarcomas and is sometimes extremity of the short arm of chromosome locus behaves as two unrelated but inter-
detected in other cancers, such as carci- 9, the letter A serving to distinguish it locked genes. The first gene, encoding
nomas or brain tumours. MDM2 thus from the CDKN2B gene, which is located p16, directly controls cell cycle progres-
behaves as an oncogene, since its activa- just 20 kilobases away. sion and senescence. The second, encod-
tion by amplification causes the inactiva- This gene is unique in that it contains two ing p14ARF, controls p53 and all its down-
tion of a tumour suppressor gene [22]. distinct reading frames, with two different stream anti-proliferative functions.
The p53 gene (and its product) is one of promoters, the same DNA being used to The CDKN2A locus is often altered by loss
the most studied genes in human cancer. synthesize two proteins that do not have a of alleles (which removes both p16 and
In the 20 years since its discovery in single amino acid sequence in common p14ARF), by mutation (most frequently in
1979, more than 15,000 publications [23] (Fig. 3.23). The first reading frame to exon 2, common to both gene products),
have addressed its structure, function be discovered encodes p16, an inhibitor of and by hypermethylation. Increased
and alteration in cancers. There have cyclin-dependent kinases 4 and 6, which methylation of specific regions of the DNA
been many attempts to exploit this associates with cyclin D1 in G1 phase of within the promoters and some of the cod-
knowledge in the development of new the cell cycle (The cell cycle, p104). The ing regions prevents adequate transcrip-
therapies based on the control of p53 p16 protein is thus an archetypal cell cycle tion and decreases the levels of protein
activity in cancer cells. Experimental brake, its loss leading to increased cell synthesized. Loss of expression due to
gene therapy has shown that it may be proliferation and, more specifically, to hypermethylation may be the most fre-
possible to restore p53 function in cells escape from replicative senescence and quent way of altering the CDKN2A locus in
that have lost the gene. More recently, extended cellular life span. The other read- many forms of cancers, particularly carci-
drugs designed to specifically target and ing frame, named p14ARF for alternative nomas.

NAMING GENES AND ative of the successful investigator. inhibitor WAF1 is also known as
PROTEINS Identification of a novel gene is often fol- CDKN1A, CAP20, MDA-6, PIC-1 and SDI-
lowed by discovery of structurally related 1. In scientific writing, all such names
Conventionally, a gene (that is, a specif- or homologous genes (and correspon- are given in the first instance, after
ic segment of DNA) is identified by a sin- ding proteins) and these may be given which a single name is used consistent-
gle name, in upper case and italicized names closely related to the first member ly in any one document. The latest esti-
(e.g. the oncogene RAS) that is indica- of the family identified. Such an mates from the Human Genome Project
tive of the character or function of the approach to nomenclature may be inade- suggest that there may be about 30,000
protein encoded, which is designated by quate, for example, in those instances in human genes.
the same name, in lower case (ras in the which a single DNA segment encodes Conventions for the naming of genes
case of the present example). Proteins multiple proteins through alternative and proteins are subject to international
are also named by reference to their splicing of messenger RNA. Multiple agreement and are continuously subject
molecular weight, with the correspon- names for the same gene or protein may to review (HUGO Gene Nomenclature
ding gene in superscript (e.g. p21WAF1 ). arise because of independent discovery Committee, http://www.gene.ucl.ac.uk/
The names of genes are often based on (and hence, naming) by different investi- nomenclature/).
acronyms, and are generally the prerog- gators. Thus, the cyclin-dependent kinase

Oncogenes and tumour suppressor genes 101


GEOGRAPHIC VARIATION IN
MUTATION PATTERNS

Mutations in cancer genes are the direct


consequence of attack on DNA by exoge-
nous or endogenous agents or of errors in
DNA repair systems. By analysing the
type and the sequence context of such
mutations, it is possible to form hypothe-
ses regarding the nature of the mutagenic
mechanism involved. The most interest-
ing genes in this respect are those altered
by missense point mutations, such as
members of the RAS family, CDKN2A/
INK4A, and, in particular, the p53 gene.

The p53 gene is the most frequently


mutated gene in human cancer, with over
16,000 mutations reported and compiled Fig. 3.24 Geographic variations in the prevalence of p53 gene mutations in breast cancers.
in a database maintained at IARC
(http://www.iarc.fr/p53). The diversity of
these mutations allows the identification class of mycotoxins which contaminates ences have been observed between geo-
of patterns which vary depending on the traditional diets (groundnuts) (Food con- graphical areas, which may provide infor-
tumour type, the geographic origin and taminants, p43). Experiments in animals mation on the nature of risk factors
the risk factors involved. These are often and in cell culture have shown that aflatox- involved.
specific for particular agents that have ins can directly induce the mutation at
caused these mutations. Thus p53 gene codon 249. This particular mutation is not In many other cancers, mutation patterns
mutations in cancers may be seen as fin- found in liver cancers in areas of the world, also vary from one region of the world to
gerprints left by carcinogens in the such as the USA, where exposure to afla- another. This variability may give clues
human genome, which may help to identi- toxins is low. about the genetic heterogeneity of popu-
fy the particular carcinogen involved. lations, as well as about the diversity of
Specific mutations have also been agents involved in causing cancers. For
A typical example of such a fingerprint observed in lung cancers from smokers example, in oesophageal cancers, muta-
is the mutation at codon 249 observed in (due to tobacco carcinogens). In skin can- tion types widely differ between high-inci-
liver cancers of patients from sub- cers, the mutations bear typical chemical dence and low-incidence regions, sug-
Saharan Africa and Eastern Asia. In these signatures of the damage inflicted to DNA gesting that specific mutagens are at
regions, liver cancer is a consequence of by exposure to solar ultraviolet radiation. In work in causing the excess incidence
chronic infection by hepatitis viruses and other instances, exemplified by patterns of seen in some parts of the world, such as
of dietary poisoning with aflatoxins, a mutation in breast cancer, marked differ- Northern Iran and Central China.

Lesions in the p16INK4A-cyclin D, CDK4-pRb cancer types have been characterized. cancer process. Moreover, many biolog-
and p14ARF-Mdm2-p53 pathways occur so Most of these have a powerful impact ical alterations leading to cancer may
frequently in cancer, regardless of patient on tumour growth. However, it is very not be detectable at the DNA level.
age or tumour type, that they appear to be likely that many critical genes with less Cancer-causing changes may result
fundamental to malignancy [24]. penetrant phenotypes remain to be from modification of RNA levels or pro-
identified. In particular, the genes cessing, and of protein structure and
Prospects for the molecular analysis of involved in stress reponses, in the con- function through a variety of epigenetic
cancer trol of oxygen metabolism and in the phenomena. The systematic profiling of
More than 200 genes that are altered at detoxification of xenobiotics are all can- gene expression in cancer cells will
variable proportions in different human didates for a role as cofactors in the probably reveal a whole new set of

102 Mechanisms of tumour development


potential cancer genes altered through tory of protein structure, dynamics and provide a basis for the development of
less conspicuous mechanisms. Apart interactions is still largely unexplored. new means for treatment and diagnosis.
from expression studies, the whole terri- An understanding of such processes will

REFERENCES WEBSITES
1. Fearon ER, Vogelstein B (1990) A genetic model for colo- share the ability to relieve the cell's requirement for cyclin American Tissue Type Collection:
rectal tumorigenesis. Cell, 61: 759-767. D1 function in G1. J Cell Biol, 125: 625-638. http://www.atcc.org/
2. Weinberg RA (1995) The molecular basis of oncogenes 15. Baker SJ, Fearon ER, Nigro JM, Hamilton SR, Centers for Disease Control and Prevention, Atlanta:
and tumor suppressor genes. Ann N Y Acad Sci, 758: 331- Preisinger AC, Jessup JM, vanTuinen P, Ledbetter DH, http://www.cdc.gov/
338. Barker DF, Nakamura Y (1989) Chromosome 17 deletions Cancer Genome Anatomy Project:
3. Savelyeva L, Schwab M (2001) Amplification of onco- and p53 gene mutations in colorectal carcinomas. http://www.ncbi.nlm.nih.gov/ncicgap/
genes revisited: from expression profiling to clinical appli- Science, 244: 217-221.
European Bioinformatics Institute:
cation. Cancer Lett, 167: 115-123. 16. Birch JM (1994) Li-Fraumeni syndrome. Eur J Cancer, http://www.ebi.ac.uk/
4. Kinzler KW, Vogelstein B (1997) Cancer-susceptibility 30A: 1935-1941.
HotMolecBase (Weizmann Institute, Israel):
genes. Gatekeepers and caretakers. Nature, 386: 761, 763. 17. Bresalier RS (1997) The gatekeeper has many keys: http://bioinformatics.weizmann.ac.il/hotmolecbase/
5. Hanahan D, Weinberg RA (2000) The hallmarks of can- dissecting the function of the APC gene. Gastroenterology,
113: 2009-2010. IARC p53 database:
cer. Cell, 100: 57-70. http://www.iarc.fr/p53/
6. Hunter T (1991) Cooperation between oncogenes. Cell, 18. Zheng L, Li S, Boyer TG, Lee WH (2000) Lessons
learned from BRCA1 and BRCA2. Oncogene, 19: 6159- Kyoto encyclopedia of genes and genomes:
64: 249-270. http://www.genome.ad.jp/kegg/kegg.html
6175.
7. Hung MC, Lau YK (1999) Basic science of HER-2/neu: OMIM (Online Mendelian Inheritance in Man):
a review. Semin Oncol, 26: 51-59. 19. Shiloh Y, Rotman G (1996) Ataxia-telangiectasia and
the ATM gene: linking neurodegeneration, immunodeficien- http://www3.ncbi.nlm.nih.gov/omim/
8. Wiesmuller L, Wittinghofer F (1994) Signal transduction cy, and cancer to cell cycle checkpoints. J Clin Immunol, Protein Data Bank (a protein structure database):
pathways involving Ras. Mini review. Cell Signal, 6: 247- 16: 254-260. http://www.rcsb.org/pdb/
267.
20. Lane DP (1992) Cancer. p53, guardian of the
9. Henriksson M, Luscher B (1996) Proteins of the Myc genome. Nature, 358: 15-16.
network: essential regulators of cell growth and differenti-
ation. Adv Cancer Res, 68: 109-182. 21. Hainaut P, Hollstein M (2000) p53 and human can-
cer: the first ten thousand mutations. Adv Cancer Res, 77:
10. Antonsson B, Martinou JC (2000) The Bcl-2 protein 81-137.
family. Exp Cell Res, 256: 50-57.
22. Momand J, Jung D, Wilczynski S, Niland J (1998) The
11. Knudson AG (1996) Hereditary cancer: two hits revis- MDM2 gene amplification database. Nucleic Acids Res,
ited. J Cancer Res Clin Oncol, 122: 135-140. 26: 3453-3459.
12. Knudson AG, Jr. (1971) Mutation and cancer: statisti- 23. Chin L, Pomerantz J, DePinho RA (1998) The
cal study of retinoblastoma. Proc Natl Acad Sci U S A, 68: INK4a/ARF tumor suppressor: one gene--two products--
820-823. two pathways. Trends Biochem Sci, 23: 291-296.
13. Bartek J, Bartkova J, Lukas J (1997) The retinoblas- 24. Sherr CJ (2000) The Pezcoller lecture: cancer cell
toma protein pathway in cell cycle control and cancer. Exp cycles revisited. Cancer Res, 60: 3689-3695.
Cell Res, 237: 1-6.
14. Lukas J, Muller H, Bartkova J, Spitkovsky D, Kjerulff
AA, Jansen-Durr P, Strauss M, Bartek J (1994) DNA tumor
virus oncoproteins and retinoblastoma gene mutations

Oncogenes and tumour suppressor genes 103


THE CELL CYCLE

tion of cellular materials between the malian cells, these mutations would have
SUMMARY daughter cells. Moreover, immediately been lethal and it would therefore have
> The control of cell division is critical to before or after the cell cycle, various fac- been impossible to characterize them.
normal tissue structure and function. It tors interact to determine whether the cell These mutants were called cdc, for cell
is regulated by a complex interplay of divides again or whether the cell becomes division cycle mutants, and many of them
many genes that control the cell cycle, committed to a programme of differentia- have been accorded wider recognition
with DNA replication (S phase) and tion or of cell death. Therefore, the term through the application of their names to
mitosis as major checkpoints. cell cycle is often used in a broad sense the mammalian homologues correspon-
> The cell cycle is tightly regulated to to refer to, as well as the basic, self-repli- ding to the yeast genes.
minimize transmission of genetic damage cating cellular process, a number of con-
to subsequent cell generations. nected processes which determine pre-
> Progression through the cell cycle is and post-mitotic commitments. These
primarily controlled by cyclins, asso- may include the commitment to stop
ciated kinases and their inhibitors. dividing in order to enter a quiescent
Retinoblastoma (RB) and p53 are major state, to undergo senescence or differen-
suppressor genes involved in the G1/S tiation, or to leave the quiescent state to
checkpoint control. re-enter mitosis.
> Cancer may be perceived as the conse-
quence of loss of cell cycle control and Molecular architecture of the cell cycle
progressive genetic instability. The molecular ordering of the cell cycle is
a complex biological process dependent
upon the sequential activation and inacti-
vation of molecular effectors at specific
points of the cycle. Most current knowl-
Classically, the cell cycle refers to the edge of these processes stems from
set of ordered molecular and cellular experiments carried out in the oocyte of
processes during which genetic material the frog, Xenopus laevis, or in yeast, either Fig. 3.25 Proliferating cells in the basal parts of
is replicated and segregates between two Saccharomyces cerevisiae (budding yeast) the colonic crypts, visualized by immunohisto-
chemistry (stained brown).
newly generated daughter cells via the or Schizosaccharomyces pombe (fission
process of mitosis. The cell cycle can be yeast). The Xenopus oocyte is, by many cri-
divided into two phases of major morpho- teria, one of the easiest cells to manipulate
logical and biochemical change: M phase in the laboratory. Its large size (over a mil-
(mitosis), during which division is evi- limetre in diameter) means that cell cycle
dent morphologically and S phase (syn- progression can be monitored visually in
thesis), during which DNA is replicated. single cells. Microinjections can be per-
These two phases are separated by so- formed for the purpose of interfering with
called G (gap) phases. G1 precedes S specific functions of the biochemical
phase and G2 precedes M phase. machinery of the cell cycle. The Xenopus
During progression through this division oocyte has proven to be an invaluable tool
cycle, the cell has to resolve a number of in the study of the biochemistry of the cell
critical challenges. These include ensuring cycle, allowing, among other findings, the
that sufficient ribonucleotides are avail- elucidation of the composition and regula-
able to complete DNA synthesis, proof- tion of maturation promoting factor (MPF),
reading, editing and correcting the newly- a complex enzyme comprising a kinase
synthesized DNA; that genetic material is (p34cdc2) and a regulatory subunit (cyclin Fig. 3.26 A human osteosarcoma cell nucleus
during mitosis. Cell division proceeds clockwise
not replicated more than once; that the B), which drives progression from G2 to M from upper right through interphase, prophase
spatial organization of the mitotic spindle phase [1]. In contrast, the exceptional (centre), prometaphase, metaphase, anaphase
apparatus is operational; that the packing genetic plasticity of yeast has allowed the and telophase. During the cycle, the chromo-
and the condensation of chromosomes is identification of scores of mutants with somes are replicated, segregated and distributed
optimal; and that there is equal distribu- defects in cell cycle progression; in mam- equally between the two daughter cells.

104 Mechanisms of tumour development


One of the earliest genes to be identified the activity of cyclin/CDK complexes. functional properties: the WAF1/CIP1
in this way was cdc2. Isolated in S. Downstream of cyclin/CDKs are effector family (p21), the KIP family (p27, p57) and
pombe, cdc2 was determined to be able molecules, essentially transcription fac- the INK4 family (p16, p15, p18) (Fig.
to correct a G2 cell cycle arrest defect. tors, which control the synthesis of pro- 3.27).
The product of this gene, a serine-threo- teins that mediate the molecular and cel- Downstream effectors of cyclin/CDKs
nine kinase of molecular weight 32- lular changes occurring during each include proteins mediating three main
34,000 daltons, was subsequently shown phase. functional categories: (1) those involved
to be the yeast homologue of the kinase CDKIs are small proteins that form com- in the control of the enzymes responsible
contained in the Xenopus MPF. This plexes with both CDKs and cyclins [3]. for DNA replication, proof-reading and
enzyme became the paradigm of a class Their role is primarily to inhibit the activi- repair, (2) those involved in chromosome
of enzymes now called cyclin-dependent ties of cyclin/CDK complexes and to neg- and chromatin remodelling and in the
kinases, or CDKs. In their active form, atively regulate cell cycle progression. control of genomic integrity and (3) those
CDKs form heterodimers with cyclins, a They constitute the receiving end of many involved in the mechanics of cell division
class of molecules synthesized in a time- of the molecular cascades signalling (including the formation of the centro-
dependent manner during the cell cycle. growth promotion or suppression of some and the mitotic spindle, and in the
The progression of the cell cycle depends growth. Thus CDKIs may be considered as resorption of the nuclear membrane).
upon the sequential activation and inacti- the interface between the cell cycle These processes require the coordinated
vation of cyclin/CDK complexes [2], a machinery and the network of molecular synthesis of hundreds of cellular pro-
process which requires the synthesis of pathways which signal proliferation, teins. Transcription factors of the E2F
cyclins, the formation of a complex death or stress responses. However, by family play a critical role in the control of
between a specific cyclin and a CDK and virtue of their complexing properties, gene transcription during cell cycle pro-
post-translational modification of the some CDKIs also play a positive role in gression (Fig. 3.28). In G1, factors of the
CDK to convert the enzyme to an active cell cycle progression by facilitating the E2F family are bound to their DNA tar-
form (Fig. 3.27). assembly of cyclin/CDK complexes. For gets but are maintained in a transcrip-
Progression through the cell cycle as example, p21, the product of the CDKN1A tionally inactive state by the binding of
mediated by cyclins is, in turn, deter- gene (also known as WAF1/CIP1), pro- proteins of the retinoblastoma (pRb) pro-
mined by factors categorized as having motes the assembly of cyclin D/cdk2 tein family. At the G1/S transition, the
either regulatory (upstream) or effector complexes in G1 at a stoichiometric 1:1 sequential phosphorylation of pRb by
(downstream) roles. Upstream of ratio, but inhibits the activities of these several cyclin/CDKs dissociates pRb
cyclin/CDKs are regulatory factors complexes when expressed at higher lev- from the complexes, allowing E2Fs to
called cyclin-dependent kinase inhibitors els. There are three main families of interact with transcription co-activators
(CDKIs), that regulate the assembly and CDKIs, each with distinct structural and and to initiate mRNA synthesis [4].

Gene (chromosome) Product Type of alteration Role in cell cycle Involvement in cancer

p53 (17p13) p53 Mutations, deletions Control of p21, 14-3-3, etc. Altered in over 50% of all
cancers

CDKN2A (9p22) p16 and p19ARF Mutations, deletions, Inhibition of CDK4 and 6 Altered in 30-60% of all
hypermethylation cancers

RB1 (13q14) pRb Deletions Inhibition of E2Fs Lost in retinoblastomas,


altered in 5-10% of other
cancers.

CCND1 Cyclin D1 Amplification Progression into G1 10-40% of many


carcinomas

CDC25A, CDC25B cdc25 Overexpression Progression in G1, G2 10-50% of many


carcinomas

KIP1 p27 Down-regulation Progression in G1/S Breast, colon and


prostate cancers

Table 3.3 Cell cycle regulatory genes commonly altered in human cancers.

The cell cycle 105


In early G2, cdk1 is in an inactive form.
Its activation requires first association
with cyclin B, followed by post-transla-
tional modification of the kinase itself.
WAF1/CIP1 This modification includes phosphoryla-
tion of a conserved threonine residue
CDK2A (Thr161) by a kinase complex called CAK
(CDK-activating kinase), as well as
CDK2B
dephosphorylation of two residues local-
WAF1/CIP1
INK4D
ized within the active site of the enzyme,
a threonine (Thr14) and a tyrosine
(Tyr15). The removal of these phosphate
WAF1/CIP1 groups is carried out by the dual-speci-
ficity phosphatases of the cdc25 group,
KIP1
WAF1/CIP1 comprising three isoforms in humans (A,
B and C). Activation of these phos-
KIP2
WAF1/CIP1 phatases is therefore crucial for the acti-
vation of cyclin B/cdk1 complexes. The
KIP1 phosphatase is directly controlled by a
number of regulators, including plk1
KIP2
(polo-like kinase), an activating kinase,
Fig. 3.27 The progression of the cell cycle depends upon the sequential activation and inactivation of pp2A, (protein phosphatase 2A), an
cyclin/CDK complexes. This process requires the synthesis of cyclins, the formation of a complex inhibitory phosphatase and 14-3-3s, a
between a specific cyclin and a CDK, and modification of the CDK to convert this enzyme to an active
form. The enzymes activity may be disrupted by a specific inhibitor, a CDKI. signal transduction molecule which com-
plexes with cdc25, sequesters it in the
cytoplasm and thus prevents it from
Through this mechanism, E2Fs exert a germ cells during the second division of dephosphorylating its nuclear targets. Of
dual function both as transcriptional meiosis: cells that have undergone the course, the action of cdc25 phos-
repressors in G1, when bound to pRb, and first, asymmetric division of the meiotic phatases is counteracted by kinases that
as transcriptional activators in G1/S and cycle arrest in G2 until completing the sec- restore the phosphorylation of Thr14 and
in S phase, after dissociation of pRb from ond division, which is triggered by fertiliza- Tyr15, named wee1 and mik1 [6].
the complex. Recent observations suggest tion. This concept of cell cycle check- Following the activation process out-
that transcriptional repression by E2Fs is points was later extended to all mam- lined above, the cyclin B/cdk1 complex
essential to prevent the premature activa- malian cells [5]. It is now common to envis- is potentially able to catalyse transfer of
tion of cell cycle effectors, which would age the mammalian cell cycle as a succes- phosphates to substrate proteins.
scramble the temporal sequence of sion of checkpoints that have to be negoti- However, in order to achieve this, it has
molecular events and preclude cell cycle ated in order for division to be achieved. to escape the control exerted by CDKIs,
progression. There is no clear agreement on how many such as p21. The function of this CDKI is
such checkpoints exist in the mammalian itself controlled by several activators,
Cell cycle checkpoints cell cycle, or on their exact position. including BRCA1, the product of a breast
The notion of cell cycle checkpoints is cancer susceptibility gene (Oncogenes
also derived from early studies in Xenopus Control of cdk1 at G2/M transition and tumour suppressor genes, p96). The
oocytes and in yeast mutants. In S. cere- The regulation of the complex between p21 protein is removed from the com-
visiae, commitment to the mitotic cycle cdk1 (also called p34cdc2) and cyclin B plex by a still poorly understood phos-
requires the crossing of a restriction exemplifies how different factors co-oper- phorylation process, which also drives
point called the start transition. Failure to ate to control the activation of rapid degradation of the protein by the
cross this transition results in cells being cyclin/CDK complexes at a cell cycle proteasome. This leaves the cyclin
blocked in the G1 phase of the cycle. checkpoint. This activation process B/cdk1 complex ready to function, after
Another control point has been clearly requires co-operation between three lev- a final step of autophosphorylation, in
identified after S phase, at the transition els of regulation: association between the which cdk1 phosphorylates cyclin B. The
between G2 and M phases. Cells unable to two partners of the complex, post-trans- complex is now fully active and ready to
cross this checkpoint may remain blocked lational modifications of the kinase and of phosphorylate many different sub-
in a pre-mitotic, tetraploid state. the cyclin, and escape from the negative strates, such as nuclear lamins, during
Physiologically, this checkpoint is active in regulation exerted by the CDKIs. entry into mitosis.

106 Mechanisms of tumour development


Regulation of the cell cycle and control
of genetic stability
During the cell cycle, a number of potential
problems may result in damage to the
genome. These problems may arise at
three distinct stages: (1) during DNA repli-
cation, especially if the cell is under condi-
tions of stress that favour the formation of
DNA damage (irradiation, exposure to car-
cinogens etc.), (2) following the termination
of DNA replication, when the cell effective-
ly switches off its DNA synthesis machin-
ery and (3) during M phase, when the cell
has to negotiate the delicate task of segre-
gating chromatids equally. A tight coupling
between these processes and cell cycle
regulation is therefore crucial to allow the
cell to pause during the cell cycle in order
to afford the time necessary for the suc-
cessful completion of all the operations of
DNA and chromosome maintenance. Fig. 3.28 Progression from G1 to S phase is regulated by phosphorylation of the retinoblastoma protein
Failure to do this may result in both genet- (pRb), in the absence of which DNA replication cannot proceed.
ic and genomic instabilities, which are hall-
marks of cancer. Genetic instability is char- The cell cycle and cancer pressor activity remains to be deter-
acterized by an increased rate of gene Genes involved in cell cycle control are mined). Unlike the CDKN2A/INK4A gene,
mutation, deletion or recombination important among those subject to the the CDKN1A gene (encoding p21) is rarely
(essentially due to defects in DNA repair). genetic alterations that give rise to can- disrupted in cancer. As p21 plays many
Genomic instability results in chromosome cer [8]. However, the proliferation of can- roles in the negative regulation of almost
translocations, loss or duplication of large cer cells requires that the cells retain all phases of the cell cycle, loss of this
chromosome fragments and aberrant chro- functional cell cycle processes. The cell function might be expected to result in
mosome numbers (aneuploidy). cycle alterations seen in cancer are main- uncontrolled cell division. This is appar-
Tens of molecules have been identified as ly confined to two major sets of regula- ently not the case, as mice lacking the
components of the signalling cascades tors: those involved in the negative con- CDKN1A gene do not show an increased
which couple detection of DNA damage trol of cell cycle progression (inactivation frequency of cancer. This observation
and regulation of the cell cycle. One of of which leads to accelerated and illustrates one of the most important char-
these is the product of the tumour sup- unchecked cell proliferation) and those acteristics of cell cycle regulatory mecha-
pressor gene p53 (Oncogenes and tumour involved in coupling the maintenance of nisms: there is a large degree of redun-
suppressor genes, p96). p53 is specifically genome integrity to the cell cycle (inacti- dancy and overlap in the function of any
activated after various forms of direct DNA vation of which results in cells having particular effector. Therefore, cancer-
damage (such as single or double strand gene alterations that progressively accu- causing deregulation of the cell cycle
breaks in DNA) and regulates the transcrip- mulate during carcinogenesis) (Table 3.3) requires a combination of many alter-
tion of several inhibitors of cell cycle pro- [9]. Most of the genes corresponding to ations in genes encoding proteins that,
gression, particularly at the G1/S and these two categories fall within the group either alone or in concert, are critical for
G2/M transitions [7]. Other important mol- of tumour suppressors, and many of them the control of cell division.
ecules in this coupling process include the are also direct participants in DNA repair Apart from inactivation of negative regula-
checkpoint kinases chk1 and chk2. Chk1 is processes. tors, a few cell cycle genes may be acti-
activated after replication blockage during The gene which encodes p16 (CDKN2A/ vated as oncogenes, in that their alter-
S-phase. In turn, chk1 activates wee1 and INK4A) has been established as a tumour ation results in enhanced activity leading
mik1, two kinases that counteract the suppressor gene [10], and mutations and to accelerated cell proliferation. The best
action of cdc25 and keep cdk1 in an inac- deletions at this site are commonly found example of such a cell cycle oncogene is
tive form. Thus, through activation of chk1, in primary human tumours, especially CCND1, the gene encoding cyclin D1, a
the cell triggers an emergency mechanism melanoma (although the contribution of G1-specific cyclin [11]. This gene is locat-
that ensures that cells with incompletely another protein encoded by the same ed on chromosome 11p13, within a large
replicated DNA cannot enter mitosis. locus on chromosome 9p, p14ARF, to sup- region that is amplified in up to 20% of

The cell cycle 107


several carcinomas (e.g. breast, head and phase cyclin) and for activating mutations extremely diverse range of possibilities for
neck, oesophageal and lung cancers). of CDK4 (one of the partners of cyclin D1) cancer-associated alterations. In this
There is also limited evidence for tran- in some cancers. Indeed, the high com- respect, cancer can be seen as, funda-
scriptional activation of cyclin A (an S- plexity of cell cycle effectors provides an mentally, a disease of the cell cycle.

TELOMERES Telomerase assays have not yet entered


AND TELOMERASE routine clinical practice, but there is con-
siderable interest in their possible use for
cancer diagnosis and prognosis. For exam-
The ends of eukaryotic chromosomes are ple, telomerase assays of urine sediments
referred to as telomeres. These contain may be useful for diagnosis of urinary tract
many copies of a repetitive DNA cancer (Kinoshita H et al., J Natl Cancer
sequence, which in vertebrates is the Inst, 89: 724-730, 1997), and telomerase
hexanucleotide TTAGGG. The telomeres of activity levels may be a predictor of out-
normal human somatic cells shorten by come in neuroblastoma (Hiyama E et al.,
50 to 150 base pairs every time cell divi- Nature Medicine, 1: 249-255, 1995).
sion occurs. This appears to act as a cell Fig. 3.29 Telomeres contain repetitive DNA
division counting mechanism: when a The catalytic subunit of human telomerase, sequences that cap the ends of chromosomes.
Quantitative fluorescence in situ hybridization
cell's telomeres have shortened below a hTERT, was cloned in 1997 (Lingner J, Cech analysis of human metaphase chromosome
critical length, the cell exits permanently TR, Curr Opin Genet Dev 8: 226-232, 1998). It spreads is shown, using oligonucleotide probes
from the cell cycle. Normal cells thus has subsequently been shown that genetic specific for telomere (white) and centromere (red)
DNA sequences, and the DNA dye DAPI (blue).
have a limited proliferative capacity, and manipulations of hTERT which result in inhi- From the laboratory of Drs J.W. Shay and W.E.
this acts as a major barrier against car- bition of telomerase activity in tumour cells Wright.
cinogenesis. Cells that have accumulated limit their proliferation and often result in cell
some carcinogenic changes are unable to death. This raises the possibility that telom-
form clinically significant cancers unless erase inhibitors may be a very useful form of A potential challenge facing telomerase
this proliferation barrier is breached. therapy for many or most types of cancer. research is the finding that some cancers
More than 85% of all cancers achieve this However, in tumours with long telomeres, it maintain their telomeres by a mechanism
by expressing an enzyme, telomerase, may take many cell divisions before telom- that does not involve telomerase,
that synthesizes new telomeric DNA to erase inhibitors exert an anti-tumour effect. referred to as alternative lengthening of
replace the sequences lost during cell When such drugs are developed they will telomeres, ALT (Bryan TM et al., Nature
division (Shay JW, Bacchetti S, Eur J therefore need to be carefully integrated with Medicine, 3: 1271-1274, 1997; Reddel RR,
Cancer, 33A: 787-791, 1997). other anticancer treatments. J Clin Invest, 108: 665-667, 2001).

REFERENCES WEBSITES
1. Hunt T (1989) Maturation promoting factor, cyclin and 7. Hainaut P, Hollstein M (2000) p53 and human cancer: Animation of the phases of the cell cycle and of mitosis:
the control of M-phase. Curr Opin Cell Biol, 1: 268-274. the first ten thousand mutations. Adv Cancer Res, 77: 81- http://www.cellsalive.com/
137. Nature Reviews, Focus on cell division:
2. Pines J (1995) Cyclins and cyclin-dependent kinases: a
biochemical view. Biochem J, 308 (Pt 3): 697-711. 8. Hartwell LH, Kastan MB (1994) Cell cycle control and http://www.nature.com/ncb/celldivision/
cancer. Science, 266: 1821-1828. The Forsburg laboratory home pages, a guide to the cell
3. Sherr CJ, Roberts JM (1999) CDK inhibitors: positive
and negative regulators of G1-phase progression. Genes 9. Kinzler KW, Vogelstein B (1997) Cancer-susceptibility cycle and DNA replication in S. pombe:
Dev, 13: 1501-1512. genes. Gatekeepers and caretakers. Nature, 386: 761, 763. http://pingu.salk.edu/~forsburg/lab.html
4. Weinberg RA (1995) The retinoblastoma protein and 10. Strohmaier H, Spruck CH, Kaiser P, Won KA, Sangfelt
cell cycle control. Cell, 81: 323-330. O, Reed SI (2001) Human F-box protein hCdc4 targets
cyclin E for proteolysis and is mutated in a breast cancer
5. Hartwell LH, Weinert TA (1989) Checkpoints: controls
cell line. Nature, 413: 316-322.
that ensure the order of cell cycle events. Science, 246:
629-634. 11. Schuuring E (1995) The involvement of the chromo-
some 11q13 region in human malignancies: cyclin D1 and
6. Zeng Y, Forbes KC, Wu Z, Moreno S, Piwnica-Worms H,
EMS1 are two new candidate oncogenes--a review. Gene,
Enoch T (1998) Replication checkpoint requires phospho-
159: 83-96.
rylation of the phosphatase Cdc25 by Cds1 or Chk1.
Nature, 395: 507-510.

108 Mechanisms of tumour development


CELL-CELL COMMUNICATION

mony with normal neighbouring cells, it is tal to the nature of malignancy, it has long
SUMMARY not surprising that the function of genes been postulated that gap junctional inter-
involved in intercellular communication cellular communication is disturbed in
> Cells communicate by means of secret- mechanisms is disrupted in many cancer. The first confirmatory evidence
ed molecules which affect neighbouring
tumours. Thus, several oncogenes (Onco- was the observation that of a reduced
cells carrying appropriate receptors,
and also by direct cell contact, including
genes and tumour suppressor genes, p96) level of gap junctional intercellular com-
specifically includes gap junctions. encode products involved in humoral inter- munication in one tumour type [8]. This
cellular communication: c-erb, c-erbB2 phenomenon has now been observed in
> Cell contact-mediated communication and c-SIS [3]. It has also become clear almost all tumours [4]. Cell lines estab-
through gap junctions is controlled by that cell contact-mediated intercellular lished from tumours, as well as cells
connexin genes and is often disrupted in communication plays a crucial role in cell transformed in vitro, usually exhibit
cancer. This may contribute to uncon- growth control [4] and genes involved are impaired function in respect of gap junc-
trolled and autonomous growth. often classified as tumour suppressor tional intercellular communication. Gap
genes [5]. Cell adhesion molecules are junctional intercellular communication
> Interventions restoring gap junction
communication may provide a basis for
also involved in cell-cell recognition. There between transformed cells and neigh-
therapy. are several lines of evidence which sug- bouring normal counterparts is selective-
gest that aberrant functions of cell adhe- ly defective in murine embryonic
sion may be involved in tumour invasion BALB/c3T3 cells (Fig. 3.31). A lack of het-
and metastasis [6]. erologous gap junctional intercellular
communication between transformed and
Gap junctional intercellular communi- normal cells has been observed using rat
In complex organisms, neighbouring cells cation and cancer liver epithelial cell lines and rat liver
behave and function in harmony for the Gap junctional intercellular communica- tumour in vivo. It appears that reduced
benefit of the whole organism through the tion is the only means by which cells gap junctional intercellular communica-
operation of cell-cell communication. exchange signals directly from the interi- tion is common to many tumour cells.
During evolution, various types of intercel- or of one cell to the interior of surround- Further studies with multistage models of
lular communication have developed, ing cells [7]. Since the extent to which rat liver and mouse skin carcinogenesis
which in mammals take two forms: (1) tumour cells deviate from cells which have revealed that there is, in general, a
humoral communication and (2) cell con- exhibit tissue homeostasis is fundamen- progressive decrease in the level of gap
tact-mediated communication (Fig. 3.30).
Humoral communication is typically medi-
ated by molecules, such as growth factors A. Humoral communication
and hormones, excreted from certain cells
and received by receptors of other cells.
Intercellular communication based on
direct cell-cell contact is mediated by var-
ious junctions, including adherence junc-
tions, desmosomes and gap junctions.
During multistage carcinogenesis, genes
critically involved in cell growth are B. Cell contact-mediated communication
altered [1]. Most such genes are known to 1.Cell adhesion 2. Gap junction
be directly or indirectly involved in the
control of cell replication (The cell cycle,
p104) or in the death of individual cells [2]
(Apoptosis, p113). Genes involved in inter-
cellular communication control cellular
growth at another level. These genes func-
tion to maintain cell growth in harmony
with that of the surrounding tissue. Since Fig. 3.30 Relationships between cells are maintained by different types of intercellular communication,
most cancer cells do not proliferate in har- which may (B) or may not (A) require cell contact.

Cell-cell communication 109


A Injection into transformed cell B junctional intercellular communication
during carcinogenesis and tumour pro-
gression.
Another line of evidence, that implies a
causal role for blockage of intercellular
communication in carcinogenesis, is that
agents or genes involved in carcinogenesis
have been shown to modulate gap junc-
tional intercellular communication. The
mouse skin tumour-promoting agent 12-O-
Phase-contrast micrographs Fluorescence micrographs tetradecanoylphorbol 13-acetate (TPA)
inhibits gap junctional intercellular commu-
nication. Many other tumour-promoting
agents inhibit gap junctional intercellular
communication [9]. In addition to such
chemicals, other tumour-promoting stimuli,
such as partial hepatectomy and skin
wounding, have been demonstrated to
inhibit gap junctional intercellular commu-
nication. Activation of various oncogenes,
C Injection into non-transformed cell D
including those which encode src, SV-40 T
Fig. 3.31 Selective gap junctional communication: cells transformed by a chemical carcinogen (spindle- antigen, c-erbB2/neu, raf, fps and ras, also
shaped and criss-crossed) communicate among themselves, but not with their surrounding non-trans- results in inhibition of gap junctional inter-
formed counterparts. When a gap junction-diffusible fluorescent dye is microinjected into a single cell
(marked with star) of a transformed focus there is communication between transformed cells but not with cellular communication. Conversely, some
the surrounding non-transformed cells (A, B). Injection of the dye into a non-transformed cell which is chemopreventive agents enhance gap junc-
located near a transformed focus results in communication between non-transformed cells, but not with tional intercellular communication [10].
transformed cells (C, D).

Cells carrying the tk gene are GCV-P cannot pass through


killed by the toxic phosphorylated the cell membrane
GCV (GCV-P) they produce

No bystander
GCV-P effect
Transfected cells
carrying the HSV-tk gene

Ganciclovir
(GCV)

Tumour cell population

Bystander
GCV-P effect

Movement of GVC-P
from cytoplasm to
cytoplasm in gap
junctions

Fig. 3.32 Role of cell-cell interaction in gene therapy. In a tumour cell population, only a few cells can be reached by vectors carrying the HSV-tk gene.
Expression of the tk gene (orange) makes these cells sensitive to ganciclovir: they produce phosphorylated ganciclovir, which is toxic. As phosphorylated gan-
ciclovir cannot pass through the cell membrane, theoretically only cells expressing the tk gene will die as a result of ganciclovir treatment. Transmembrane dif-
fusion of phosphorylated ganciclovir from cytoplasm to cytoplasm can induce a bystander effect sufficient to eradicate a tumour cell population, even if only a
few cells express the tk gene [13].

110 Mechanisms of tumour development


Connexin genes and tumour suppres- Enhancement of cancer therapy by Signal transduction from intercellular
sion cell-cell communication network
The first indication that normal cells may A decade ago it was demonstrated that The main physiological function of gap
suppress the growth of malignant cells gap junctional intercellular communica- junctional intercellular communication is
with which they are in contact came from tion could be exploited to distribute ther- probably to maintain homeostasis by
the work of Stoker and colleagues [11]. apeutic agents among cancer cells and keeping the level of signals mediated by
More recent evidence for such a direct thereby enhance cancer therapy [12]. agents of low molecular weight at equilib-
role of gap junctional intercellular com- One principle of gene therapy is the rium among cells linked by gap junctions.
munication in tumour suppression has mediation of selective cytotoxicity by the This implies that intercellular communica-
come from experiments in which connex- introduction, into malignant cells, of a tion may control cell growth indirectly. As
in genes were transfected into gap junc- gene that activates an otherwise innocu- already noted, such an activity is distinct
tional intercellular communication-defi- ous drug. In practice, only a fraction of from that mediated by genes which are
cient malignant cell lines. In many cases, the total number of tumour cells sought directly involved in cell growth and death.
connexin gene expression reduced or to be eliminated, are successfully trans- One particularly important pathway link-
eliminated tumorigenicity of recipient fected with the gene in question. ing intercellular interaction to signal trans-
cells [10]. However, at least in the case of brain duction involves the cell adhesion mole-
Although tumour suppressor genes, most tumour therapy based upon the thymi- cule -catenin. If the level of -catenin in
notably p53, are mutated in a high pro- dine kinase gene from herpes simplex the cytoplasm and nucleus rises, it acti-
portion of tumours, few mutations of con- virus (HSV-tk), not only are the cells vates transcription factors of the TCF(T-
nexin genes have yet been found in transfected with the gene affected by cell factor)/LEF family and increases
rodent tumours and none has been treatment with the drug ganciclovir, but activity of genes including C-MYC, cyclin
reported for any human cancer. Although neighbouring cells are also killed in the D1 and connexin-43. Normally the levels
this suggests that connexin gene muta- presence of ganciclovir. Several studies of -catenin in the cytoplasm and nucleus
tions are rare in carcinogenesis, only a have provided strong evidence that this are kept very low because a complex of
few studies (all from one laboratory) on a phenomenon, termed the bystander proteins including the APC gene product
limited number of connexin genes (Cx32, effect (Fig. 3.32), is due to connexin- (adenomatous polyposis coli, Colorectal
Cx37[4] and Cx43) have been conduct- mediated gap junctional intercellular cancer, p198), axin and glycogen synthe-
ed. Several polymorphisms in connexin communication; that is, ganciclovir phos- sis kinase 3 bind the free -catenin and
genes in humans and rats have been phorylated by HSV-tk can diffuse through put a phosphate group onto it which
described, although there was no appar- gap junctions and even those cells with- marks it for destruction [14].
ent correlation between such polymor- out HSV-tk gene can be killed. The role of In normal cells the level of free -catenin
phisms and the cancer sites examined connexin genes in this effect has been is regulated by the Wnt ("wingless homo-
[10]. confirmed [13]. logue") signal from outside the cell, which

Gene Cancer site/cancer Changes observed

Integrin Skin, liver, lung, osteosarcoma Reduced expression

E-cadherin Stomach, colon, breast, prostate Mutations: reduced expression

-catenin Stomach, colon, breast, prostate, Reduced expression


oesophagus, kidney, bladder, etc
-catenin Melanoma, colon Mutations: reduced expression

-catenin Breast, colon Loss of expression, translocation into nuclei

Connexins Liver, skin etc Reduced expression, aberrant localization

Table 3.4 Examples of cell-cell interaction genes involved in carcinogenesis [10].

Cell-cell communication 111


increases the level by transiently reducing accepts the phosphate allow the levels of occur in many cancers, including those of
the activity of the kinase. However, muta- -catenin to rise; when this happens the the colon, breast and endometrium,
tions in either the APC gene or in the part TCF/LEF-controlled genes are permanent- shows the importance of this pathway and
of the -catenin (CTNNB1) gene which ly activated. The fact that mutations of the emphasizes the connection between cell-
codes for the part of the molecule which CTNNB1, AXIN1, AXIN2 and APC genes cell contact and signal transduction.

REFERENCES
1. Fearon ER, Vogelstein B (1990) A genetic model for 7. Bruzzone R, White TW, Paul DL (1996) Connections autoradiographic studies using marked cells. J Cell Sci, 2:
colorectal tumorigenesis. Cell, 61: 759-767. with connexins: the molecular basis of direct intercellular 293-304.
2. Kinzler KW, Vogelstein B (1997) Cancer-susceptibility signaling. Eur J Biochem, 238: 1-27. 12. Yamasaki H, Katoh F (1988) Novel method for selective
genes. Gatekeepers and caretakers. Nature, 386: 761, 763. 8. Loewenstein WR, Kanno Y (1966) Intercellular commu- killing of transformed rodent cells through intercellular
3. Heldin CH (1996) Protein tyrosine kinase receptors. nication and the control of tissue growth: lack of commu- communication, with possible therapeutic applications.
Cancer Surv , 27: 7-24. nication between cancer cells. Nature, 209: 1248-1249. Cancer Res, 48: 3203-3207.

4. Krutovskikh V, Yamasaki H (1997) The role of gap junc- 9. Trosko JE, Chang CC, Madhukar BV, Klaunig JE (1990) 13. Mesnil M, Yamasaki H (2000) Bystander effect in her-
tional intercellular communication (GJIC) disorders in Chemical, oncogene and growth factor inhibition gap junc- pes simplex virus-thymidine kinase/ganciclovir cancer
experimental and human carcinogenesis. Histol tional intercellular communication: an integrative hypothe- gene therapy: role of gap-junctional intercellular communi-
Histopathol, 12: 761-768. sis of carcinogenesis. Pathobiology, 58: 265-278. cation. Cancer Res, 60: 3989-3999.

5. Hirohashi S (1998) Inactivation of the E-cadherin-medi- 10. Yamasaki H, Omori Y, Zaidan-Dagli ML, Mironov N, 14. Behrens J, Jerchow BA, Wurtele M, Grimm J, Asbrand
ated cell adhesion system in human cancers. Am J Pathol, Mesnil M, Krutovskikh V (1999) Genetic and epigenetic C, Wirtz R, Kuhl M, Wedlich D, Birchmeier W (1998)
153: 333-339. changes of intercellular communication genes during mul- Functional interaction of an axin homolog, conductin, with
tistage carcinogenesis. Cancer Detect Prev, 23: 273-279. beta-catenin, APC, and GSK3beta. Science, 280: 596-599.
6. Birchmeier EJ, Behrens J (1994) Cadherin expression in
carcinoma: Role in the formation of cell junctions and pre- 11. Stoker MG (1967) Transfer of growth inhibition
vention of invasiveness. Biochim Biophys Acta, 1198: 11-26. between normal and virus-transformed cells:

112 Mechanisms of tumour development


APOPTOSIS

engulfing respectively [1]. The regulato- dependent on definition of the ced genes
SUMMARY ry phase includes all the signalling path- in the nematode Caenorhabditis elegans,
ways that culminate in commitment to members of this gene family being vari-
> The term apoptosis refers to a type of cell death. Some of these pathways regu- ously homologous to human BCL2 (which
cell death that occurs both physiologi-
late only cell death, but many of them suppresses apoptosis), APAF-1 (which
cally and in response to external stimuli,
including X-rays and anticancer drugs.
have overlapping roles in the control of mediates caspase activation) and the
cell proliferation, differentiation, respons- caspases themselves (proteases which
> Apoptotic cell death is characterized by es to stress and homeostasis. Critical to mediate cell death). The centrality of
distinctive morphological changes dif- apoptosis signalling are the initiator apoptosis to cancer biology is indicated
ferent from those occurring during caspases (including caspase-8, caspase-9 by excess tumorigenesis in BCL2-trans-
necrosis, which follows ischaemic injury and caspase-10) whose role is to activate genic and p53-deficient mice. An appreci-
or toxic damage. the more abundant effector caspases ation of apoptosis provides a basis for the
(including caspase-3 and caspase-7) further development of novel and conven-
> Apoptosis is regulated by several dis- which, in turn, brings about the morpho- tional cancer therapy
tinct signalling pathways. Dysregulation
logical change indicative of apoptosis.
of apoptosis may result in disordered
cell growth and thereby contribute to
Finally, the engulfing process involves the The role of cell death in tumour
carcinogenesis. recognition of cellular remains and their growth
elimination by the engulfing activity of Apoptosis, or lack of it, may be critical to
> Selective induction of apoptosis in tumour surrounding cells. tumorigenesis [2]. BCL2, a gene mediat-
cells is among current strategies for the Identification of genes mediating apopto- ing resistance to apoptotic stimuli, was
development of novel cancer therapies. sis in human cells has been critically discovered at the t(14:18) chromosomal

APOPTOSIS
Apoptosis is a mode of cell death that
facilitates such fundamental processes as Condensation and
fragmentation of
development (for example, by removal of chromatin
unwanted tissue during embryogenesis)
and the immune response (for example,
by elimination of self-reactive T cells). NECROSIS
This type of cell death is distinguished
from necrosis both morphologically (Fig.
3.33) and functionally. Specifically, apop-
Shrinking/
tosis involves single cells rather than Organelle rounding up,
areas of tissue and does not provoke disruption fragmentation
inflammation. Tissue homeostasis is and break- of cell and
down, nucleus
dependent on controlled elimination of cell swelling
unwanted cells, often in the context of a
continuum in which specialization and
maturation is ultimately succeeded by
cell death in what may be regarded as the
final phase of differentiation. Apart from
elimination in a physiological context, Membrane
cells that have been lethally exposed to blebbing,
cytotoxic drugs or radiation may be sub- residual Engulfing by
ghost cell neighbouring cell
ject to apoptosis. as apoptotic-
The process of apoptosis can be body
described by reference to distinct phases,
termed regulation, effector and Fig. 3.33 Apoptosis and necrosis are distinguished by characteristic morphological changes.

Apoptosis 113
translocation in low grade B cell non-
DNA DAMAGE REPLICATION STRESS
Hodgkin lymphoma. It thus became
apparent that neoplastic cell expansion
could be attributable to decreased cell
death rather than rapid proliferation.
Defects in apoptosis allow neoplastic
cells to survive beyond senescence, Rad17
Rad9, Rad1, Hus1
thereby providing protection from hypoxia ATM + ATR
and oxidative stress as the tumour mass
expands. Growth of tumours, specifically
in response to chemical carcinogens, has Chk1 Chk2 Mdm2 p53 BRCA1 Nbs1 cAbl
been correlated with altered rates of
apoptosis in affected tissues as cell pop-
ulations with altered proliferative activity Cdc25 p21
14-3-3
emerge. Paradoxically, growth of some
cancers, specifically including breast, has
Cdk
been positively correlated with increasing
CELL CYCLE APOPTOSIS TRANSCRIPTION DNA REPAIR
apoptosis [3]. ARREST

Fig. 3.34 Response to DNA damage is mediated by several signalling pathways and may include apoptosis.
Interrelationships between mitogenic
and apoptotic pathways
A dynamic relationship between regula- damage caused by ionizing radiation, con-
tion of growth/mitosis and apoptosis may trolling the initial phosphorylation of pro-
be demonstrated using a variety of rele- teins such as p53, Mdm2, BRCA1, Chk2
vant signalling pathways. Many differing and Nbs1. Other sensors of DNA damage
promoters of cell proliferation have been include mammalian homologues of the
found to possess pro-apoptotic activity PCNA-like yeast proteins Rad1, Rad9 and
[4]. Thus, ectopic expression of the C-MYC Hus1, as well as the yeast homologue of
oncogene (normally associated with prolif- replication factor C, Rad17. Specific mole-
erative activity) causes apoptosis in cul- cules detect nucleotide mismatch or inap-
tured cells subjected to serum deprivation propriate methylation. Following exposure
(which otherwise prevents proliferation). of mammalian cells to DNA-damaging
Fig. 3.35 Apoptotic cell death requires gap junc-
Oncogenes that stimulate mitogenesis agents, p53 is activated and among many tional intercellular communication. Expression
can also activate apoptosis. These include targets consequently upregulated are the and subcellular location of connexin 43 in healthy
oncogenic RAS, MYC and E2F. Mutations cyclin-dependent kinase inhibitor p21 (A) and in apoptotic (B) rat bladder carcinoma
in E2F that prevent its interaction with the (which causes G1 arrest) and Bax (which cells. Arrows indicate location of connexin 43 in
areas of intercellular contact between apoptotic
retinoblastoma protein (pRb) accelerate S induces apoptosis). Thus, the tumour sup- (B) and non-apoptotic (A) cells. Counterstaining of
phase entry and apoptosis. A function of pressor gene p53 mediates two responses DNA with propidium iodide reveals fragmentation
pRb is to suppress apoptosis: pRb-defi- to DNA damage by radiation or cytotoxic of the nucleus typical of apoptosis (B).
cient cells seem to be more susceptible to drugs: cell cycle arrest at the G1 phase of
p53-induced apoptosis. the cell cycle and apoptosis (Oncogenes
Agents such as radiation or cytotoxic drugs and tumour suppressor genes, p96). The
cause cell cycle arrest and/or cell death serine/threonine kinase Chk2 is also able
[5]. The DNA damage caused by radiation to positively interact with p53 and BRCA1.
or drugs is detected by various means (Fig. Chk2 and the functionally related Chk1
3.34). DNA-dependent protein kinase and kinase appear to have a role in the inhibi-
the ataxia-telangiectasia mutated gene tion of entry into mitosis via inhibition of
(ATM) (as well as the related ATR protein) the phosphatase Cdc25 (The cell cycle,
bind to damaged DNA and initiate phos- p104).
phorylation cascades to transmit damage
signals. DNA-dependent protein kinase is The regulatory phase Fig. 3.36 Apoptotic cells in an adenoma, visual-
ized by immunohistochemistry (red). Apoptosis is
believed to play a key role in the response Two major apoptotic signalling pathways restricted to single cells, unlike necrosis, which
to double-stranded DNA breaks. ATM plays have been identified in mammalian cells typically involves groups of cells. Apoptosis does
an important part in the response to DNA (Fig. 3.37). The extrinsic pathway not produce an inflammatory response.

114 Mechanisms of tumour development


activates the Fas receptor in the absence
of ligand. TRAIL (TNF-related apoptosis-
CD95L
inducing ligand, Apo-2L) has 28% amino
acid identity to FasL. TRAIL induces cell
CD95 death only in tumorigenic or transformed
cells and not in normal cells [8].
FADD

Procaspase-8 The regulation of apoptosis by BCL2


DNA damage
c-FLIP family genes
p53
While the members of the death recep-
Bid BclxL tor family and their ligands have struc-
Bax tural elements in common, agents and
stimuli initiating the mitochondrial path-
Caspase-8
way to apoptosis are diverse. Common to
Bcl2
these stimuli, however, is a change in
Truncated bid mitochondrial function, often mediated by
members of the BCL2 family [9]. In
Procaspase-3
humans, at least 16 homologues of BCL2
Cytochrome c
have been identified. Several family mem-
bers (including Bcl-2, Bcl-xL, Bcl-W) sup-
Apoptosis-inducing press apoptosis, while others induce
factor (AIF)
Apoptosome
apoptosis and may be subdivided on the
basis of their ability to dimerize with Bcl-
Caspase-3
Smac/DIABLO 2 protein (Bad, Bik, Bid) or not (Bax, Bak).
Apaf-1
Procaspase-9 Phosphorylation of Bad protein by a spe-
cific (Akt/PKB) and other kinases pre-
Inhibitors of
vents dimerization with Bcl-2 and pro-
Apoptotic substrates apoptosis (IAPs) motes cell survival. At least two distinct
mechanisms of action are recognized: the
Fig. 3.37 Apoptosis occurs when specific proteases (caspases) digest critical proteins in the cell. The
binding of Bcl-2 (or other members of the
caspases are normally present as inactive procaspases. Two pathways lead to their activation. The death
receptor pathway (at the top and left side of the figure) is triggered when ligands bind to death receptors family) with either pro- or anti-apoptotic
such as CD95/Fas. The mitochondrial pathway is triggered by internal insults such as DNA damage as members of the Bcl-2 family or the for-
well as by extracellular signals. In both pathways, procaspases are brought together. They then cleave mation of pores in mitochondrial mem-
each other to release active caspase. The binding of ligand (FasL or CD95L) to CD95 brings procaspase branes. Bcl-xL is a potent death suppres-
8 molecules together; release of mitochondrial components bring procaspases 9 together. The active
caspase 8 and 9 then activate other procaspases such as procaspase 3. sor that is upregulated in some tumour
types. Bax is a death promoter that is
inactivated in certain types of colon can-
depends upon the conformational change ramifications. The best-characterized cer, stomach cancer and in haematopoi-
in certain cell surface receptors following receptors belong to the tumour necrosis etic malignancies. By dint of relevant
the binding of respective ligands. The factor (TNF) receptor gene superfamily binding sites, Bax is under the direct tran-
intrinsic pathway involves mitochondrial [7]. In addition to a ligand-binding scriptional control of p53.
function and is initiated by growth factor domain, death receptors contain homolo-
deprivation, corticosteroids or DNA dam- gous cytoplasmic sequence termed the Involvement of mitochondria
age induced by radiation or cytotoxic death domain. Members of the family Apoptosis induced by cytotoxic drugs is
drugs. include Fas/APO-1/CD95 and TNF-1 accompanied by critical changes in mito-
receptor (which binds TNF). Activation chondria. Such apoptotic stimuli induce
Cell surface receptors of the Fas (or CD95) receptor by its spe- translocation of Bax from cytosol to mito-
Apoptosis may be induced by signalling cific ligand (FasL or CD95L) results in a chondria, which induces release of
molecules, usually polypeptides such as conformational change such that the cytochrome c. Loss of transmembrane
growth factors or related molecules, death domain interacts with the adap- potential follows cytochrome c release
which bind to death receptors on the tor molecule FADD which then binds pro- and is dependent on caspase activation
cell surface [6]. Such cell death was ini- caspase-8. In some cell types, drug- (see below), whereas cytochrome c
tially investigated in relation to the induced apoptosis is associated with Fas release is not. Bcl-2 and Bcl-xL reside
immune response, but has much wider activation. Ultraviolet irradiation directly chiefly in the outer mitochondrial mem-

Apoptosis 115
brane. Bcl-2, Bcl-xL and Bax can form ion tion of caspases. However, nitric oxide is sors conserved throughout evolution. The
channels when they are added to synthet- involved in several aspects of apoptosis IAP protein survivin is overexpressed in
ic membranes, and this may be related to and may act both as a promoter and a large proportion of human cancers.
their impact on mitochondrial biology [10]. inhibitor depending on conditions [11]. Little is known about the involvement of
In the cytosol after release from mito- caspase mutations in cancer.
chondria, cytochrome c activates the cas- The effector and engulfing phases
pases through formation of a complex (the In mammals at least 13 proteases which Caspase substrates and late stages of
apoptosome) with Apaf-1 (apoptotic- mediate the breakdown of cell structure apoptosis
protease activating factor-1), procaspase- during apoptosis have been identified and Apoptosis was initially defined by refer-
9 and ATP. It appears that Bcl-2/Bcl-xL are designated caspases-1 through -13 ence to specific morphological change. In
may suppress apoptosis by either prevent- [12]. All possess an active site cysteine fact, both mitosis and apoptosis are char-
ing release of cytochrome c or interfering and cleave substrates after aspartic acid acterized by a loss of substrate attach-
with caspase activation by cytochrome c residues. They exist as inactive zymogens, ment, condensation of chromatin and
and Apaf-1. Sustained production of nitric but are activated by different processes phosphorylation and disassembly of
oxide (NO) may cause the release of mito- which most often involve cleavage of their nuclear lamins. These changes are now
chondrial cytochrome c into the cyto- pro-forms (designated procaspase-8, etc.) attributable to caspase activation and its
plasm and thus contribute to the activa- at particular sites, thereby generating sub- consequences.
units which form active proteases consist- Most of the more than 60 known caspase
ing of two large and two small subunits. substrates are specifically cleaved by cas-
Proteolytic cascades may occur with pase-3 and caspase-3 can process pro-
some caspases operating as upstream ini- caspases-2, -6, -7 and -9 [13]. Despite the
tiators (which have large N-terminal multiplicity of substrates, protease activi-
prodomains and are activated by protein- ty mediated by caspases is specific and
protein interaction) and others being seems likely to account for much of the
downstream effectors (activated by pro- morphological change associated with
tease cleavage). As noted earlier, at least apoptosis. Caspases cleave key compo-
two pathways of caspase activation can nents of the cytoskeleton, including actin
A be discerned: one involving FADD or simi- as well as nuclear lamins and other struc-
lar protein-protein complexes and the tural proteins. Classes of enzymes cleaved
other mediated by release of cytochrome by caspases cover proteins involved in
c. In the former, affinity labelling suggests DNA metabolism and repair exemplified
that caspase-8 activates caspases-3 and by poly(ADP-ribose) polymerase and DNA-
-7 and that caspase-3 in turn may activate dependent protein kinase. Other classes
caspase-6. On the other hand, release of of substrates include various kinases, pro-
cytochrome c into the cytoplasm results in teins in signal transduction pathways and
the activation of caspase-9 which in turn proteins involved in cell cycle control,
activates caspase-3. exemplified by pRb. Cleavage of some
B Though the intrinsic pathway to caspase-3 substrates is cell-type specific. Caspase
activation may be distinguished from the activity accounts for internucleosomal
extrinsic pathway (i.e. that activated by cleavage of DNA, one of the first charac-
Fas, etc.), some interaction is demonstra- terized biochemical indicators of apopto-
ble. Thus, caspase-9 is able to activate sis. ICAD/DFF-45 is a binding partner and
caspase-8. Nonetheless, the pathways are inhibitor of the CAD (caspase-activated
separate to the extent that caspase-8 null DNAase) endonuclease, and cleavage of
animals are resistant to Fas- or TNF- ICAD by caspase-3 relieves the inhibition
induced apoptosis while still susceptible and promotes the endonuclease activity
to chemotherapeutic drugs; cells deficient of CAD.
C in caspase-9 are sensitive to killing by
Fas/TNF but show resistance to drugs Therapeutic implications
Fig. 3.38 Neuroblastoma cells treated with ioni-
zing radiation undergo apoptosis. The TUNEL assay
and dexamethasone. Finally, death of In theory, knowledge of critical signalling
was used to visualize apoptotic cells (green), some cells may occur independently of or effector pathways which bring about
before (A) and 24 hours after (B) treatment with X- caspase-3. Caspases-3, -7 and 9 are apoptosis provides a basis for therapeutic
rays (5 Gray). Close-up shows that the nuclei of the inactivated by proteins of the inhibitor of intervention, including the development of
apoptotic cells are fragmented (C). apoptosis family (IAPs) which are suppres- novel drugs to activate particular path-

116 Mechanisms of tumour development


ways. Several options are under investiga- tumour types. In experimental systems, suppression of Bcl -2 by an antisense
tion [14]. More immediately, attempts are cells acquiring apoptosis defects (e.g. p53 oligonucleotide has been shown to retard
being made to exploit knowledge of apop- mutations) can more readily survive hypox- tumour growth and the approach is cur-
totic processes to increase the efficacy or ic stress and the effects of cytotoxic drugs rently subject to clinical trial. Likewise,
specificity of currently available therapy. [15]. However, clinical studies have not antisense oligonucleotides directed at
Simple answers have not emerged. Thus, consistently established that mutation of survivin are being evaluated. The possi-
for example, relatively increased expres- p53 is associated with poor response to bility of using recombinant TRAIL to
sion of Bcl-2 (which, under many experi- chemotherapy [16]. induce apoptosis in malignant cells is
mental conditions, inhibits apoptosis) is not The function of Bcl-2 family members may under investigation. TRAIL is implicated as
necessarily indicative of poor prognosis be subject to interference by small mole- the basis of all-trans-retinoic treatment of
and the reverse appears true for some cules [17]. In preclinical animal models, promyelocytic leukaemia [18]. Also note-

Ligand
DRUGS TARGETING SIGNAL
Receptor
TRANSDUCTION PATHWAYS Cell membrane

GRB SOS Ras

In complex multicellular organisms, cell


proliferation, differentiation and survival PLC PI3K Raf
are regulated by a number of extracellular
Intracellular messengers

hormones, growth factors and cytokines.


DAG Akt MEK
These molecules are ligands for cellular
receptors and communicate with the nucle-
us of the cell through a network of intracel- BAD P7056K
lular signalling pathways. In cancer cells,
key components of these signal transduc- PKC
tion pathways may be subverted by proto- BclxL ERK
oncogenes through over-expression or
NUCLEAR TRANSCRIPTION
mutation, leading to unregulated cell sig- APOPTOSIS
nalling and cellular proliferation. Because a DNA
number of these components may be pref-
erentially over-expressed or mutated in Signaling pathways targeted by anticancer agents. PI3K = phosphoinositide-3-kinase; PLC = phos-
human cancers, the cell signalling cascade pholipase C; PKC = protein kinase C; MEK = mitogen-activated protein kinase kinase; ERK = extracellular sig-
provides a variety of targets for anticancer nal-regulated kinase; Akt = protein kinase B (PKB); BAD = Bcl-XL/Bcl-2-associated death protein; VEGF = vas-
cular endothelial growth factor; HER = human epidermal growth factor receptor family; PDGF = platelet
therapy (Adjei AA, Current Pharmaceutical derived growth factor; FGF = fibroblast growth factor; SOS = son of sevenless guanine nucleotide exchange
Design, 6: 471-488, 2000). protein; GRB = growth factor receptor-bound protein.

Different approaches have been used to


attack these targets and include classical ing. The drug Gleevec is already in clinical > Receptors, anti-receptor antibodies and
cytotoxic agents as well as small molecule use (Leukaemia, p242). It is hoped that in tyrosine kinase receptor inhibitors
drug inhibitors. In addition, antisense future, a combination of agents targeting > RAS farnesyltransferase inhibitors
oligonucleotides, vaccines, antibodies, parallel pathways, as well as combinations > RAF inhibitors
ribozymes and gene therapy approaches with classical cytotoxic agents will improve > MEK inhibitors
have been utilized. the outcome of cancer patients. > Rapamycin analogues
Classes of agents and their potential tar- > Protein kinase C (PKC) inhibitors
The diagram illustrates cell signalling gets include: > Inhibitors of protein degradation
pathways that are targeted by anticancer > Inhibitors of ligands, such as recombinant > Inhibitors of protein trafficking
agents currently undergoing clinical test- human antibody to VEGF (rHu mAbVEGF)

Apoptosis 117
worthy is the development of caspase 4-hydroxyphenylretinamide. Butyrate, a nase enzyme (COX-2) expression may
inhibitors for the treatment of certain short-chain fatty acid produced by bacter- modulate intestinal apoptosis via changes
degenerative (non-cancerous) diseases ial fermentation of dietary fibre, inhibits in Bcl-2 expression. Aspirin and similar
characterized by excess apoptosis. cell growth in vitro and promotes differen- drugs which inhibit COX-2 may promote
Drugs shown to induce apoptosis specifi- tiation; it also induces apoptosis. Both apoptosis and prevent tumour formation.
cally include chemopreventive agents roles may contribute to its prevention of
(Chemoprevention, p151), exemplified by colorectal cancer. Moreover, cyclo-oxyge-

REFERENCES WEBSITE
1. Strasser A, O'Connor L, Dixit VM (2000) Apoptosis sig- early events that modulate caspase activation during apop- The European Cell Death Organization:
naling. Annu Rev Biochem, 69: 217-245. tosis. Nat Cell Biol, 2: 318-325. http://www.ecdo.dote.hu/
2. Kaufmann SH, Gores GJ (2000) Apoptosis in cancer: 11. Chung HT, Pae HO, Choi BM, Billiar TR, Kim YM (2001)
cause and cure. Bioessays, 22: 1007-1017. Nitric oxide as a bioregulator of apoptosis. Biochem
Biophys Res Commun, 282: 1075-1079.
3. Parton M, Dowsett M, Smith I (2001) Studies of apop-
tosis in breast cancer. BMJ, 322: 1528-1532. 12. Kumar S (1999) Regulation of caspase activation in
apoptosis: implications in pathogenesis and treatment of
4. Choisy-Rossi C, Yonish-Rouach E (1998) Apoptosis and
disease. Clin Exp Pharmacol Physiol, 26: 295-303.
the cell cycle: the p53 connection. Cell Death Differ, 5:
129-131. 13. Porter AG, Janicke RU (1999) Emerging roles of cas-
pase-3 in apoptosis. Cell Death Differ, 6: 99-104.
5. Rich T, Allen RL, Wyllie AH (2000) Defying death after
DNA damage. Nature, 407: 777-783. 14. Nicholson DW (2000) From bench to clinic with apop-
tosis-based therapeutic agents. Nature, 407: 810-816.
6. Peter ME, Krammer PH (1998) Mechanisms of CD95
(APO-1/Fas)-mediated apoptosis. Curr Opin Immunol, 10: 15. Zhou BB, Elledge SJ (2000) The DNA damage
545-551. response: putting checkpoints in perspective. Nature, 408:
433-439.
7. Yeh WC, Hakem R, Woo M, Mak TW (1999) Gene tar-
geting in the analysis of mammalian apoptosis and TNF 16. Brown JM, Wouters BG (1999) Apoptosis, p53, and
receptor superfamily signaling. Immunol Rev, 169: 283- tumor cell sensitivity to anticancer agents. Cancer Res, 59:
302. 1391-1399.
8. Griffith TS, Lynch DH (1998) TRAIL: a molecule with 17. Zheng TS (2001) Death by design: the big debut of
multiple receptors and control mechanisms. Curr Opin small molecules. Nat Cell Biol, 3: E43-E46.
Immunol, 10: 559-563. 18. Altucci L, Rossin A, Raffelsberger W, Reitmair A,
9. Gross A, McDonnell JM, Korsmeyer SJ (1999) BCL-2 Chomienne C, Gronemeyer H (2001) Retinoic acid-induced
family members and the mitochondria in apoptosis. Genes apoptosis in leukemia cells is mediated by paracrine action
Dev, 13: 1899-1911. of tumor-selective death ligand TRAIL. Nat Med, 7: 680-
686.
10. Matsuyama S, Llopis J, Deveraux QL, Tsien RY, Reed
JC (2000) Changes in intramitochondrial and cytosolic pH:

118 Mechanisms of tumour development


INVASION AND METASTASIS

synonymous with poor prognosis. Current


SUMMARY methods of detecting new tumours, Malignant tumour

> The ability of tumour cells to invade and including computed tomography (CT) Growth
colonize distant sites is a major feature scans or magnetic resonance imaging Angiogenic Viable cells
distinguishing benign growths from (MRI), ultrasound, or measurement of cir- factors
malignant cancer. Cytokines Zone of sublethal
culating markers such as carcinoembry- hypoxia
> Most human tumours lead to death through onic antigen (CEA), prostate-specific anti- Growth Necrosis
widespread metastasis rather than the gen (PSA) or cancer antigen 125 (CA125) factors
adverse local effects of the primary neo- are not sufficiently sensitive to detect
plasm. micrometastases. A greater understand- Rapid accumulation
ing of the molecular mechanisms of Inflammatory of genetic damage
> Often, metastatic spread first involves cells in sublethal zone
metastasis is required. It is clear that mediated by oxygen
regional lymph nodes, followed by
haematogenous spread throughout the
metastatic growth may reflect both gain free radicals
(NO= nitric oxide)
body. Metastases may become clinically and loss of function, and indeed the
manifest several years after surgical resec- search for metastasis suppressor genes
Reperfusion of sub-
tion of the primary tumour. has been more fruitful than identification lethal zone containing
of genes which specifically and reliably cells with varying
> Current methods are inadequate for the degrees of genetic
potentiate metastasis [1]. damage. Some have
routine detection of micrometastases and acquired a metastatic
the search for effective, selective phenotype
therapies directed toward metastatic
The genetics of metastasis
growth remains a major challenge. With the publication of the human
genome sequence, and various major ini- Metastasis
tiatives such as the Cancer Genome
Fig. 3.40 The hypoxia hypothesis suggests that
Project in the UK and the Cancer Genome the progression of malignant tumours to a
Anatomy Project in the USA, the search metastatic phenotype is mediated by deficiency of
for genes selectively upregulated, mutat- oxygen and resulting tumour necrosis.
Metastasis (from the Greek meaning
change in location) refers to growth of
secondary tumours at sites distant from a Growth Angiogenesis Decreased cell-cell adhesion
primary neoplasm. Metastasis thus dis- Protease activation Increased protease production
tinguishes benign from malignant lesions Selection Increased cell-matrix adhesion
and is the ultimate step in the multistage 1.Localized tumour 2.Breakthrough 3.Invasion
process of tumour progression.
Metastatic growth is the major cause of
treatment failure and the death of cancer
patients. Although secondary tumours
may arise by shedding of cells within body
cavities, the term metastasis is generally
reserved for the dissemination of tumour 4.Transport
cells via the blood or lymphatics. Spread
in the cerebrospinal fluid and
transcoelomic passage may also occur.
Most (60-70%) cancer patients have overt
or occult metastases at diagnosis, and
the prognosis of the majority of these
5.Lodgement 6.Extravasation 7.Metastasis
patients is poor (Box: TNM Classification
Adhesion Proliferation Angiogenesis
of Malignant Tumours, p124). Protease activation/production
Protease production
There is a critical need to identify reliable Metastasis of metastases
indicators of metastatic potential, since Fig. 3.41 The stages in the metastatic process, illustrated in relation to the spread of a primary tumour
clinical detection of metastatic spread is from a surface epithelium to the liver.

Invasion and metastasis 119


metastasis, and a great deal is known
about the cellular and molecular events
Gene Cancer type(s) Mechanism
that underlie the process. However the
ability to predict which patient has occult
micrometastases, and the discovery of
nm23 Family (H1-6) of Breast Cell migration?
nucleoside diphosphate (liver, ovary, melanoma) Signalling via G proteins, effective, selective therapies for metasta-
kinases microtubule assembly tic disease, remain major challenges in
oncology.
PTEN/MMAC1 Prostate, glioma, breast Migration, focal adhesions

KAI1/CD82/C33 Prostate, stomach, colon, Cell-cell adhesion, motility The biology of metastasis
breast, pancreas, lung Growth of tumours beyond a few millime-
tres in diameter cannot progress without
CAD1/E-cadherin Many adenocarcinomas Cell-cell adhesion,
epithelial organization neovascularization, and there is a growing
appreciation of how this phenomenon is
MKK4/SEK1 Prostate Cellular response to linked to metastasis [3]. Many genetic
stress?
changes associated with malignant pro-
KiSS-1 Melanoma, breast cancer Signal transduction? gression (mutation of HRAS, over-expres-
Regulation of MMP-9? sion of ERBB2 oncogenes, loss of p53)
induce an angiogenic phenotype (devel-
BRMS1 Breast Cell communication,
motility oping blood vessels) via induction of
cytokines, such as vascular endothelial
DPC4 Colon, pancreas ? growth factor (VEGF-A). VEGF-A is also
upregulated by hypoxia in tumours, partly
Table 3.5 Putative metastasis suppressor genes. by host cells such as macrophages. The
presence of hypoxic areas is a character-
ed or lost in metastatic cancers (Table metastasis, although many others so iden- istic of solid tumours and has been relat-
3.5) has gained momentum. It is now pos- tified are also associated with tumour ed to poor response to conventional ther-
sible, using laser capture microdissection growth or developmental processes. apies (Fig. 3.40). In addition, activation of
and serial analysis of gene expression The events which lead to cancer metasta- epithelial growth factor receptor (EGFR)
(SAGE), to isolate invasive cancer cells sis include changes in cell-cell and cell- and other oncogenic signalling pathways
and compare their gene or protein expres- matrix adhesion, alterations in cell shape, can also upregulate VEGF-C, a known
sion with non-invasive or normal cells deformability and motility, invasion of sur- lymphangiogenic cytokine [4]. The recep-
from the same patient [2]. Prior to this, rounding normal tissues, gaining access tors for these cytokines (Flk-1 and Flk-4)
transfection of chromosomes or DNA to lymphatic or vascular channels, dis- are expressed on tumour vasculature,
from metastatic to non-metastatic cells semination via blood or lymph, survival of and both (in addition to acting as potent
(or vice versa), subtractive hybridiza- host defence mechanisms, extravasation mitogens for endothelial cells) also
tion/differential display PCR, cDNA array and colonization of secondary sites (Fig. enhance vessel permeability. Thus activa-
and other strategies resulted in identifica- 3.41). There are now many features of tion of these signalling pathways may
tion of some genes specifically linked to cancer cells recognized to potentiate potentiate both vascular and lymphatic
invasion and tumour spread. Basic fibrob-
last growth factor (bFGF) is often upregu-
lated in cancers, particularly at the inva-
sive edge where tumour cells interact
with host cells [5].
Epithelial cells are normally bounded by
basement membranes which separate
them from the underlying stroma and
mesenchymal compartments. Breaching
this barrier is the first step in the transi-
tion from carcinoma in situ to invasive
and potentially metastatic carcinoma.
Basement membrane is composed of a
Fig. 3.42 Multiple metastatic growths of an intes- Fig. 3.43 Multiple metastases to the brain from a variety of structural proteins including
tinal carcinoma in the liver. lung carcinoma. collagen IV (the major component),

120 Mechanisms of tumour development


laminin, entactin and also heparan sulfate affinity can be profoundly influenced by lized during lymphocyte homing, and a
proteoglycans. Interactions of tumour the local microenvironment and soluble change from the standard epithelial pat-
cells with the basement membrane have factors, enabling the tumour cell to tern to expression of splice variants asso-
been considered to comprise three steps, respond to different conditions encoun- ciated with haematopoietic cells has
which can readily be demonstrated in tered throughout the metastatic cascade. been proposed to assist carcinoma cells
vitro: adhesion, matrix dissolution/prote- in haematogenous dissemination. Throm-
olysis and migration [6]. Other molecules involved in adhesion bospondin may mediate adhesion bet-
Epithelial cells are normally polarized and Other adhesion molecules implicated in ween circulating tumour cells, platelets
firmly attached to each other via desmo- cancer progression include selectins such and endothelial cells, promoting emboliza-
somes, tight junctions and intercellular as sialyl Lex and members of the tion (vessel obstruction) and arrest.
adhesion molecules such as E-cadherin, immunoglobulin superfamily, including Tumour cells then gain access to the sub-
and also bound to the basement mem- intercellular adhesion molecules (ICAM-1, endothelial basement membrane when
brane via other adhesion molecules ICAM-2, VECAM and PECAM). The latter endothelial cells retract in response to
including integrins. Changes in cell-cell are upregulated on activated endothelial these emboli, and can adhere to exposed
and cell-matrix adhesive interactions are cells, and can interact with integrins on proteins. Synthetic peptides containing
common in invasive cancer (Cell-cell com- leukocytes and circulating tumour cells, sequences of amino acids which compete
munication, p109). Indeed, E-cadherin assisting their arrest and extravasation. with binding to laminin or fibronectin can
may be designated a tumour suppressor CD44 is another adhesion molecule uti- inhibit colonization of the lung by intra-
gene, since its loss or functional inactiva-
tion is one of the most common charac-
teristics of metastatic cancer, and its re-
introduction into cells can reverse the
malignant phenotype. The adenomatous
polyposis coli gene (APC), which is mutat-
ed in many inherited and sporadic colon
cancers, normally regulates the expres-
sion of -catenin, a protein which inter-
acts with E-cadherin. Mutations in APC
(or -catenin) increase cellular levels of
the latter and facilitate interactions with
transcription factors such as T-cell fac-
tor/lymphoid enhancer factor (TCF/LEF)
which drive the expression of genes
involved in inhibiting apoptosis and stim-
ulating cell proliferation. Other genes
commonly lost in cancers (e.g. DCC,
Deleted in Colon Carcinoma) also encode
adhesion molecules.

Integrins
Integrins are heterodimeric proteins that
mediate adhesion between cells and the
extracellular matrix or other cellular ele-
ments. Ligand specificity is determined by
the subunit composition; many integrins
bind multiple substrates and others are
more selective. Far from being an inert
glue, they are capable of transmitting
important signals regulating cell survival,
differentiation and migration [7]. Many dif-
ferences in integrin expression between
benign and malignant cells have been doc- Fig. 3.44 MRI scan showing skeletal metastases in a patient with a primary prostatic carcinoma (front
umented, but the patterns are complex. In and back views). Some of the larger metastases are marked by arrows. Note the numerous metastases
addition, their expression and binding in the ribs and in the spine.

Invasion and metastasis 121


venously injected cells in experimental laminin and also activates gelatinases.
models. Thus, upregulation of these enzymes in
cancers leads to proteolytic cascades and
RHO potential for invasion of the basement
The RHO gene family of small GTP- membrane and stroma.
hydrolysing proteins contains several Metalloproteinases also contribute to
members known to be involved in cell tumour growth and metastasis by other
migration via regulation of actomyosin- means [9]. During angiogenesis, inva-
based cytoskeletal filament contraction sion of capillary sprouts requires local
and the turnover of sites of adhesion. proteolysis (mediated in part by upregu-
Overexpression of RhoC alone in lated MMP-2 and MMP-9 together with
melanoma cells is sufficent to induce a uPA) and in addition MMP-9 has been
highly metastatic phenotype [8]. implicated in the angiogenic switch by
releasing VEGF from sequestration in the
Enzyme functions in invasion and extracellular matrix [10]. Furthermore,
metastasis these proteases can contribute to the
Invasive tumour cells show increased sustained growth of tumours by the
expression of many enzymes due to ectodomain cleavage of membrane-
upregulation of genes, enhanced activa- bound pro-forms of growth factors, and
tion of pro-enzymes or reduced expres- the release of peptides which are mito-
sion of inhibitors such as tissue inhibitors genic and chemotactic for tumour cells.
of metalloproteinases (TIMPs). In addi-
tion, tumour cells may also induce expres- Heparanase
sion of enzymes by neighbouring host Apart from the structural proteins cleaved
cells and hijack these to potentiate by metalloproteinases in the basement
invasion. membrane and extracellular matrix, the
other major components are glycos-
Matrix metalloproteinases aminoglycans, predominantly heparan
Fig. 3.45 Location of metastases at autopsy for One important group is the matrix metal- sulfate proteoglycan (HSPG). Heparanase
some common cancers, indicating that the site of loproteinases (MMP). Different cancers is an important enzyme which degrades
metastasis is not random.
may show different patterns of expres- the heparan sulfate side-chains of HSPGs
sion; for instance squamous carcinomas and, like the proteases described above,
frequently have high levels of gelatinase B not only assists in the breakdown of extra-
(MMP-9), stromelysins 1-3 (MMP-3, cellular matrix and basement membrane,
Primary tumour Site of metastasis MMP-10 and MMP-11, normally stromal but is also involved in the regulation of
enzymes, but also expressed by these growth factor and cytokine activity. Basic
carcinomas) and matrilysin (MMP-7). fibroblast growth factor (bFGF, another
Bronchial cancer Adrenal Adenocarcinomas such as breast may potent mitogen and chemotactic factor
(often bilateral) have increased levels of gelatinase A for endothelial cells) and other heparin-
(MMP-2) and colon carcinomas common- binding growth factors are sequestered by
Breast ductal Liver ly overexpress MMP-7. In addition, MT1- heparan sulfate, providing a localized
carcinoma
MMP, which activates MMP-2, is often depot available for release by heparanase.
Breast lobular Diffuse peritoneal upregulated in tumour and/or neighbour- Similarly, uPA and tissue plasminogen
carcinoma seeding ing host tissues. The major substrate of activator (tPA) can be released from
Breast Bone, ovary the gelatinases is collagen IV, a major heparan sulfate by heparanase, further
component of the basement membrane, potentiating proteolytic and mitogenic
Lung Brain whereas the stromelysins prefer laminin, cascades.
Ocular melanoma Liver fibronectin and proteoglycans, and can
also activate procollagenase (MMP-1), Tissue-specific growth factors
Prostate Bone which in turn degrades the fibrillar colla- Finally, it is possible that release of tissue-
gens of the interstitial tissues. Urokinase specific growth factors may play a role in
Melanoma Brain
plasminogen activator (uPA) is also fre- organ selectivity of metastasis. For exam-
Table 3.6 Some sites of metastasis which are not quently upregulated in cancer. It controls ple, colorectal carcinoma cells over-
explicable by circulatory anatomy. the synthesis of plasmin, which degrades expressing EGFR have a predilection for

122 Mechanisms of tumour development


Target Example of agent Comments

Adhesion/attachment RGD-toxin constructs and RGD-targeted Have not reached clinical trials
gene therapy

Anti-avfl3 monoclonal antibody Cytostasis in patients; anti-tumour and


(Vitaxin, Medi522) anti-angiogenic in animal models

Proteolysis Matrix metalloproteinase inhibitors Cytostatic in patients; rare occurence of


tumour partial regressions; stromal fibrosis;
activity seen in multiple animal models and
in combination with chemotherapy; new agents
with varied MMP specificity under development
Motility No selective agents
Signal pathways Squalamine (NHE-3 inhibitor) Selective for endothelial cells

PDGFR, KDR and EGFR small molecule Active in vitro in animal models; preclinical
inhibitors activity in combinations; phase I trials
completed for several agents, some tumour
stabilization or regression

Anti-EGFR monoclonal antibody Neutralizing antibody; active in vitro in


(C225) animal models; phase I trials ongoing

Anti-VEGF antibody Blocking antibody; active in vitro in animal


models; preclinical activity alone and in
combination; phase I-III trials ongoing

CAI (non-voltage-gated Ca++ uptake Active in vitro in animal models; preclinical


inhibitor) activity in combinations; phase I trials of single
agents and combinations, some tumour
stabilization or regression

Extracellular matrix Pirfenidone Suppresses stromal/inflammatory cell


Remodelling by stromal expression of TGF-
Phase I trials for pulmonary fibrosis

Table 3.7 Therapeutic agents directed towards stroma-tumour interactions.

growth in the liver where there are high transforming growth factor alpha, epider- nating cells. Most metastases occur in
concentrations of its ligands. All of these mal growth factor and platelet-derived the first capillary bed or lymph node
require proteolytic cleavage for activation. growth factor, can induce chemotactic encountered. The number of involved
Other enzymes which have been implicat- responses in tumour cells expressing the nodes is a key prognostic factor for many
ed in metastasis include the cysteine pro- cognate receptors. Scatter factor (also cancers, and this has led to efforts to
teinases, notably cathepsins B and D. For known as hepatocyte growth factor, HGF) identify sentinel lymph nodes in order
most of the enzymes described, there are is a potent host-derived motility factor, to improve predictions of cancer spread.
active research programmes seeking and tumour cells themselves secrete a Lymphatic channels present less of a
selective inhibitors (some of which have variety of autocrine motility factors includ- challenge to tumour cell entry than capil-
reached phase II and III clinical trials) to ing autotaxin and neuroleukin/phospho- laries since they have scanty basement
prevent or treat metastatic disease. hexose isomerase. membrane. Once in the lymphatics,
Motility, coupled with proteolysis, is the tumour cells are carried to the subcapsu-
basis of tumour cell invasion, and is also Organ preference of metastases lar sinus of draining nodes, where they
important during intravasation and The organ distribution of metastases may arrest and grow, succumb to host
extravasation of blood and lymphatic ves- depends on the type and location of the defences, or leave the node via the effer-
sels. Many motility factors have been primary tumour, with 50-60% of the sec- ent lymphatics. The propensity for a
described which may be tumour- or host- ondary sites being dictated by the tumour cell to generate a lymphatic
derived. Many growth factors, such as anatomical route followed by the dissemi- metastasis may depend upon its ability to

Invasion and metastasis 123


TNM CLASSIFICATION OF T = primary tumour
MALIGNANT TUMOURS TX Primary tumour cannot be assessed
T0 No evidence of primary tumour
Tis Carcinoma in situ
The TNM system for the classification of T1 Tumour invades submucosa
malignant tumours (http://tnm.uicc.org/) T2 Tumour invades muscularis propria
is a form of clinical shorthand used to T3 Tumour invades through muscularis propria into subserosa or into non-
describe the anatomic extent (staging) of peritonealized pericolic or perirectal tissues
a cancer in terms of: T4 Tumour directly invades other organs or structures and/or
T - the primary tumour perforates visceral peritoneum
N - regional lymph nodes
M - distant metastases N = regional lymph nodes
NX Regional lymph nodes cannot be assessed
These components are given a number
N0 No regional lymph node metastasis
that reflects the absence or presence and
N1 Metastasis in 1 to 3 regional lymph nodes
extent of the disease. For example, a N2 Metastasis in 4 or more regional lymph nodes
tumour of the colon that is classified as
T2N1M0 would have extended into the M = distant metastasis
colons muscular wall, spread to 1 to 3 MX Distant metastasis cannot be assessed
regional lymph nodes but without evi- M0 No distant metastasis
dence of distant metastasis. Evaluation M1 Distant metastasis
by the TNM system can therefore help in
Table 3.8 TNM classification of cancer of the colon and rectum.
the planning of treatment by the oncolo-
gist and in monitoring the efficacy of this
treatment, as well as giving some indica- single international TNM classification Uniform Use, 2nd Edition, Wiley 2001)
tion of prognosis. Moreover, the use of a should be formulated. This is achieved via with rules and explanations.
standardized system facilitates the dis- meetings of experts that update existing The challenge for the future is the incor-
semination of information in the clinical classifications, as well as develop new poration into TNM of information from
community. ones. The present TNM edition (Eds. Sobin new diagnostic and imaging technologies
The TNM system was developed by Pierre LH and Wittekind Ch, TNM Classification of (such as endoscopic ultrasound, magnet-
Denoix (President of the UICC, 1973-1978) Malignant Tumours, 6th Edition, Wiley, ic resonance imaging, sentinel node bio-
between 1943 and 1952 (Sobin LH, TNM 2002) contains guidelines for classification psy, immunohistochemistry and poly-
principles, history and relation to other and staging that correspond exactly with merase chain reaction). There is an
prognostic factors. Cancer, 91:1589-92, those of the 6th edition of the AJCC Cancer expanding array of known and potential
2001). In 1968, a series of brochures pub- Staging Manual (2002). TNM, now the most prognostic factors (Eds. Gospodarowicz M
lished by UICC describing the classifica- widely used system to classify tumour et al., Prognostic Factors in Cancer, Wiley,
tion of cancers at 23 body sites were com- spread, is published in 12 languages and is 2001) with which TNM could be integrat-
bined to produce the Livre de Poche, accompanied by an illustrated TNM Atlas ed to form a comprehensive prognostic
which has been subject to regular re-edi- (Eds. Hermanek P et al., 4th Edition, system. Such integration could potential-
tion, enlargement and revision over sub- Springer-Verlag, 1997), a TNM Mobile ly be exploited to enhance the prediction
sequent years. In order to prevent Edition (Wiley, 2002) and a TNM supple- of prognosis, and individualize cancer
unwanted variations in the classification ment (Eds. Wittekind Ch et al., TNM patient treatment.
by its users, in 1982 it was agreed that a Supplement 2001. A Commentary on

adhere to reticular fibres in the subcapsu- there; colon carcinoma cells enter the that specific cancer cells (the seed) had
lar sinus. These fibres contain laminin, portal circulation which delivers cells to an affinity for certain organs (the soil).
fibronectin and collagen IV, and different the liver, and so on (Fig. 3.45). However, a In experimental systems there are many
integrins expressed by different tumour non-random element in metastatic pat- examples showing that primary tumours
cells may be responsible for adhesion to terns has long been recognized (Table are heterogeneous, and that cloned cells
these structures and to the lymphatic 3.6). Stephen Paget developed the seed can vary in their ability to metastasize to
endothelial cells [11]. and soil hypothesis in 1889, based on his different sites. Some of the patterns
Sarcomas tend to metastasize to lungs observations from autopsies of over 700 relate to the ability of malignant cells to
because the venous drainage returns women with breast cancer. He proposed adhere to the endothelial cells in target

124 Mechanisms of tumour development


organs, and to respond to local growth stages, both in experimental models and chain reaction (PCR), individual tumour
factors once they have extravasated. It in man, many tumour cells reach distant cells (or specific genetic markers) can be
used to be thought that escape from the sites but may remain dormant, either due found in blood, nodes, bone marrow, body
primary tumour and survival in the circu- to lack of appropriate growth factors, or fluids etc, but the significance of posi-
lation were the major rate-limiting steps their failure to induce neoangiogenesis. tive results, and whether they can be
for successful metastasis. However, while Indeed, using sensitive assays such as used to predict subsequent overt metas-
there is a good deal of attrition at these immunocytochemistry and polymerase tases is not yet established.

REFERENCES WEBSITE
1. Yoshida BA, Sokoloff MM, Welch DR, Rinker-Schaeffer 7. Berman AE, Kozlova NI (2000) Integrins: structure and The Metastasis Research Society (UK):
CW (2000) Metastasis-suppressor genes: a review and per- functions. Membr Cell Biol, 13: 207-244. http://www.metastasis.icr.ac.uk
spective on an emerging field. J Natl Cancer Inst, 92: 1717-
1730. 8. Ridley A (2000) Molecular switches in metastasis.
Nature, 406: 466-467.
2. Simone NL, Paweletz CP, Charboneau L, Petricoin EF,
Liotta LA (2000) Laser capture microdissection: Beyond 9. McCawley LJ, Matrisian LM (2000) Matrix metallopro-
functional genomics to proteomics. Mol Diagn, 5: 301-307. teinases: multifunctional contributors to tumor progres-
3. Fidler IJ (2000) Angiogenesis and cancer metastasis. sion. Mol Med Today, 6: 149-156.
Cancer J Sci Am, 6 Suppl 2: S134-S141. 10. Bergers G, Brekken R, McMahon G, Vu TH, Itoh T,
4. Eccles SA (2000) Cell biology of lymphatic metastasis. Tamaki K, Tanzawa K, Thorpe P, Itohara S, Werb Z, Hanahan
The potential role of c-erbB oncogene signalling. Recent D (2000) Matrix metalloproteinase-9 triggers the angio-
Results Cancer Res, 157: 41-54. genic switch during carcinogenesis. Nat Cell Biol, 2: 737-
5. Fidler IJ (1999) Critical determinants of cancer metas- 744.
tasis: rationale for therapy. Cancer Chemother Pharmacol,
43 Suppl: S3-10. 11. Brodt P (1991) Adhesion mechanisms in lymphatic
metastasis. Cancer Metastasis Rev, 10: 23-32.
6. Stracke ML, Liotta LA (1992) Multi-step cascade of
tumor cell metastasis. In Vivo, 6: 309-316.

Invasion and metastasis 125

You might also like