Sie sind auf Seite 1von 29

THE YEAR IN EVOLUTIONARY BIOLOGY 2009

Evolutionary Theory of Cancer


Camille Stephan-Otto Attolini and Franziska Michor
Computational Biology Program, Memorial Sloan-Kettering Cancer Center, New York,
New York, USA

As Theodosius Dobzhansky famously noted in 1973, “Nothing in biology makes sense ex-
cept in the light of evolution,” and cancer is no exception to this rule. Our understanding
of cancer initiation, progression, treatment, and resistance has advanced considerably
by regarding cancer as the product of evolutionary processes. Here we review the litera-
ture of mathematical models of cancer evolution and provide a synthesis and discussion
of the field.

Key words: evolution; cancer; mathematical modeling

Introduction 1983; Komarova & Wodarz 2005; Michor et al.


2006b) (Fig. 1).
The use of mathematics in the study of med- Cancer results from evolutionary processes
ical conditions has a long history. The earli- occurring within the body (Nowell 1976). Since
est work dates back to the 8th century, when evolution describes the temporal changes of
Al-Kindi attempted to apply mathematics to a population of individuals due to variation
pharmacological questions such as describing and selection, the concept is highly relevant
the strength of drugs (Prioreschi 2002). In the to neoplasia. Tumors can be viewed from
18th century, Bernoulli analyzed the morbid- an evolutionary standpoint as collections of
ity and mortality of smallpox and demon- cells that accumulate genetic and epigenetic
strated the efficacy of vaccination (Bernoulli & changes, which are then subjected to the selec-
Blower 2004). The interaction between mathe- tion pressures within a tissue. These normally
matics and medicine has proven beneficial heritable variations can lead to adaptations
for understanding the underlying biology and of the cells such as induction of angio-
for designing treatments and diagnosing dis- genesis or evasion of the immune system.
eases. Due to its clinical importance, cancer Beneficial heritable changes can cause rapid
has been of particular interest to theoretical expansion of the mutant clone since they en-
investigators. Since the early 1940s (Charles & able their carriers to outcompete cells that have
Luce-Clausen 1942), mathematical approaches not accumulated similar improvements. Muta-
have been developed to explain regularities tions advantageous to the cancer cell are nor-
seen in incidence data (Armitage & Doll 1957; mally detrimental to the organism, ultimately
Fisher 1958; Knudson 2001), hereditary pre- causing death of both the patient and the
disposition to disease (Knudson 1986; Frank tumor. Therefore, neoplastic processes serve
2007), cancer progression (Tomlinson et al. as an example for selection acting on dif-
1996; Desper et al. 1999; Michor et al. 2006c), ferent hierarchical levels (Buss 1987): clonal
and response to treatment (Goldie & Coldman evolution generally selects for increased pro-
liferation, survival, and evolvability on the
cellular level and leads to progression, inva-
sion, and resistance; the latter effects are se-
Address for correspondence: Franziska Michor, Computational Biol-
ogy Program, Memorial Sloan-Kettering Cancer Center, New York, NY
lected against on the level of multicellular
10065. Voice: 646-888-2802; fax: 646-422-0717. michorf@mskcc.org organisms.

The Year in Evolutionary Biology 2009: Ann. N.Y. Acad. Sci. 1168: 23–51 (2009).
doi: 10.1111/j.1749-6632.2009.04880.x  c 2009 New York Academy of Sciences.

23
24 Annals of the New York Academy of Sciences

Figure 1. Contributions to an evolutionary theory of cancer. We show examples of theoretical approaches


to cancer initiation (black outline), genetic instabilities (dark gray), progression (gray), and resistance (light
gray). Online version: cancer initiation (purple), genetic instabilities (green), progression (dark blue), and
resistance (light blue).

The investigation of cancer evolution re- Cancer Initiation


quires mechanistic, quantitative models that in-
corporate realistic properties of biological sys- An understanding of the mechanisms of can-
tems such as stochasticity and nonlinearity. As cer initiation has straightforward implications
the outcomes of such interactions cannot be de- for prevention, diagnosis, and treatment of the
termined by verbal reasoning alone, they must disease. A genetic cause of cancer was first pro-
be computed from general integrative mod- posed by Boveri in 1914 (Boveri 1914). Charles
els of carcinogenesis (Gatenby & Maini 2003; & Luce-Clausen (1942) presented one of the
Michor et al. 2004a; Merlo et al. 2006). Theo- earliest mathematical models of cancer. They
retical approaches to tumorigenesis have led to studied the incidence of skin carcinomas in
considerable insights into the natural history of mice painted with a carcinogen, finding a lin-
the disease and have begun to transform cancer ear relationship between the square root of the
research into a rational and predictive science. number of tumors and the time since the first
In this review we present a historical and topical painting. Their model was based on the as-
view of mathematical models of cancer evolu- sumption that each application of the carcino-
tion. We center our attention on several areas gen causes a certain number of cells to acquire
of interest in the analysis of cancer—initiation, mutations, but that these cells initiate abnor-
progression, genomic instabilities, differentia- mal growth only once both alleles of a partic-
tion and heterogeneity, and drug resistance— ular gene have been mutated. The predictions
and provide a synthesis of the field. of the model were in good agreement with the
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 25

experimental data, reinforcing the notion that


cancer might have a genetic cause and, in par-
ticular, could be explained by the accumulation
of two mutations.
In the first half of the last century, the main
source of cancer-related data was age-specific
incidence. For most cancer types in adults, the
number of cases per age class increases with a
high power of age. Fisher & Hollomon (1951)
presented a multicellular model in which mu-
tations were assumed to occur in different cells
within the same cell population and only the
combination of all mutants led to cancer. De-
spite the dismissal of this model as being unable
to convincingly explain tumorigenesis, the im-
portance of cooperation and co-evolution of
different cell types in tumors, the surrounding
stroma, and the immune system has recently
been highlighted (Vincent & Gatenby 2008).
As an alternative to the multicellular hypoth-
esis, Nordling suggested in 1953 that muta-
tions must occur sequentially in the same cell
for transformation into a neoplastic phenotype
(Nordling 1953). He noticed that when cancer
incidence data are plotted on a doubly logarith-
mic plane, the resulting curve is a straight line
with a slope close to six. He hypothesized that
in general, cancer data with a slope of n can
be explained by n + 1 hits (Fig. 2A). In 1957,
Armitage and Doll presented a more detailed

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
time (Knudson 1971). He concluded that a single so-
Figure 2. The study of age-specific incidence matic mutation is sufficient to cause cancer in hered-
data of human cancers. (A) Age at death from var- itary cases and two somatic mutations are neces-
ious epithelial cancers. The plots show the number sary for the sporadic cases. Figure adapted from
of deaths of male patients per 100,000 versus the Weinberg (2007). (C) Colon cancer incidence. The
age at death for different countries on a doubly log- panel shows the numerical simulation of equation (1)
arithmic plane. According to Nordling (1953), the from Liso et al. (2008) and the adjusted cumulative
slopes of the curves indicate that the accumulation of colon cancer incidence; data were downloaded from
five or six rate-limiting events is required to cause SEER (www.seer.cancer.gov) and adjustments were
a lethal cancer; the slopes differ slightly between performed as in Table 1 of Michor et al. (2005a).
cancer types (data not shown). Figure adapted from Parameter values are mutation rate 5 × 10−4 , initial
Weinberg (2007). (B) Kinetics of sporadic and hered- population size (i.e., number of stem cells per colonic
itary retinoblastoma. Knudson studied the retinoblas- crypt) 10, carrying capacity (i.e., final population
toma incidence in children and found that in heredi- size after clonal expansion) 1013 , fitness advantage
tary cases, the percentage of cases not yet diagnosed of mutated cells 1%, average time between stem cell
decreases linearly with age while in sporadic divisions 10 days, and probability of diagnosis per
cases, this percentage decreases quadratically with mutated cell 10−10 .
26 Annals of the New York Academy of Sciences

mathematical model of the successive accumu- mathematical model based on a comparison


lation of mutations (Armitage & Doll 1957). of incidence data of sporadic and hereditary
They allowed for different mutation rates at cancer (Ashley 1969). He studied the differ-
different ages and assumed a fixed order in the ences between colorectal cancer patients with
sequence of mutations. The model was tested and without Familial Adenomatous Polyposis
with age-specific mortality curves of 17 types (FAP), a hereditary condition now known to
of cancer and for patients between ages 25 and be caused by a germline mutation in the APC
74. For a subset of cancer types, the accumula- tumor-suppressor gene, which results in hun-
tion of seven mutations before diagnosis seemed dreds to thousands of polyps in the colons of
to agree well with the observed data. Never- teenage carriers. Ashley proposed that some
theless, they noticed that a second subset did of the genetic changes leading to cancer could
not fit the model assuming n +1 hits; the au- cause increased ratios of cell division to cell
thors attributed this deviation to the influence death. Under this assumption, a slope of n of
of external factors such as exposure to carcino- the incidence curve could result from either n
gens or the influence of endocrine secretions. or n − 1 hits since the accelerated net growth
These considerations led to the formulation of rate reduces the number of necessary muta-
two distinct mathematical descriptions of can- tions. When comparing the incidence data of
cer evolution—one assuming constant muta- FAP with that of sporadic colorectal cancer,
tion rates and the other considering variable the difference in the slopes was found to be
rates of mutation depending on age, sex, and approximately two for both men and women.
the site of the disease (Armitage & Doll 1957). Ashley concluded that a mutation in the APC
These authors’ hypothesis that cancer results gene accounts for a slope of two or three in the
from the accumulation of multiple mutations incidence curve of colorectal cancer, depend-
in the same cell eventually became known as ing on whether clonal expansion is considered.
the multistage theory of carcinogenesis. Knudson’s investigation in 1971 then led to the
Fisher (1958) suggested that the incidence identification of tumor-suppressor genes by the-
curves of most cancers can be explained by oretical techniques and became known as the
the accumulation of only three mutations. Ac- “two hit” hypothesis of tumor-suppressor inac-
cording to his approach, the slope of the in- tivation (Knudson 1971). The model was de-
cidence curve, s, is proportional to a function veloped from the observation that retinoblas-
of the number of mutations, m, and the age toma, a childhood eye cancer, presents in a
t at diagnosis, t 3(m−1) . This formula was de- hereditary unilateral version and a sporadic bi-
rived from a modified version of Armitage and lateral type. While hereditary cases accumulate
Doll’s model that included the possibility of linearly with age, sporadic cases increase with
early mutations leading to clonal expansion, the second order of age in frequency (Fig. 2B).
thereby altering their epidemiological conse- Knudson developed a statistical model predict-
quences. Fisher found that if the radius of the ing that two somatic mutations in an “anti-
area covered by the mutant clone grows at a oncogene,” as he called it, cause the sporadic
constant rate, sequential waves of faster than cases, while a single somatic mutation com-
linear growth are expected. In the context of bined with a germline mutation results in the
this model, a slope of six in the incidence curve hereditary cases. The gene later identified to
is caused by three genetic changes, each fol- cause retinoblastoma if inactivated in both al-
lowed by a quadratic increase in the population leles, RB1, became known as the first tumor-
size of the tumor. suppressor gene (Friend et al. 1986).
The next steps in the development of theories Inspired by Knudson’s work in the 1970s,
of cancer incidence were taken by Ashley (1969) several investigations into the predisposition for
and Knudson (1971). Ashley developed the first and inheritance of cancer were initiated (Frank
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 27

2004a). Nunney (2003) used a probabilistic both copies of the gene, while in intermediately
population genetics model of multistage car- large populations, a tumor-suppressor gene is
cinogenesis to arrive at three conclusions con- inactivated in a single rate-limiting hit (since
cerning the inheritance of cancer-related muta- the rates of these hits limit the evolutionary
tions: lethal or sterilizing cancers are caused by dynamics, they are called rate-limiting). These
alteration of more than one gene, with the ex- kinetics are due to a cell with two inactivated
ception of retinoblastoma; cancers that occur alleles taking over the population before a cell
in prereproductive ages infrequently have an with one inactivated allele reaches fixation; this
inherited predisposition; and cancers depend- phenomenon also emerges in different scenar-
ing on the mutation of several genes appear ios and has been called “stochastic tunneling”
with increased incidence late in life because (Iwasa et al. 2004). In a large population of cells,
these cancers do not decrease reproductive fit- it takes zero rate-limiting hits to accumulate the
ness. Frank (2004b) developed a computational two mutations because the magnitude of the po-
model to show that the larger the number of pulation size enables mutations to emerge
mutations needed to cause cancer is, the smaller rapidly. Therefore, a tumor-suppressor gene
the mortality rates in the population are; nev- is inactivated with different kinetics depend-
ertheless, the initial mutations in the process ing on the number of cells in the population,
of carcinogenesis are easily accumulated be- and the epidemiological implications are not
cause reproductive selection cannot decrease straightforward.
their frequency. Frank (2005) presented a math- Further, evolutionary models investigating
ematical model that accounts for inherited mu- the incidence of leukemias have shown that a
tations accelerating the rate of cancer initia- single mutation is sufficient to explain incidence
tion. He studied the age incidence of hereditary curves with slopes of up to four (Michor et al.
and sporadic colon cancer as well as retinoblas- 2006a; Liso et al. 2008). A stochastic model of
toma, arriving at the same conclusions as Ash- cancer initiation and diagnosis leads to three
ley and Knudson: since cancer progression is waiting times, the magnitude of which deter-
driven by the accumulation of genetic changes, mines the slope of the incidence curve: (i) the
it is expected that cancers in families with in- waiting time until the production of the first
herited genetic aberrations will progress faster successful mutant cell, (ii) the time for clonal
through the different stages of carcinogenesis expansion of its lineage, and (iii) the waiting
than cancers in families with no inherited al- time until diagnosis of the disease (Michor et al.
terations. He emphasized that mutations that 2006a). If the fitness advantage of the mutant
are recessive at the cellular level can neverthe- cell is small and the population size of the tis-
less be inherited in a dominant fashion. sue and/or the carrying capacity of the tumor
Fisher’s work, together with papers pub- are large, then a one-mutation model can ex-
lished in recent years, demonstrated that the plain incidence curves with large slopes. Hence
number of genetic changes necessary to cause a large slope of cancer incidence data may re-
cancer cannot simply be read off the age- sult from a small number of mutations together
specific incidence curve. In an effort to study with clonal expansion of mutant lineages and
the dynamics of tumor-suppressor gene inac- the probabilistic process of diagnosis.
tivation, Nowak et al. (2004) designed a pop- Despite the enormous importance of early
ulation genetics model and investigated how work for establishing the multistage hypothesis
the kinetics of mutation accumulation depends of carcinogenesis, interpretations of cancer in-
on the number of cells in a compartment and cidence curves must be done with careful con-
the cellular mutation rates. The authors found sideration of the population genetics of muta-
three different laws: in small populations of tions accumulating in tissues. To demonstrate
cells, it takes two rate-limiting hits to inactivate the inability of cancer incidence data alone to
28 Annals of the New York Academy of Sciences

inform about the number of mutations nec- only in the field of cancer initiation, but also
essary to cause cancer, we fit the age-specific in progression and its implications in treatment
incidence of colorectal cancer with a stochas- and resistance. Several experimental investiga-
tic model assuming that a single mutation is tions were initiated to study mutator pheno-
sufficient to cause invasive cancer (model from types, leading to the characterization of two
Liso et al. 2008) (Fig. 2C). For reasonable pa- main types of genetic instabilities (Lengauer
rameter values, this model can explain the data et al. 1998) (Fig. 3A): while chromosomal in-
even though it has been demonstrated that sev- stability (CIN) leads to increased rates of los-
eral genetic changes must be accumulated for ing (parts of) chromosomes triggered by genetic
colorectal cancer to arise (Fearon & Vogelstein alteration of so-called CIN genes (Kolodner
1990). Therefore, the implications of the slope et al. 2002), microsatellite instability (MIN) re-
of incidence data must be re-evaluated, and sults in elevated point-mutation rates due to
final proof of the number of mutations neces- a deficiency in the mismatch repair (MMR)
sary to cause a particular cancer must come pathway (Kinzler & Vogelstein 1996; Perucho
either from experimental evidence or from a 1996). CIN can emerge due to one dominant
detailed knowledge of the effects of particular mutation, whereas MIN requires the accumu-
mutations together with mathematical model- lation of two recessive genetic changes (Fig. 3B).
ing. Additionally, as the investigations into the Many mathematical models have been
genetic causes of cancer advanced (Vogelstein developed after Loeb’s initial publication that
& Kinzler 2002), other questions started to either support or reject the hypothesis that mu-
emerge. tator phenotypes are necessary for carcinogen-
esis (Tomlinson et al. 1996; Herrero-Jimenez
The Mutator Phenotype in Cancer et al. 2000; Luebeck & Moolgavkar 2002;
Initiation and Progression Komarova et al. 2003; Michor et al. 2003a,
2004b; Komarova & Wodarz 2004). Tomlinson
In 1991, Loeb presented mathematical evi- et al. (1996) published one of the first responses
dence showing that a moderately large number to Loeb’s hypothesis, in which they showed that
of mutations cannot be accumulated in the life- normal mutation rates can indeed produce all
time of an individual under the assumption of genetic changes necessary for carcinogenesis
normal mutation rates (Loeb 1991). By multi- if fitness effects of mutations are considered.
plying the baseline mutation rate by the num- By means of computer simulations based on
ber of cell divisions occurring in a tissue, he a simple model of cell birth and death, they
showed that hundreds to thousands of cancer demonstrated that in situations in which two
cells are generated within a tissue if one or two mutations must be accumulated for cancer ini-
mutations are sufficient for carcinogenesis. If tiation, it is more likely to mutate genes that
the accumulation of more than two mutations confer a fitness advantage to the cell than to
is necessary to cause cancer, however, then a additionally accumulate a mutation leading to
tumor could not arise within the lifetime of an a mutator phenotype. From this observation,
individual assuming normal mutation rates. As the authors concluded that as long as mutations
possible solutions to this problem, Loeb pro- result in a growth advantage of tumor cells, ge-
posed mutagenic hotspots, mutational events netic instability is not the main driving force
affecting more than one gene (such as chro- of tumorigenesis—the concept of selection is
mosome amplifications and deletions), the ac- sufficient to explain the onset of cancer. Never-
tivity of carcinogens, mutations conferring a theless, as the number of mutations needed for
growth advantage to the cell, and, most im- initiating clonal expansion increases, the im-
portantly, a mutator phenotype. The latter hy- portance of genetic instability is enhanced. In
pothesis has since caused an intense debate not particular, if six neutral mutations are needed to
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 29

Figure 3. The mutator phenotype in cancer evolution. (A) Examples of mitotic abnormalities in cancer
cells. Left, aneuploidy in colorectal cancer cells. Chromosomes have been painted with chromosome-specific
hybridization probes, allowing the inspection of chromosome structure and function. Center, abnormal mitotic
spindles in oral cancer cells. This cell was stained for kinetochore regions of chromosomes (gray), spindle
microtubules (white) and chromosomal DNA, revealing an aberrant tetrapolar spindle (arrows). Right, am-
plification of centrosomes in prostate cancer. This cell contains six pericentrin-containing centrosomes (white
dots), which nucleate multiple microtubules (gray). Figure from Jallepalli & Lengauer (2001). Online ver-
sion: kinetochore regions of chromosomes (red), spindle microtubules (yellow), chromosomal DNA (green).
Right, pericentrin-containing centrosomes (yellow), microtubules (green). (B) Pathways to genetic instability.
Different types of genetic instability require different numbers of mutational hits to produce the respective
instability phenotype. Top, in a heterozygote with one defective MMR (mismatch repair) allele (step 1), all
that is required to begin to develop mutations at a high rate (microsatellite instability, MIN) is the inactivation
of the normal allele (step 2). Bottom, chromosomal instability (CIN) has a dominant quality because a single
hit (in a gene such as hBUB1, a component of the mitotic spindle checkpoint (MSC)) is sufficient to produce
the CIN phenotype. (C) The role of CIN in tumorigenesis. The early steps of colon cancer initiation occur in
small crypts, each of which is replenished by a small number of stem cells. If the effective population size of
a crypt (i.e., the number of stem cells) is small, then there is a high probability that crypts contain cells of
only one type at any time. Then a stochastic process can be designed that describes the evolution of the stem
cell population toward a more malignant phenotype (by inactivation of the APC tumor suppressor, top row).
At any time, a mutation leading to a mutator phenotype (chromosomal instability, CIN, vertical arrows) can
emerge. Stochastic tunnels emerge if an intermediate cell type does not reach fixation (gray arrows). This
stochastic process can be used to evaluate the chance that CIN emerges before inactivation of APC. Figure
adapted from Nowak et al. (2002). Online version: Stochastic tunnels emerge if an intermediate cell type
does not reach fixation (blue arrows).
30 Annals of the New York Academy of Sciences

initiate a neoplasm, then mutation rates must of parameters, a CIN or MIN mutation likely
be increased by four orders of magnitude in precedes the homozygous inactivation of APC.
order for a tumor to emerge within a human This effect is particularly pronounced if the rate
life span (Tomlinson et al. 1996). The effects of triggering genetic instability is large and the
of neutral mutations on cancer progression are selective cost of such a phenotype is low. A sub-
further discussed below. sequent paper investigated the rate of chromo-
Herrero-Jimenez et al. (2000) presented some loss optimal for tumor growth that is initi-
a statistical analysis of colorectal cancer in- ated by inactivation of tumor-suppressor genes
cidence data accounting for demographic (Komarova & Wodarz 2004). Evolutionary the-
stochasticity, that is, heterogeneity in the pa- ory predicts that higher mutation rates acceler-
tient population due to inherited traits and ate the rate of evolution but can lead to an error
environmental exposures. The authors fit a catastrophe if the rate exceeds a certain thresh-
two-stage initiation–promotion model to the old (Eigen & Schuster 1977). It is therefore in-
cancer incidence data and calculated adenoma teresting to perform a cost–benefit analysis of
growth rates, the number of mutations nec- large mutation rates leading to the advantages
essary for cancer initiation, and the rate of of inactivating tumor-suppressor genes as well
chromosome loss; the latter was found to be as the disadvantages of chromosomal losses.
significantly higher than in normal cells, sup- The stochastic approach taken by Komarova
porting the hypothesis of mutator phenotypes. and Wodarz (Komarova & Wodarz 2004) indi-
In contrast to the findings of those authors, cates that tumor initiation and progression are
a similar initiation–promotion model in the optimized if the rate of chromosome loss is of
hands of Luebeck and Moolgavkar (2002) led the order of 10−2 to 10−3 per cell division—a
to the conclusion that genetic instability is not value that coincides with experimentally deter-
necessary for fitting the incidence curve of colo- mined rates in CIN cell lines (Lengauer et al.
rectal cancer. They found that two rare events 1997). Subsequently, Michor and colleagues
(interpreted as the inactivation of both copies of presented a model in which the spatial arrange-
APC) followed by a high-frequency event (ac- ment of cells was explicitly considered (Michor
cumulation of another mutation driving can- et al. 2004b). Colorectal stem cells were as-
cer progression) are sufficient to explain the sumed to give rise to independent lineages of
incidence data. However, a scenario in which differentiating cells, which, after undergoing a
one of the rare early events is accounted for certain number of cell divisions, are shed into
by a mutation causing genetic instability, which the gut lumen. By considering mutations in the
then accelerates the rate of APC inactivation APC tumor-suppressor gene and in genes caus-
and progression to cancer, cannot be excluded. ing chromosomal instability, it was found that
In 2002, a stochastic population genetics the presence of a few genes of the latter type is
model was used to propose that the emergence sufficient to ensure that the emergence of chro-
of genetic instability is an early event in tu- mosomal instability precedes the inactivation
mors, such as colon cancer, initiated by the in- of APC. This finding showed that the hierar-
activation of a tumor-suppressor gene (Nowak chical structure of colonic crypts reinforces the
et al. 2002) (Fig. 3C). This paper led to other hypothesis of the mutator phenotype, since the
investigations of particular situations in tumori- organization of colorectal stem cells into small
genesis. Komarova et al. (2003) designed a compartments reduces the protective effect of
mathematical model of the mutation-selection negative selection against genes causing genetic
network consisting of the tumor suppressor instabilities (Michor et al. 2003a).
APC as well as genes causing chromosomal At around the same time, several papers were
and microsatellite instabilities, CIN and MIN. published that investigated the importance and
The authors found that within a broad range effect of mutator phenotypes in the progression
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 31

of low-grade tumors to more aggressive and tions such as the number of cell divisions that
invasive cancers. Solé and Deisboeck (2004) have occurred in a tumor, the number of dom-
addressed the question of the existence of a mu- inant and recessive genes conferring a fitness
tation threshold in cancer by utilizing the qua- disadvantage, and the percentage of genomic
sispecies model of Eigen and Schuster (1977). mutations affecting them, as well as mutation
Under the assumption that a mutator pheno- rates and the number of genes present in the
type exists only in a subpopulation of cells, they genome. The authors proposed that although
concluded that a limited amount of genetic in- deleterious mutations do indeed occur, the ef-
stability is advantageous for a cell clone; if the fect of negative selection is negligible since
mutation rate surpasses a threshold, the repli- disadvantageous mutants are lost from the can-
cation rate is reduced and eventually drives cer cell population while the main clone con-
the clone to extinction. The authors proposed tinues to proliferate. However, other authors
that cancer cell populations tend to maxi- have suggested that most of the genes in the
mize both mutation and replication rates. Later human genome function to constrain cellu-
on, Brumer et al. (2006) used a similar ap- lar growth and coordinate differentiation path-
proach to study error thresholds in situations ways (Rajagopalan et al. 2003). It might be pos-
in which both CIN and MIN are present. The sible, they argued, that most genetic changes
authors analyzed the semiconservative quasi- arising during tumorigenesis are beneficial to a
species model of such tumors and, considering neoplastic cell. Finally, in 2007, Enderling and
the role of postmethylation DNA repair in tu- co-workers used a system of differential equa-
mor cells, found that CIN and MIN tumors tions to emphasize the necessity of genetic in-
are individually viable, while a cell containing stability for driving cancer progression unless
both types of instability cannot survive. This the presence of a large number of stem cells
study of error thresholds in different situations and/or tumor-suppressor genes in the genome
provided an explanation for experimental find- is postulated (Enderling et al. 2007).
ings that CIN and MIN are mutually exclusive By now it has become generally accepted
(Lengauer et al. 1998). that genetic instabilities play an important role
Michor and colleagues (2005b) extended in the initiation and progression of cancers.
their investigation of colorectal cancer to ex- The widespread presence and clinical effects
plore the role of genetic instability during the of instabilities lead to questions about phar-
accumulation of all mutations considered nec- macological strategies that may be used to
essary to cause invasive cancer. They found exploit this cancer trait to the advantage of
that the conditions for early instabilities are met patients. For example, genomically unstable tu-
even more easily when a larger number of mu- mors could be treated with agents that inflict
tations are considered, since the cost of accu- further DNA damage on cells (such as alkylat-
mulating a mutation causing a mutator pheno- ing drugs) that serve to push the tumor cells
type is balanced by the larger benefit of that across the error threshold. However, in ge-
phenotype accelerating the inactivation of ev- netically stable tumors the amount of DNA
ery successive tumor-suppressor gene. damage incurred might be tolerable and hence
One argument against the hypothesis that could potentially accelerate cancer progression
mutator phenotypes are necessary for tumori- and the evolution of resistance. A more exten-
genesis was that with high mutation rates, the sive theoretical investigation of optimum treat-
probability of accumulating deleterious mu- ment strategies would be useful to assess the
tations increases. This effect leads to nega- risks and benefits of DNA-damaging agents uti-
tive clonal selection of mutator phenotypes. lized to treat potentially unstable tumors. Fur-
Beckman and Loeb (2005) developed a dif- thermore, the number and identities of genes
ferential equation model including considera- causing genomic instability when mutated are
32 Annals of the New York Academy of Sciences

still mostly unknown, and a systems biology & Weinberg 2000) and was investigated by
approach to the identification of such genes, Komarova and Wodarz with a stochastic model
their tissue specificity, and membership of path- (Wodarz & Komarova 2007). They argued that
ways is needed. high rates of apoptosis increase cell turnover,
thus generating more mutants and increasing
Considerations of Cancer the probability of cancer progression. They
Progression found that an absence of apoptosis would drive
the population to an evolutionary dead end,
Understanding and preventing cancer pro- resulting in benign lesions unable to surpass fit-
gression is one of the central goals of cancer ness barriers; an optimum relationship between
research. The natural history of a tumor is de- rates of cell death and of mutations exists such
termined not only by the genetic and epigenetic that enough mutants emerge without disrup-
changes accumulating in the cancer cell popu- tion of other pathways needed for cell viabil-
lation, but also by the tumor’s interactions with ity. Also in 2007, Wodarz developed a stochas-
the microenvironment and the immune system, tic model with which he studied the effect of
as well as by the dynamics of different cell clones cell turnover and mutational mechanisms on
within the tumor. the processes of cancer progression and aging
Bodmer and Tomlinson (1995) showed that (Wodarz 2007). He found that if mutations oc-
clonal expansion caused by altered cell death cur independently of cell division, an increased
or differentiation rates can result in the popu- cell turnover results in a higher probability of
lation growing to higher plateaus in size. Us- tumor progression and a lower degree of ag-
ing a mathematical model of discrete-time dif- ing, therefore pointing to the existence of an
ferential equations, they investigated possible equilibrium that maximizes the life span of the
scenarios for cancer growth when rates of cell organism. If mutations occur only during cell
death and differentiation are altered by mu- division, both aging and cancer risk increase
tations. This model was able to explain long with cell turnover, thus favoring low turnover
lags in tumor progression and the existence rates.
of benign lesions growing to an equilibrium Neutral mutations have been at the center
cell number. In a later contribution, d’Onofrio of interest of evolutionary biologists ever since
and Tomlinson incorporated fluctuations in the Motoo Kimura introduced the theory of neu-
parameters and nonlinearity in the equations tral evolution in 1968; in this landmark pa-
into the model (d’Onofrio & Tomlinson 2007). per, he argued that most genetic variation is
They found that fluctuations of the population selectively neutral and hence neither subject
size at distinct differentiation stages increase the to nor explicable by natural selection (Kimura
probability of exponential growth, which is ir- 1968). Rather, most evolutionary change re-
reversible once initiated. In another extension sults from random drift of nonselected alle-
of Bodmer and Tomlinson’s work, Johnston les. The emergence of neutral mutations dur-
and colleagues (2007) relaxed the need for syn- ing cancer progression and clonal evolution is
chronous cell divisions and presented a con- of great importance since such changes could
tinuous approximation of the model. They serve as evolutionary “bottlenecks” (Maley &
concluded that feedback controls of the cell Forrest 2001): after the initial model by Tom-
population add stability to the system, ren- linson and colleagues in 1996 (Tomlinson et al.
dering exponential expansion impossible un- 1996), Maley and Forrest (2001) introduced a
less these regulatory mechanisms are altered computational model to investigate the rela-
by mutations. tionship between the numbers of neutral and
The importance of apoptosis in carcino- selected mutations as well as those changes
genesis has often been emphasized (Hanahan that cause mutator phenotypes. The model was
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 33

based on computer simulations of a cell popula- output of this analysis was a cancer mutation
tion proliferating on a two-dimensional lattice. prevalence (CaMP) score for each gene ana-
In a large parameter search, the authors found lyzed (Sjöblom et al. 2006). The CaMP score
that the number of selected mutations positively represents the probability that the number of
correlates with the number of neutral muta- mutations observed in a gene reflects a muta-
tions needed to progress to cancer. Contrary to tion frequency higher than expected by chance
the arguments against the necessity of mutator given the background mutation rate. However,
phenotypes for cancer evolution, the authors this method of estimating selection across the
argued that, with the normal mutation rate in genome has been criticized by several authors
human cells, a small number of selected muta- (Forrest & Cavet 2007; Getz et al. 2007; Rubin
tions cannot be accumulated in reasonable time & Green 2007; Chittenden et al. 2008). Dis-
frames if neutral mutations are also needed for cussions about the P-values used by Sjöblom
carcinogenesis. and colleagues as well as their estimates of the
The identification of neutral and positively background mutation rate have led to the sug-
selected mutants in tumorigenesis has attracted gestion that the CaMP score in its original form
much interest since the advent of powerful severely overestimated the number of driver
genome-wide analysis tools. The observation mutations in the analyzed datasets. These criti-
that some genetic aberrations are present in cisms were countered by the authors of the orig-
large fractions of tumor samples of the same, inal study, who argued that the experimental
and sometimes even different, cancer types sug- setup warranted a modification of the statisti-
gests a defining role of the implicated genes in cal approach that, if incorporated into the other
the process of tumorigenesis, but it has been groups’ models, would predict almost identical
difficult to systematically identify such “driver” numbers of proposed driver mutations across
mutations. To initiate a systematic analysis of the investigations (Parmigiani et al. 2007).
genetic alterations in cancer, Vogelstein and Such statistical issues cannot be ade-
colleagues (Sjöblom et al. 2006), as well as Strat- quately resolved without employing theoreti-
ton and colleagues (Yuen et al. 2007), deter- cal methodologies unrelated to the above tech-
mined the sequence of protein-coding genes niques, as well as a functional validation of the
in a total of 232 diverse human tumor sam- mutations that are ranked highly by the algo-
ples. The studies identified 189 genes (Sjöblom rithms. Such validation is essential for identi-
et al. 2006) and 120 genes (Yuen et al. 2007) fying true driver mutations and also computa-
that were mutated at significant frequency. To tional techniques that correctly predict which
distinguish genes likely to contribute to tumori- genes are functionally relevant for tumorige-
genesis from those in which passenger muta- nesis. In the following we review alternative
tions occurred by chance, Sjöblom and co- approaches to the identification of functionally
workers (2006) developed statistical methods to significant mutations in cancer.
estimate the probability that the number of mu- Maley and collaborators (2004a) used a sta-
tations in a given gene is greater than expected tistical approach to identify mutations confer-
from the background mutation rate. For each ring selective advantages in Barrett’s esopha-
gene, this analysis incorporated the number of gus. Biopsies sampled from different regions
somatic alterations observed in a genomic mu- of patients’ esophagi were analyzed for loss of
tation screen, the number of tumor samples heterozygosity, microsatellite shifts, point muta-
studied, and the number of nucleotides suc- tions, and methylation of selected loci. Genetic
cessfully analyzed. Because the mutation fre- alterations were sorted according to the pro-
quencies vary with nucleotide type and context portion of proliferating cells that carry the al-
and are different in different tumor types, these teration per sample, as well as the frequency of
factors were included in the calculations. The that alteration among patients. Highly ranked
34 Annals of the New York Academy of Sciences

changes according to this approach were de- nomic datasets and has led to several impor-
fined as driver mutations, and any genetic tant findings (Weir et al. 2007; TCGA 2008).
alteration that occurred exclusively together Taylor and co-workers (2008) presented a sim-
with such a driver was interpreted as a hitch- ilar statistical approach, RAE, to distinguish
hiker mutation. The authors found that ho- functionally neutral from causal chromosomal
mozygous p16 inactivation followed by p53 alterations in tumors. The key differences be-
mutations has a strong selective advantage in tween this method and earlier approaches are
Barrett’s esophagus. that RAE (i) distinguishes between four classes
In 2007, Sander and colleagues presented of genomic gains and losses to reflect the bio-
the Online Mutation Assessor (OMA), a com- logical significances of such alterations, (ii) ren-
putational approach incorporating informa- ders these four scoring models sample-specific,
tion about the sequence evolution and three- adapting to individual tumors to account for
dimensional structures of proteins and of their their differences, (iii) uses soft discrimination
interactions in macromolecular complexes, as rather than hard thresholds for improved sig-
well as their placement in molecular path- nal extraction, and (iv) generates a random
ways (Reva et al. 2007). Given a nonsynony- aberration model using a background of seg-
mous sequence variant, OMA provides a func- mental DNA rather than independent array
tional report and shows the mutated residues markers. When the performance of RAE and
in the context of a protein family alignment GISTIC were compared (TCGA 2008), the ge-
view and a 3-D structure view. The result is netic alterations that scored as statistically sig-
a prioritized list of mutations ranked by func- nificant were almost identical, suggesting that
tional score as well as background information both methodologies are equally good at identi-
that leads to the score, including evolutionarily fying significant aberrations in cancer.
conserved patterns across organisms as well as Although many different statistical and evo-
structure placement of the mutation. This algo- lutionary approaches to identifying driver mu-
rithm is promising since it incorporates infor- tations have been proposed, the field is still
mation about different aspects of the mutation lacking a comprehensive comparison of these
and might therefore lead to a more robust iden- techniques together with experimental efforts
tification of functionally important genes than to validate purported driver mutations. Only
approaches that incorporate only the frequency a functional validation in cell line and mouse
of mutations. experiments can prove that a specific genetic
Also in 2007, Beroukhim and colleagues de- alteration leads to cancer initiation or an accel-
veloped a statistical methodology called Ge- eration of malignant growth. Such endeavors
nomic Identification of Significant Targets in will be of crucial importance in the coming
Cancer (GISTIC) (Beroukhim et al. 2007). years.
GISTIC was designed to identify signifi- In 2007, Beerenwinkel and colleagues de-
cant copy-number changes in cancer genomes veloped a mathematical model to investigate
through two key steps: it first calculates a statis- the waiting time to cancer (Beerenwinkel et al.
tic that takes into account the frequency and 2007). Observations of the mutational patterns
amplitude of the genetic change, and then as- in colorectal cancer led to an estimation of
sesses the statistical significance of each genetic the number of driver mutations of around 20.
change by comparing the statistic derived in Based on this hypothesis, the authors studied
the first step to the results that would be ex- tumor growth and genotype dynamics and re-
pected by chance (Fig. 4A). This method iden- lated the time for a tumor to reach a given
tifies regions of aberration that are most likely size to the mutation rate, population size, and
to drive cancer pathogenesis. Since its publi- fitness advantage of the driver mutations. Sim-
cation, GISTIC has been applied to many ge- ulations and analytical results showed that for
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 35

Figure 4. Identification of driver mutations and their time of emergence during tumorigenesis. (A) The
GISTIC algorithm. After identifying the locations and magnitudes of chromosomal aberrations in multiple
tumors (left), GISTIC scores each genomic marker with a score that is proportional to the total magnitude of
aberrations at each location (upper center). In addition, by permuting the locations in each tumor, GISTIC
determines the frequency with which a given score would be attained if the events were due to chance and
therefore randomly distributed (lower center). A significance threshold (vertical line) is determined indicating
the value beyond which significant scores are unlikely to occur by chance alone. Alterations are deemed
significant if they occur in regions that surpass this threshold (right). From Beroukhim et al. (2007). (B)
A maximum likelihood tree for the karyotypic evolution of clear cell renal cell carcinoma, based on the
chromosomal aberrations seen in more than 10% of the cases. Deletions (amplifications) are denoted by a
minus (plus) sign followed by the chromosome number and arm. Figure adapted from von Heydebreck et al.
(2004).

reasonable parameter values, a fitness advan- for a large benign lesion to evolve into cancer
tage conferred by driver mutations (assuming but less than 2 years for cells within that cancer
normal mutation rates) could account for the to acquire the ability to metastasize; (ii) it takes
generation of a tumor within a human life span. few, if any, selective events to transform a highly
The model also predicts the high level of het- invasive cancer cell into one that has the ability
erogeneity seen in colorectal cancer patients, to metastasize; and (iii) the rates at which point
which, in the context of the model, is accounted mutations develop in advanced cancers are sim-
for by the emergence of different mutations af- ilar to those of normal cells. Such hypotheses,
fecting the same mechanistic pathways. The even though interesting, require experimental
authors showed that the assumption of a large validation and remain a theory without wet-lab
number of drivers with small fitness advantages efforts.
fits well with the data and the incidence of colo- It is of interest not only to identify all driver
rectal cancer. In 2008, the same group used mutations implicated in a particular cancer, but
mathematical modeling to arrive at some gen- also to uncover the temporal sequence in which
eral conclusions about colorectal carcinogene- these mutations arise. The earlier a driver mu-
sis (Jones et al. 2008): (i) it takes about 17 years tation emerges during tumorigenesis, the more
36 Annals of the New York Academy of Sciences

likely it is to induce “oncogenic addiction” It was shown that mixture trees could be used to
(the dependence of a cancer cell on the ac- investigate independent mutational pathways,
tivity of a particular oncogene) and therefore which were enigmatic when using single-tree
could represent the most promising drug tar- reconstruction. The authors also developed a
get (Weinstein 2002). In 1990, Fearon and Vo- measure of progression to estimate the sur-
gelstein published their paradigmatic result re- vival times of individual patients and validated
garding mutational pathways, suggesting that their predictions with clinical datasets. In 2002,
the accumulation of a linear series of genetic Newton developed a method to find combina-
changes transforms colonic tissue into invasive tions of genomic aberrations from CGH data
cancer (Fearon & Vogelstein 1990); however, (Newton 2002). This approach involved a joint
this so-called “Vogelgram” has since been criti- probability distribution of the samples’ profiles
cized for describing the tumorigenetic processes from which ensembles of genomic abnormali-
of only a subset of colorectal cancers (Smith ties were inferred. One important characteris-
et al. 2002). The identification of evolutionary tic of the model is that no preselection of rel-
trajectories toward cancer has become a hot evant genetic abnormalities is necessary, since
topic in theoretical research, particularly be- such changes are automatically inferred by the
cause the experimental conditions under which algorithm.
Fearon and Vogelstein’s results were obtained A variation of these tree reconstruction
are hard or even impossible to reproduce for methods was presented by von Heydebreck
other cancer types. and colleagues in 2004 (von Heydebreck et al.
A few years later, Desper and colleagues 2004). In their model, the leaves of the tree
(1999) presented one of the first theoretical represent mutational events while intermedi-
approaches to this question. They introduced ate nodes denote “hidden” events, which might
branching mutational trees—so-called oncoge- be interpreted biologically as intermediate (ge-
netic trees—to the investigation of cancer pro- netic) events (Fig. 4B). Starting with the wild-
gression. The method was applied to cross- type node, stochastic experiments are realized
sectional comparative genomic hybridization that evolve according to the joint probabili-
(CGH) data obtained from tumor samples. The ties between events. The highest-scoring tree
goal of the algorithm was to identify the tree is constructed using a likelihood-maximization
that best describes the observed joint proba- algorithm, resulting in a tree topology with
bilities of each pair of mutations. Their use branch lengths representing the probability to
of tree reconstruction methods allows for the move from one node to the other; the tree
formulation of temporal relations between ge- therefore contains information about the likely
netic changes and can lead to the identifica- temporal occurrence of events in a mutational
tion of the order in which mutations occur. pathway.
One drawback to the approach of Desper and Even though these phylogenetic algorithms
colleagues is that events in the leaves of the promise to reconstruct the temporal sequence
tree can only occur once all previous events in which driver mutations arise during tumori-
have happened, resulting in large portions of genesis, they have not been validated using the
the tree with zero likelihood of occurring. To one dataset for which the answer is known: col-
overcome this problem, Beerenwinkel and co- orectal tumorigenesis. Once an algorithm has
workers (2005) extended Desper’s algorithm by been validated with available data, the predic-
introducing mixture trees. These trees combine tions of the model should be tested in in vivo
suboptimal trees, including a special topology models to investigate the stage at which the
that contains all events independently; due to mutation arises experimentally as well as the
this modification, all genetic alterations can oc- effects it has on cell turnover, physiological de-
cur without depending on previous mutations. pendencies, and other issues.
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 37

Finally, in 2006, Michor and colleagues de- netic changes, or do other cellular adaptations
veloped stochastic mathematical models to in- provide for such capabilities? Can a mathe-
vestigate the dynamics of metastasis forma- matical model incorporating mechanical forces
tion in patients (Michor et al. 2006c; Michor within a tumor explain the patterns of metas-
& Iwasa 2006). The authors considered situ- tasis seen in patients? How can the evolution
ations in which one or two mutations–either of metastatic sites best be prevented by thera-
single activating mutations in genes such as pies? Future theoretical investigations aimed at
rat sarcoma virus homolog (RAS) and mye- reducing the mortality associated with invasion
locytomatosis viral oncogene homolog (MYC) will be useful for the clinical management of
(Michor et al. 2006c) or inactivation of both al- late-stage cancer.
leles of metastasis-suppressor genes like Meta-
stasis inhibition factor (NM23) (Michor &
Iwasa 2006)—are necessary to enable cells to Stem Cells, Differentiation,
metastasize. They considered the dynamics of and Heterogeneity
metastasis-enabling mutations in a cell popula-
tion of constant size and investigated whether The role of tissue-specific stem cells, differ-
metastatic potential is the property of all cells entiation hierarchies, and the resulting tumor
in the primary tumor or of only a small subset. heterogeneity in carcinogenesis has attracted
They found that if most metastases are caused the interest of both experimentalists and theo-
by mutations that confer a fitness advantage to reticians (Fig. 5). Tissues are organized into dif-
the cell in the primary tumor, then most or all ferentiation hierarchies of cells: the most prim-
cells in the primary tumor will carry metastasis- itive cells are pluripotent stem cells capable
enabling mutations; this effect arises because, in of proliferation, self-renewal, and the produc-
that case, a mutant cell is likely to reach large tion of many types of differentiated progeny
frequencies in the tumor. In contrast, if most (Reya et al. 2001). Stem cells produce commit-
metastases derive from disadvantageous muta- ted progenitors, which in turn produce even
tions, then only a small fraction of cells in the more committed cells. Differentiated cells typ-
primary tumor will have metastatic potential. ically proliferate to fulfill organ-specific tasks.
To compensate for the selective disadvantage in Once fully differentiated, however, such cells
the primary tumor, such mutations must be able may lose the ability to replicate, as illustrated
to successfully found metastases elsewhere with by the loss of nuclei in erythrocytes and ker-
a probability that is many orders of magnitude atinocytes (Bach et al. 2000). In fact, the orga-
higher than that for advantageous mutations. nization of tissues into hierarchical structures
Since this scenario is unlikely, the authors con- might have evolved to minimize the risk of car-
clude, most metastases should arise from mu- cinogenesis, since stem cells are more prone
tations that have reached a large frequency in to accumulating mutations that lead to cancer
the primary tumor. This finding is supported than differentiated cells due to their enhanced
by experimental evidence (Ramaswamy et al. self-renewal capabilities and should therefore
2003). These models were later extended to represent only a small fraction of cells in a tis-
situations in which the growth of the main tu- sue (Michor et al. 2003b).
mor is described by a branching process and Cairns (1975) was the first to put forward the
the tumor may undergo exponential expansion hypothesis that stem cells retain an “immortal
(Dingli et al. 2007a). strand” of genetic material. He proposed that to
Evolutionary approaches to invasion and keep from accumulating mutations, stem cells
metastasis are still underrepresented in the lit- could divide asymmetrically, retaining the orig-
erature, and many open questions remain. Is inal strand of DNA while passing the newly
metastatic ability conferred exclusively by ge- synthesized, and possibly mutated, strand to a
38 Annals of the New York Academy of Sciences

daughter cell. This hypothesis has since led to ture with a shorter lineage has a reduced risk
controversy, with Conboy and associates pro- of cancer. If the mutation rates of stem cells are
viding supporting evidence by showing tem- significantly smaller than those of progenitors
plate strand cosegregation in muscle stem cells or differentiated cells, then the risk of cancer
(Conboy et al. 2007), and Kiel and colleagues depends on the number of mutations neces-
(2007) insisting that hematopoietic stem cells sary to drive tumorigenesis: for large numbers
do not asymmetrically segregate chromosomes. of mutations, more transit and fewer stem cell
This controversy awaits resolution. divisions are favored. As the number of mu-
In 2002, Cairns presented a mathematical tations decreases, a structure with shorter lin-
model of cancer initiation based on the as- eage has a decreased risk of cancer. In the
sumption that mutant stem cells undergo apop- same year, Nowak and co-workers presented
tosis due to deficient DNA repair mechanisms a linear model of mutation accumulation that
and are replaced by progenitors that may have takes the spatial relationship between cells ex-
accumulated mutations (Cairns 2002). His set plicitly into account (Nowak et al. 2003). Using
of simple formulas showed good concordance stochastic processes, the authors showed that in
with observed tumor incidence of mice treated such a spatial model, fitness differences between
with carcinogens. Cairns argued that the bio- non–stem cells are unimportant since they are
logical features represented in his model could “washed out” of the system, leaving stem cells
explain the onset of cancer even with the low as the only source of lasting variation (Fig. 5B).
mutation rates of human cells. The authors also concluded that such spatial
Recently, Pepper and colleagues presented a organization results in an overall reduction in
model of cell differentiation and somatic evo- the probability of cancer initiation. Later on,
lution. By means of a system of differential Dingli and colleagues investigated the effects of
equations, the authors studied the effects of stochasticity on the dynamics of stem cells given
mutations as well as changing replication and the small number of replicating cells in a com-
differentiation rates in a hierarchically struc- partment (Dingli et al. 2007b). They showed
tured population (Pepper et al. 2007). Several that remission of tumors and rapid expansion
conclusions were drawn: carcinogenesis can of mutant clones could both be explained by
be initiated both in stem cells and transient- means of stochastic dynamics alone.
amplifying cells, genetic lesions disrupting dif- Such theoretical investigations of differenti-
ferentiation pathways are critical to tumorigen- ation structures provide important insights into
esis and should be considered among the most the evolutionary effects of tissue design. How-
fundamental hallmarks of cancer, and the struc- ever, in the absence of a clear connection to
ture of serial differentiation is a general strat- experimental results, these endeavors remain
egy for the suppression of somatic evolution in abstract. It would be interesting to see mathe-
tissues. matical approaches to situations in which dif-
Adding to the importance of differentiation ferentiation is disrupted by specific mutations.
structures is the fact that long lineages in- Such investigations can lead to suggestions of
crease the probability of accumulating muta- therapeutic intervention based on predicted ef-
tions, since there are more possibilities for mu- fects of perturbations of the system.
tations to arise if the number of cell divisions Genetic and phenotypic heterogeneity of
a clone has undergone is large. Frank and col- tumors is not only caused by differentiation
leagues (2003) developed a model to quantify hierarchies, but also by cancer progression
the risk of cancer initiation for different tissue (Maley et al. 2006), mutator phenotypes and the
architectures. The authors found that, if the evolution of resistance (Komarova & Wodarz
mutation rates of stem and differentiated cells 2003), and interactions of cancer cells with
are of comparable magnitude, a tissue struc- their microenvironment (Gatenby & Vincent
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 39

2003a) (Fig. 5A). Historically, special empha- mor. The author suggested that this “hypertu-
sis has been devoted to studying the mecha- mor” mechanism may be the cause of necrosis
nisms of generation and maintenance of such observed in many vascularized tumors. A simi-
heterogeneity, and a number of computational lar mechanism was later applied to study tumor
approaches have been developed. In 2002, incidence across species (Nagy et al. 2007).
González-Garcı́a and colleagues investigated a In 2006, Maley and colleagues demonstrated
stochastic spatial model of tumor growth and for the first time a direct relationship between
showed that coexistence of diverse clones with clonal diversity in premalignant lesions and
different fitness values in the same tumor is pos- progression to cancer (Maley et al. 2006). They
sible due to the complexity of the microenviron- investigated the heterogeneity of cellular clones
ment (González-Garcı́a et al. 2002). The evolu- in patients with Barrett’s esophagus using eco-
tion of a tumor in three-dimensional space was logical measures of diversity, finding that di-
simulated utilizing simple mutation–selection versity was highly correlated with risk of pro-
rules. The authors showed that maintenance of gression to esophageal carcinoma (Fig. 5C).
diversity can be explained by spatial dynam- Diversity was measured by three different in-
ics since the exclusion of competitors takes a dices, which incorporated information about
long time as compared to the time scale of loss of heterozygosity (LOH), microsatellite
tumor growth. A similar approach was used shifts, and sequence mutations across the
by Zhang and colleagues (2009). They imple- genome. A high level of clonal diversity was
mented a three-dimensional model of tumor found to increase the risk of progression to ma-
growth as a computer simulation and consid- lignancy, but to stem from genetic instability
ered several levels of environmental interac- only if such variation leads to viable pheno-
tions. They investigated the accumulation of types. Normal apoptotic pathways may thus
a linear sequence of mutations, each capable of prevent progression of the tumor by limiting
changing the phenotypic response of the cell to population diversity and eliminating cells with
the environment. The model predicted consid- a mutator phenotype.
erable heterogeneity at the molecular level and Theoretical investigations of this nature
different rates of evolution in distinct regions of are important both for an evolutionary
the tumor. For instance, in those clones located understanding of tumorigenesis as well as for
near blood vessels, diversity was reduced due a guideline for clinical interventions: it is im-
to the expansion of fast-growing phenotypes portant to predict which patients with prema-
which led to an increased rate of progression to lignant lesions are most likely to develop malig-
more aggressive variants; regions close to the nant tumors such that the risks of cancer and
core of the tumor show a sustained increase in of unnecessary surgery can be minimized. Un-
diversity since in that environment coexistence fortunately, few systems are amenable to ap-
of different clones is possible. In 2004, Nagy proaches relying on measurements of prema-
designed a differential equation model to de- lignant tissue. However, it is an urgent goal of
scribe three aspects of a solid tumor: change the field to identify methodologies and histolo-
in cell mass over time, change in tumor vascu- gies in which such important studies can be
larization over time, and competition between performed.
two different cancer cell types (Nagy 2004). The In 2008, Vincent, Gatenby, and Gillies pro-
model also describes the number of immature posed that cancer cells must surpass several
vascular endothelial cells, which build blood environmental barriers in order to acquire
vessels. The model predicts that natural selec- an aggressive–invasive phenotype (Vincent &
tion always favors more aggressive cancer cell Gatenby 2008; Gatenby & Gillies 2008). The
phenotypes, even if the growth of such pheno- authors presented an evolutionary game the-
types eventually destroys all or part of the tu- ory model to investigate the characteristics of
40 Annals of the New York Academy of Sciences

Figure 5. Differentiation hierarchies, spatial arrangements, and heterogeneity in cancer.


(A) Cancer stem cells and clonal succession. Cancer stem cells (middle) can differentiate into
transit-amplifying cells (horizontal arrows), but can also accumulate further mutations that drive
cancer progression (vertical arrows). Both differentiation and progression processes contribute
to tumor heterogeneity. From Weinberg (2007). (B) “Linear process” of somatic evolution.
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 41

tumor initiation, promotion, and progression strongly depend on changes in tumor microen-
(Vincent & Gatenby 2008). Their analysis was vironment, rather than exclusively on variation
based on the idea of adaptive landscapes, in of intracellular processes such as cell cycle reg-
which selective traits are altered as the microen- ulation or apoptotic mechanisms. In a similar
vironment changes; the microenvironment is setting, Gatenby and Gillies made several im-
in turn shaped by the evolving populations portant observations: the environment selects
of cells. Selective barriers arise from cell–cell for phenotypes instead of genotypes, the full
interactions, tissue organization, blood supply, cancer phenotype is attained only once all the
the stage of carcinogenesis, and other intra- barriers have been surpassed, and adaptation
and extra-cellular factors. Numerical simula- to one barrier may change the adaptive fitness
tions coincided with the observed biological response to subsequent ones (Gatenby & Gillies
features of a multistep process: initially, muta- 2008).
tions occur mostly in tumor suppressor genes— Basanta (2008) devised a model based on
these mutations are selectively (almost) neutral evolutionary game theory to analyze the inter-
but set the fitness landscape for the second actions between three different tumor cell phe-
phase of carcinogenesis, in which limited pro- notypes defined by autonomous growth, anaer-
liferation is initiated. This slow expansion is obic glycolysis, and tissue invasion. He found
surpassed only in the third stage of carcinogen- that the invasive phenotype is more likely to
esis when glycolytic and acid-resistant pheno- evolve after appearance of the glycolytic pheno-
types evolve. The authors concluded that nor- type, which would explain the presence of inva-
mal tissue is situated in an adaptive landscape, sive growth in many malignant tumors. These
which allows for the coexistence of multiple cell results suggest that therapies which increase the
types—a condition needed for the evolution fitness cost of switching to aerobic glycolysis
of multicellularity. While normal conditions in might decrease the probability of emergence
this landscape impose strict limitations on pro- of more invasive phenotypes; when applied to
liferation, populations of benign cells are vul- glioma progression, the model explains aspects
nerable to invasion of fitter genotypes, which such as the emergence of diffuse tumor cell in-
initiate tumor growth. Further progression is vasion in low-grade tumors.
limited by available blood supply, but accel- The application of concepts from evolu-
erated by promoters such as inflammation or tionary game theory to cancer enables re-
wounding which can increase the flow of nu- searchers to investigate the effects of frequency-
trients, as well as mutations leading to adapta- dependent fitness on tumorigenesis. Such
tions to acidic–hypoxic environments. The au- models may be superior to approaches us-
thors proposed that the stages of tumorigenesis ing frequency-independent fitness in situations

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
In this process, cells are arranged in a row and labeled i = 1, . . ., N . For reproduction, cells are chosen
proportional to their reproductive rate. The reproducing cell is replaced by two daughter cells, and all cells
to the right are shifted by one position. The right-most cell undergoes apoptosis (falls off the edge of the
one-dimensional table). This architecture can delay the onset of cancer since only mutations arising in the
left-most cell, the stem cell, remains in the population. From Nowak et al. (2003). (C) Clonal diversity in
individuals with Barrett’s esophagus. Barrett’s esophagus is an abnormal change in the cells of the lower end
of the esophagus thought to be caused by damage from chronic acid exposure and is considered to be a
premalignant condition associated with an increased risk of esophageal cancer. It is important to identify
those lesions likely to progress to cancer since unnecessary surgery should be avoided. In the panel, we
show genetically distinct fractions of a biopsy with their frequencies. Clones and total frequencies within a
segment were used to calculate the Shannon diversity index and mean pairwise genetic divergence scores.
Such measures of diversity are able to predict the risk of progression to esophageal cancer. From Maley et al.
(2006).
42 Annals of the New York Academy of Sciences

in which the cellular growth rate or some gence of resistance in the 1980s (Goldie & Cold-
other cell-specific parameter is a function of man 1983, 1984; Coldman & Goldie 1986). In
the behavior of other cells. For example, sce- 1983, they presented a mathematical model of
narios in which some cells secrete substances cancer treatment to investigate the risk of re-
that are beneficial to the population as a sistance, and found that the probability P of
whole but costly to produce for the cells them- having a resistant cell at any given time is a
selves (such as production of vascular endothe- sigmoid function of the number of cells in the
lial growth factor [VEGF] to initiate vascu- tumor, N , and the mutation rate, α, given by
larization) require game-theoretic approaches P = 1 − exp[−αN ] (Goldie & Coldman 1983).
(Axelrod et al. 2006). However, for some sit- The shape of this function plotted against the
uations arising in cancer it has not been logarithm of the population size is constant
found that a trait depends on the compo- across mutation rates—once the probability of
sition of the population. In those cases, the cure starts decreasing from the initial plateau,
consideration of frequency-dependent fitness small delays in the start of treatment lead to sig-
does not lead to appreciable additional insights nificant reductions in the chance of successful
and should be neglected due to unnecessary treatment (Fig. 6A). The authors proposed two
complexity. strategies to maximize the probability of suc-
The investigation of cellular heterogeneity cessful therapy: first, treatment must be started
in tumors has led to many insightful results as soon as possible. This conclusion was drawn
regarding its emergence, maintenance, and from the fact that the probability of cure de-
pathological importance. The interest of in- creases as the size of the tumor increases, and
vestigators in tumor heterogeneity was further also from the prediction that larger tumors
piqued by the clinical phenomenon of drug re- present a higher level of heterogeneity, which is
sistance, and many models were developed to inversely related to the probability of success-
study the dynamics of tumors during treatment ful therapy (Goldie & Coldman 1984); second,
and the kinetics of resistance mutations. multiple drugs should be used in combination
whenever possible, or otherwise alternated. As-
suming that the growth rate of sensitive cells is
Anti-Cancer Therapy and the decreased by the effect of the drug, short treat-
Evolution of Resistance ment breaks to limit the toxicity ensure that
clones do not reach large abundances when
The first wave of contributions to mathemat- the drug is not administered. The authors also
ical modeling of cancer treatment was started utilized stochastic models of tumor growth to
in 1964 by Skipper and colleagues who inves- calculate the risk that clones with multiple mu-
tigated the response of murine leukemias to tations emerge; their approach lead to the sug-
chemotherapy, establishing the so-called “log gestion of strategies maximizing the probability
kill” law: they showed that in an exponentially of successful therapy (Coldman & Goldie 1986).
growing population, the fraction of cells killed The prediction that early initiation of therapy
by a drug is independent of the total number and the use of multiple drugs could reduce the
of tumor cells (Skipper et al. 1964). Thereafter, risk of resistance became known as the Goldie-
other models of anti-cancer therapy were de- Coldman hypothesis.
veloped to estimate tumor growth rates and Coldman and Murray presented in 2000 a
to optimize chemotherapeutic dosing sched- stochastic model to investigate the optimum
ules (Norton & Simon 1977). The investigation administration strategies of chemotherapeutic
of cancer therapy from an evolutionary view- drugs for different tumor growth dynamics
point, however, was initiated only after Goldie (Coldman & Murray 2000). They considered
and Coldman started thinking about the emer- situations in which a patient is treated with two
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 43

Figure 6. The evolution of resistance to anti-cancer therapy. (A) The mathematical inves-
tigation of resistance mutations was initiated by Goldie and Coldman, who found that the
probability of cure depends on the mutation rate and the population size of the tumor and is
given by the exponential of the negative mutation rate times the population size. The panel
shows the probability of cure for two different mutation rates, A and B, versus the logarithm of
the population size. Equation from Goldie & Coldman (1983). (B) A stochastic process model
can be used to investigate the risk of resistance emerging in a mutation-selection network. If,
for example, three positions must be mutated for resistance to anti-cancer therapy and the
three mutations can be accumulated in any order, then the evolutionary trajectories from a
wild-type cell (three unmutated positions, 000) to an escape mutant (three mutated positions,
111) can be studied and the risk of resistance quantified. From Iwasa et al. (2003). (C) The
probability of producing resistant mutants before treatment depends on the death rate of tumor
cells, D. In this example, the mutation rate is set to zero once treatment is started, and mutants
are assumed not to die. The tumor size, N , is shown at which the probability of treatment
failure due to preexisting resistance equals 0.01. While the dependence is linear for a single
drug, it becomes stronger with an increasing number of drugs. Here the mutation rate is 10−6
and the growth rate of cells is one. From Komarova & Wodarz (2005).
44 Annals of the New York Academy of Sciences

drugs and assumed that mutations conferring of resistance by considering parameters like
resistance to one drug are sensitive to the other. apoptotic efficiency, the cost of DNA repair,
The authors optimized drug dosage strategies and the magnitude of the mutation rate. Their
to reduce toxicity and increase the probabil- stochastic model predicted that genetic insta-
ity of cure, showing that for equally effective bility is positively selected for in clones with
drugs, the optimum treatment regimen consists low mutation rates since that phenotype confers
of combined symmetric doses administered as enhanced evolvability to cells; in situations in
often as possible. The model predicts that an which mutation rates are high, however, genet-
early control of resistant clones—attained by ically stable cells experience a selective advan-
high initial doses followed by a decreasing dose tage since cells with a mutator phenotype ac-
for the remainder of the treatment time—is im- cumulate deleterious mutations at a high rate.
portant. Iwasa and colleagues (2003) designed The authors found that the outcome of the sys-
a stochastic mathematical model based on tem depends on two parameters: the difference
multi-type branching processes to investigate in the ability to repair DNA damage between
the evolutionary dynamics of resistance against stable and unstable cells and the cost of gen-
anti-cancer therapy. They considered a sce- erating lethal mutations due to genetic insta-
nario in which a heterogeneous population of bilities. In the same year, Gatenby and Vincent
cancer cells is subjected to chemotherapy, and combined population biology and evolutionary
calculated the probability of success or failure game theory to study the dynamics of tumor
of biomedical interventions consisting of one and healthy cells when treated with cytotoxic
or more drugs (Fig. 6B). They found that the drugs (Gatenby & Vincent 2003b). A model
chance of cure is largest when many drugs are reminiscent of Lotka–Volterra equations was
administered simultaneously since only if tu- used to describe the interactions of normal and
mor cells evolve many mutations will they be cancer cells interpreted as a predator–prey sys-
able to escape from eradication by therapy. tem, and the authors found that, in general,
The scenarios investigated in these contri- treatment with cytotoxic drugs alone is insuf-
butions represent just a subset of the situa- ficient to eradicate the tumor. This model was
tions that can emerge when tumor cells evolve used to identify two main barriers to achieving a
drug resistance. Other scenarios include the complete remission of the tumor: evolving pop-
treatment of a cancer patient with a cocktail ulations tend to produce resistant clones, which
of drugs that contains both specific (molecu- are able to drive the system back to equilibrium
larly targeted) and unspecific (general cytotoxic) abundance, and drug-induced alterations of the
agents, the necessity of altering several molecu- tumor microenvironment change the selection
lar pathways for a cell to evade chemotherapy pressure and select for tumor cells with larger
and immune attacks, dose-limiting toxicity and evolvability.
side effects, and the emergence of radiation- or The existence of resistance mutations at the
chemotherapy-induced secondary tumors. The time of diagnosis has been of considerable in-
literature on models optimizing therapeutic terest to the cancer research community since
strategies for such more complicated scenarios such pre-existing resistance influences treat-
is still small, and it is essential to obtain a quan- ment choices. The investigation of resistance
titative understanding of anti-cancer treatment mutations emerging while a population under-
strategies and their long-term risks and bene- goes exponential expansion was initiated by
fits such that drug resistance can be prevented; Luria and Delbrück in 1943 (Luria & Delbrück
the field is in dire need of optimization studies 1943). They were interested in the mutations
guiding therapy decisions. of bacteria that confer resistance to phages
Komarova and Wodarz (2003) investigated and performed experiments as well as cal-
the effects of mutator phenotypes on the risk culations to quantify the rate at which such
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 45

mutations emerge during bacterial growth. function of the detection size times the muta-
Their analytical results describing the distri- tion rate, and that a tumor with larger apop-
bution of the number of mutants in an expo- tosis rates has a higher incidence of resistance.
nentially growing population became known as Haeno and collaborators (2007) extended this
the Luria–Delbrück distribution. This distribu- stochastic model to incorporate resistance due
tion is also useful for situations arising in cancer to the accumulation of two mutations and ar-
and has been studied for more than half a cen- rived at similar conclusions. Later Komarova
tury (Skipper 1983; Sarkar 1991; Zheng 1999; and colleagues (2007) presented a variation of
Angerer 2001; Frank 2003; Dewanji et al. 2005; the Luria–Delbrück model for a stochastically
Iwasa et al. 2006; Haeno et al. 2007; Komarova growing population of cancer cells. As in prior
et al. 2007; Zheng 2008). Several different mod- papers, it was shown that a higher cell-death
els have been suggested that are based on pure rate leads to a larger number of mutants. Fur-
birth processes and do not include the possibil- thermore, irreversible mutations were found to
ity of cell death (Zheng 1999). However, in most act as a selective force in large cell populations
examples of cancer growth, cancer cell death even if they do not confer a fitness advantage
cannot be neglected. to the cell; this effect results from considering
In 2003, Frank developed a branching pro- unidirectional mutations, which continuously
cess model to investigate the distribution of reduce the pool of unmutated cells.
cells with different numbers of mutations (Frank These investigations of resistance mutations
2003). He found that the distribution of cells existing prior to diagnosis are important for a
with two mutations provides information about quantification of the risk of resistance as well as
the effective time of occurrence of the first mu- for deciding on appropriate treatment choices
tation; the latter—depending on its frequency at diagnosis. Unfortunately, cell-specific param-
in different tissues in the body—could even eters such as growth and death rates and muta-
have arisen during embryonal development. tion rates are unknown for most cancer types.
The time at which the first mutation emerges To adequately predict the risk of pre-existing
contains information about the number and resistance for a given tumor type, quantitative
types of tissues harboring this mutation, which measurements of these parameters are essen-
in turn influences cancer prognoses and pro- tial. It is therefore import to perform in vitro
vides information about tissue specificity ef- experiments to determine these rates.
fects. In 2005, Dewanji and colleagues de- In 2005, Komarova and Wodarz developed
veloped an extension of the Luria–Delbrück a stochastic mathematical model to investigate
model that considered non-exponential growth treatment strategies involving multiple drugs
dynamics (Dewanji et al. 2005). The authors (Komarova & Wodarz 2005). The effect of ther-
found that including the possibility of cell death, apy was modeled by increasing the ratio of cell
as well as the assumption of Gompertzian death and cell birth in cancer cells. The authors
growth of mutant cells, leads to larger varia- assumed that a single mutation is sufficient to
tions in the number of cells harboring resistance confer resistance to one particular drug; hence,
mutations. resistance against therapies consisting of n dif-
Iwasa and co-workers (2006) designed a ferent drugs requires the accumulation of n mu-
branching process model to calculate the prob- tations. The authors investigated the stochas-
ability of resistance mutations existing at the tic process model for situations with different
time of diagnosis, as well as the expected num- magnitudes of cell turnover and evaluated the
ber of resistant cells, as a function of detection importance of resistance mutations emerging
size and mutation rate. These authors also ex- during treatment with multiple drugs. They
plicitly considered cell death. They concluded found that the treatment phase is unimportant
that the probability of resistance is an increasing for the production of resistant cells and can
46 Annals of the New York Academy of Sciences

be neglected when determining the total risk sider, and there are other situations in cancer
of resistance—the majority of resistance muta- that should also be addressed. For instance,
tions arise before the tumor is diagnosed. The it is of interest to study the evolutionary dy-
chance of resistance increases with enhanced namics of cancer cell resistance when migra-
turnover rates independently of the number of tion and adaptation to foreign environments
drugs administered. The model was also used are included; a change in microenvironment
to find the optimum number of drugs such that might lead to modifications in gene expression,
both toxicity and the evolution of resistance are which can in turn modulate the risk of resis-
minimized; this number depends on the growth tance. Furthermore, the interaction between
and death rates of cells, the mutation rate, and tumor cells, stroma, immune system cells, and
the population size of cancer cells at the start of vascular endothelial cells may alter the risk
therapy (Fig. 6C). When the model was applied of resistance. Future studies of resistance dy-
to chronic myeloid leukemia (CML) to investi- namics should address these more complicated
gate the optimum number of drugs, it was found situations.
that administration of three chemotherapeu- While most mathematical models have fo-
tics minimizes the risk of resistance. Later on, cused on the action of cytotoxic or targeted
Komarova and Wodarz studied the importance drugs whose objective is to kill cancer cells,
of stem cell quiescence in the evolution of resis- Maley and colleagues recently studied the pos-
tance against anti-cancer treatment (Komarova sibility of using benign cell “boosters” to in-
& Wodarz 2007). The authors used a stochastic crease the fitness of healthy cells (Maley et al.
process model to investigate the dynamics of a 2004b). The authors developed a computa-
population of cancer stem cells that cycle be- tional model to investigate the dynamics of
tween an active, proliferative state and an inac- mutations emerging in cells that proliferate on
tive, quiescent state. The model was applied to a two-dimensional lattice. In this model, the
CML and could reproduce the biphasic decline genetic information of cells is represented by
in blood counts seen in CML patients (Michor a small number of loci acting as oncogenes,
et al. 2005a), which in the context of this model tumor suppressor genes, genes preventing
was explained by the differential response of mutator phenotypes, and genes conferring sen-
cycling and dormant stem cells to treatment sitivity to drugs. The population size is as-
with the targeted agent Gleevec. The authors sumed to be constant, thus exploiting com-
also predicted that the probability of resistance petition among different clones. The authors
would be independent of the presence and ex- showed that therapies that increase the fitness
tent of quiescence if therapy involves only a sin- of benign cells, so-called boosters, are effec-
gle drug. When two or more drugs are used, the tive in all stages of the disease when com-
risk of resistance increases with the percentage bined with traditional chemotherapy. They
of quiescent stem cells. In line with previous also suggested that increasing the fitness of
results obtained by these authors, the risk of chemosensitive cells prior to treatment would
resistance emerging during therapy is negligi- improve treatment outcomes. In a different ap-
ble when compared with resistance arising be- proach to cancer therapy, Wodarz designed a
fore the start of treatment. These findings have mathematical model to investigate the use of
important implications for the design of treat- viruses as anti-tumor agents (Wodarz 2001).
ment strategies since that showed that the use He studied a system of differential equations
of drugs that reduce the number of quiescent for three possible scenarios of the interaction
cancer stem cells cannot decrease the chance between virus particles and tumor cells, arriv-
of resistance effectively. ing at predictions about the necessary con-
The effect of quiescence on the risk of re- ditions for successful therapy. Such alterna-
sistance is an important phenomenon to con- tive approaches to treating cancer are worth
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 47

exploring and should be investigated with information, and other data. Such data should
quantitative techniques. not only provide spatial, but also temporal in-
formation about tumors. The development of
inexpensive, high-throughput single-cell assays,
Conclusions and Outlook as well as next-generation sequencing meth-
ods, will be beneficial for theoretical endeav-
The application of evolutionary thinking to ors. Furthermore, most evolutionary parame-
the study of cancer initiation, progression, het- ters such as mutation rates, fitness landscapes
erogeneity, and resistance has produced many of neoplasms, and population sizes and struc-
successful studies. However, several aspects of tures are virtually unknown. These parame-
cancer remain underrepresented in the evo- ters can be used to validate evolutionary mod-
lutionary literature and could benefit tremen- els; for example, the predictions of the Luria–
dously from more intensive quantitative inves- Delbrück distribution of resistance mutations
tigations. The putative cancer stem cells and pre-existing to therapy (Skipper 1983; Zheng
their dynamics during cancer progression and 1999; Sarkar 1991; Angerer 2001; Frank 2003;
therapy, for example, have not received much Dewanji et al. 2005; Iwasa et al. 2006; Haeno
attention. This fact may in part be due to et al. 2007; Komarova et al. 2007; Zheng 2008)
the relative novelty of experimental studies of should be tested in cell line experiments that
cancer stem cells and the comparatively small can directly assay the frequency of apoptosis
amount of biological information available to as well as the growth and mutation rates of
modelers; however, quantitative and evolution- cells. Such studies of experimental evolution
ary approaches to cancer stem cells will be remain rare but would provide valuable infor-
necessary for driving the investigations prov- mation to modelers. Once more data are acces-
ing (or disproving) their existence, their mech- sible to theoreticians, the evolution of the field
anisms of treatment insensitivity, and related will be accelerated. Interdisciplinary investiga-
aspects. Similarly, a full understanding of re- tions are furthermore hindered by the compli-
sistance from an evolutionary viewpoint still cations of securing funding. Most major medi-
requires considerable work. Many cancers re- cal funding sources prefer traditional molecular
lapse despite therapy shown to be effective biology or biochemical studies to mathemati-
in vitro, and the causes of refractory disease re- cal investigations, and the field would benefit
main to be identified in many cases. Pharma- tremendously from a funding source for inter-
cological modeling such as the investigation of disciplinary work. Also, the training of the next
pharmacokinetics and pharmacodynamics in generation of evolutionary modelers of can-
combination with the emergence of resistance cer requires the design of a useful curriculum,
similarly requires further input from evolution- since the sheer amount of knowledge necessary
ary theorists. The questions about the optimum (cancer biology, evolution, applied mathemat-
dosing strategies that utilize small therapeutic ics, bioinformatics, and statistics) renders most
windows need to be answered with sophisti- study courses superficial. Along the same line,
cated mathematical models that push the field evolutionary training of oncologists and clini-
beyond the current state. cians would be beneficial for the understanding
The main barriers to theoretical investiga- of cancer.
tions of cancer remain a lack of generally acces- In closing, we believe that the application of
sible and quality-controlled databases of treat- evolutionary modeling to cancer will continue
ment response data, mutation status of cancers to increase our understanding of neoplasms
for resistance investigations, heterogeneity in- and will eventually contribute to the design of
formation in the form of genomic data such as systems biology-based strategies to detect and
SNP arrays, gene expression data and sequence manage the disease.
48 Annals of the New York Academy of Sciences

Acknowledgments netic instability and the quasispecies model. J. Theor.


Biol., 241, 216–222.
We would like to thank Steven Frank, Carlo Buss, L. W. (1987). The evolution of individuality. Princeton,
Maley, Robert Downey, and the Michor lab for NJ: Princeton University Press.
critical reading and comments. Cairns, J. (1975). Mutation selection and the natural his-
tory of cancer. Nature, 255, 197–200.
Cairns, J. (2002). Somatic stem cells and the kinetics of
Conflicts of Interest mutagenesis and carcinogenesis. Proc. Natl. Acad. Sci.
USA, 99, 10567–10570.
The authors declare no conflicts of interest. Charles, D., & Luce-Clausen, E. (1942). The kinetics
of papilloma formation in benzpyrene-treated mice.
Cancer Research, 2, 261–263.
References Chittenden, T. W., Howe, E. A., Culhane, A. C., & Sul-
tana, R. (2008). Functional classification analysis of
somatically mutated genes in human breast and col-
Angerer, W. P. (2001). An explicit representation of the orectal cancers. Genomics, 91, 508–511.
Luria-Delbrück distribution. Journal of Mathematical Coldman, A. J., & Goldie, J. H. (1986). A stochastic model
Biology, 42, 145–174. for the origin and treatment of tumors containing
Armitage, P., & Doll, R. (1957). A two-stage theory of drug-resistant cells. Bull. Math. Biol., 48, 279–292.
carcinogenesis in relation to the age distribution of Coldman, A. J., & Murray, J. M. (2000). Optimal con-
human cancer. Br. J. Cancer, 11, 161–169. trol for a stochastic model of cancer chemotherapy.
Ashley, D. J. (1969). Colonic cancer arising in polyposis Mathematical Biosciences, 168, 187–200.
coli. Journal of Medical Genetics, 6, 376–378. Conboy, M. J., Karasov, A. O., & Rando, T. A. (2007).
Axelrod, R., Axelrod, D. E., & Pienta, K. J. (2006). Evo- High incidence of non-random template strand seg-
lution of cooperation among tumor cells. Proc. Natl. regation and asymmetric fate determination in di-
Acad. Sci. USA, 103, 13474–13479. viding stem cells and their progeny. Plos Biol., 5,
Bach, S. P., Renehan, A. G., & Potten, C. S. (2000). Stem e102.
cells: the intestinal stem cell as a paradigm. Carcino- Basanta, D., Simon, M., Hatzikirou, H., & Deutsch, A.
genesis, 21, 469–476. 2008. Evolutionary game theory elucidates the role
Beckman, R. A., & Loeb, L. A. (2005). Negative clonal of glycolysis in glioma progression and invasion. Cell
selection in tumor evolution. Genetics, 171, 2123– Proliferation, 41, 980–987.
2131. d’Onofrio, A., & Tomlinson, I. P. (2007). A nonlinear
Beerenwinkel, N. (2005). Mtreemix: a software package mathematical model of cell turnover, differentiation
for learning and using mixture models of mutage- and tumorigenesis in the intestinal crypt. J. Theor.
netic trees. Bioinformatics, 21, 2106–2107. Biol., 244, 367–374.
Beerenwinkel, N., Antal, T., Dingli, D., Traulsen, A., Desper, R., Jiang, F., Kallioniemi, O. P., Moch, H., Pa-
Kinzler, K. W., Velculescu, V. E., et al. (2007). Ge- padimitriou, C. H., & Schäffer, A. A. (1999). Infer-
netic progression and the waiting time to cancer. ring tree models for oncogenesis from comparative
PLoS Comput. Biol., 3, e225. genome hybridization data. J. Comput. Biol., 6, 37–
Bernoulli, D., & Blower, S. (2004). An attempt at a new 51.
analysis of the mortality caused by smallpox and of Dewanji, A., Luebeck, E. G., & Moolgavkar, S. H. (2005).
the advantages of inoculation to prevent it. 1766. Rev. A generalized Luria–Delbrück model. Mathematical
Med. Virol., 14, 275–288. Biosciences, 197, 140–152.
Beroukhim, R., Getz, G., Nghiemphu, L., Barretina, J., Dingli, D., Michor, F., Antal, T., & Pacheco, J. M. (2007a).
Hsueh, T., Linhart, D., et al. (2007). Assessing the The emergence of tumor metastases. Cancer Biol.
significance of chromosomal aberrations in cancer: Ther., 6, 383–390.
methodology and application to glioma. Proc. Natl. Dingli, D., Traulsen, A., & Pacheco, J. M. (2007b).
Acad. Sci. USA, 104, 20007–20012. Stochastic dynamics of hematopoietic tumor stem
Bodmer, W., & Tomlinson, I. (1995). Failure of pro- cells. Cell Cycle, 6, 461–466.
grammed cell death and differentiation as causes Dobzhansky, T. (1973). Nothing in biology makes sense
of tumors: Some simple mathematical models. Proc. except in the light of evolution. American Biology
Natl. Acad. Sci. USA: 11130–11134. Teacher, 35, 125–129.
Boveri, T. (1914). Zur Frage der Entstehung maligner Tumoren. Eigen, M., & Schuster, P. (1977). The hypercycle. A prin-
Jena, Germany: Verlag G. Fischer. ciple of natural self-organization. Part A: Emergence
Brumer, Y., Michor, F., & Shakhnovich, E. I. (2006). Ge- of the hypercycle. Naturwissenschaften, 64, 541–565.
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 49

Enderling, H., Chaplain, M. A., Anderson, A. R., & gin of drug resistance in neoplasms: implications for
Vaidya, J. S. (2007). A mathematical model of breast systemic therapy. Cancer Research, 44, 3643–3653.
cancer development, local treatment and recurrence. González-Garcı́a, I., Solé, R. V., & Costa, J. (2002).
J. Theor. Biol., 246, 245–259. Metapopulation dynamics and spatial heterogeneity
Fearon, E. R., & Vogelstein, B. (1990). A genetic model in cancer. Proc. Natl. Acad. Sci. USA, 99, 13085–13089.
for colorectal tumorigenesis. Cell, 61, 759–767. Haeno, H., Iwasa, Y., & Michor, F. (2007). The evolution
Fisher, J. C. (1958). Multiple-mutation theory of carcino- of two mutations during clonal expansion. Genetics,
genesis. Nature, 181, 651–652. 177, 2209–2221.
Fisher, J. C., & Hollomon, J. H. (1951). A hypothesis for Hanahan, D., & Weinberg, R. A. (2000). The hallmarks
the origin of cancer foci. Cancer, 4, 916–918. of cancer. Cell, 100, 57–70.
Forrest, W. F., & Cavet, G. (2007). Comment on “The Herrero-Jimenez, P., Tomita-Mitchell, A., & Furth, E. E.
Consensus Coding Sequences of Human Breast and (2000). Population risk and physiological rate param-
Colorectal Cancers.” Science, 317, 1500b. eters for colon cancer. The union of an explicit model
Frank. (2003). Somatic mosaicism and cancer: inference for carcinogenesis with the public health records of
based on a conditional Luria-Delbrück distribution. the United States. Mutation Research-Fundamental and
J. Theor. Biol., 223, 405–412. Molecular Mechanisms of Mutagenesis, 447, 73–116.
Frank. (2004a). Genetic predisposition to cancer – insights Iwasa, Y., Michor, F., & Nowak, M. A. (2003). Evolution-
from population genetics. Nat. Rev. Genet., 5, 764– ary dynamics of escape from biomedical interven-
772. tion. Proc. Biol. Sci., 270, 2573–2578.
Frank. (2004b). Genetic variation in cancer predisposi- Iwasa, Y., Michor, F., & Nowak, M. A. (2004). Stochas-
tion: mutational decay of a robust genetic control tic tunnels in evolutionary dynamics. Genetics, 166,
network. Proc. Natl. Acad. Sci. USA, 101, 8061–8065. 1571–1579.
Frank (2005). Age-specific incidence of inherited versus Iwasa, Y., Nowak, M. A., & Michor, F. (2006). Evolution
sporadic cancers: a test of the multistage theory of of resistance during clonal expansion. Genetics, 172,
carcinogenesis. Proc. Natl. Acad. Sci. USA, 102, 1071– 2557–2566.
1075. Jallepalli, P. V., & Lengauer, C. (2001). Chromosome seg-
Frank (2007). Dynamics of cancer: Incidence, inheritance, and regation and cancer: cutting through the mystery.
evolution. Princeton, NJ: Princeton University Press. Nat. Rev. Cancer, 1, 109–117.
Frank, Iwasa, Y., & Nowak, M. A. (2003). Patterns of cell Johnston, M. D., Edwards, C. M., Bodmer, W. F., Maini, P.
division and the risk of cancer. Genetics, 163, 1527– K., & Chapman, S. J. (2007). Mathematical model-
1532. ing of cell population dynamics in the colonic crypt
Friend, S. H., Bernards, R., Rogelj, S., & Weinberg, R. and in colorectal cancer. Proc. Natl. Acad. Sci. USA,
A. (1986). A human DNA segment with properties 104, 4008–4013.
of the gene that predisposes to retinoblastoma and Jones, S., Chen, W. D., Parmigiani, G., Diehl, F., Beeren-
osteosarcoma. Nature, 323, 643–646. winkel, N., Antal, T., et al. (2008). Comparative le-
Gatenby, R. A., & Gillies, R. J. (2008). A microenviron- sion sequencing provides insights into tumor evolu-
mental model of carcinogenesis. Nat. Rev. Cancer, 8, tion. Proc. Natl. Acad. Sci. USA, 105, 4283–4288.
56–61. Kiel, M. J., He, S., Ashkenazi, R., Gentry, S. N., & Teta,
Gatenby, R. A., & Maini, P. K. (2003). Mathematical M. (2007). Haematopoietic stem cells do not asym-
oncology: cancer summed up. Nature, 421, 321. metrically segregate chromosomes or retain BrdU.
Gatenby, R. A., & Vincent, T. L. (2003a). An evolutionary Nature, 449, 238–243.
model of carcinogenesis. Cancer Research, 63, 6212– Kimura, M. (1968). Evolutionary rate at the molecular
6220. level. Nature, 217, 624–626.
Gatenby, R. A., & Vincent, T. L. (2003b). Applica- Kinzler, K. W., & Vogelstein, B. (1996). Lessons from
tion of quantitative models from population biology hereditary colorectal cancer. Cell, 87, 159–170.
and evolutionary game theory to tumor therapeutic Knudson, A. G. (1971). Mutation and cancer: statistical
strategies. Mol. Cancer Ther., 2, 919–927. study of retinoblastoma. Proc. Natl. Acad. Sci. USA, 68,
Getz, G., Hofling, H., Mesirov, J., Golub, T., Meyerson, 820–823.
M., Tibshirani, R., & Lander, E. (2007). Comment Knudson, A. G. (1986). Genetics of Human Cancer. An-
on “The Consensus Coding Sequences of Human nual Reviews in Genetics, 20, 231–251.
Breast and Colorectal Cancers”. Science, 317, 1500. Knudson, A. G. (2001). Two genetic hits (more or less) to
Goldie, J. H., & Coldman, A. J. (1983). Quantitative model cancer. Nat. Rev. Cancer, 1, 157–162.
for multiple levels of drug resistance in clinical tu- Kolodner, R. D., Putnam, C. D., & Myung, K. (2002).
mors. Cancer Treatment Reports, 67, 923–931. Maintenance of Genome Stability in Saccharomyces
Goldie, J. H., & Coldman, A. J. (1984). The genetic ori- cerevisiae. Science, 297, 552–557.
50 Annals of the New York Academy of Sciences

Komarova, N. L., Sengupta, A., & Nowak, M. A. (2003). Maley, C. C., & Forrest, S. (2001). Exploring the rela-
Mutation-selection networks of cancer initiation: tu- tionship between neutral and selective mutations in
mor suppressor genes and chromosomal instability. cancer. Artif. Life, 6, 325–345.
J. Theor. Biol., 223, 433–450. Merlo, L. M., Pepper, J., Reid, B., & Maley, C. (2006).
Komarova, N. L., & Wodarz, D. (2003). Evolutionary Cancer as an evolutionary and ecological process.
dynamics of mutator phenotypes in cancer: impli- Nat. Rev. Cancer, 6, 924–935.
cations for chemotherapy. Cancer Research, 63, 6635– Michor, F., Frank, May, R. M., Iwasa, Y., & Nowak, M.
6642. A. (2003a). Somatic selection for and against cancer.
Komarova, N. L., & Wodarz, D. (2004). The optimal rate J. Theor. Biol., 225, 377–382.
of chromosome loss for the inactivation of tumor Michor, F., Hughes, T. P., Iwasa, Y., Branford, S., Shah,
suppressor genes in cancer. Proc. Natl. Acad. Sci. USA, N. P., Sawyers, C. L., et al. (2005a). Dynamics of
101, 7017–7021. chronic myeloid leukaemia. Nature, 435, 1267–1270.
Komarova, N. L., & Wodarz, D. (2005). Drug resistance Michor, F., & Iwasa, Y. (2006). Dynamics of metastasis
in cancer: principles of emergence and prevention. suppressor gene inactivation. J. Theor. Biol., 241, 676–
Proc. Natl. Acad. Sci. USA, 102, 9714–9719. 689.
Komarova, N. L., & Wodarz, D. (2007). Effect of cellular Michor, F., Iwasa, Y., Lengauer, C., & Nowak, M. A.
quiescence on the success of targeted CML therapy. (2005b). Dynamics of colorectal cancer. Semin. Cancer
PLoS ONE, 2, e990. Biol., 15, 484–493.
Komarova, N. L., Wu, L., & Baldi, P. (2007). The fixed- Michor, F., Iwasa, Y., & Nowak, M. A. (2004a). Dynamics
size Luria–Delbruck model with a nonzero death of cancer progression. Nat. Rev. Cancer, 4, 197–205.
rate. Mathematical Biosciences, 210, 253–290. Michor, F., Iwasa, Y., & Nowak, M. A. (2006a). The age
Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1997). incidence of chronic myeloid leukemia can be ex-
Genetic instability in colorectal cancers. Nature, 386, plained by a one-mutation model. Proc. Natl. Acad.
623–627. Sci. USA, 103, 14931–14934.
Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1998). Michor, F., Iwasa, Y., Rajagopalan, H., Lengauer, C., &
Genetic Instabilities In human cancers. Nature, 396, Nowak, M. A. (2004b). Linear model of colon cancer
643–649. initiation. Cell Cycle, 3, 358–362.
Liso, A., Castiglione, F., Cappuccio, A., & Stracci, F. Michor, F., Nowak, M. A., Frank, & Iwasa, Y. (2003b).
(2008). A one-mutation mathematical model can ex- Stochastic elimination of cancer cells. Proc. Biol. Sci.,
plain the age incidence of AML with mutated nucle- 270, 2017–2024.
ophosmin (NPM1). Haematologica, 93, 1219–1226. Michor, F., Nowak, M. A., & Iwasa, Y. (2006b). Evolution
Loeb, L. A. (1991). Mutator phenotype may be required of resistance to cancer therapy. Curr. Pharm. Des., 12,
for multistage carcinogenesis. Cancer Research, 51, 261–271.
3075–3079. Michor, F., Nowak, M. A., & Iwasa, Y. (2006c). Stochastic
Luebeck, E. G., & Moolgavkar, S. H. (2002). Multistage dynamics of metastasis formation. J. Theor. Biol., 240,
carcinogenesis and the incidence of colorectal can- 521–530.
cer. Proceedings of the National Academy of Sciences, 99, Nagy, J. D. (2004). Competition and natural selection in a
15095–15100. mathematical model of cancer. Bull. Math. Biol., 66,
Luria, S. E., & Delbrück, M. (1943). Mutations of bacteria 663–687.
from virus sensitivity to virus resistance. Genetics, 28, Nagy, J. D., Victor, E. M., & Cropper, J. H. (2007).
491–511. Why don’t all whales have cancer? A novel hy-
Maley, C., Galipeau, P., Finley, J., Wongsurawat, V., Li, pothesis resolving Peto’s paradox. Integr. Comp. Biol.
X., Sanchez, C., et al. (2006). Genetic clonal diversity doi: 10.1093/icb/icm062.
predicts progression to esophageal adenocarcinoma. Newton, M. A. (2002). Discovering Combinations of Ge-
Nat. Genet., 38, 468–473. nomic Aberrations Associated With Cancer. Journal
Maley, C., Galipeau, P., Li, X., Sanchez, C., Paulson, T., of the American Statistical Association, 97, 931–942.
& Reid, B. (2004a). Selectively advantageous muta- Nordling, C. O. (1953). A new theory on cancer-inducing
tions and hitchhikers in neoplasms: p16 lesions are mechanism. Br. J. Cancer, 7, 68–72.
selected in Barrett’s esophagus. Cancer Research, 64, Norton, L., & Simon, R. (1977) Growth curve of an exper-
3414–3427. imental solid tumor following radiotherapy. J. Natl.
Maley, C., Reid, B., & Forrest, S. (2004b). Cancer preven- Cancer Inst., 58, 1735–1741.
tion strategies that address the evolutionary dynam- Nowak, M. A., Komarova, N. L., Sengupta, A., &
ics of neoplastic cells: simulating benign cell boosters Jallepalli, P. V. (2002). The role of chromosomal in-
and selection for chemosensitivity. Cancer Epidemiol. stability in tumor initiation. Proc. Natl. Acad.f Sci. USA,
Biomarkers Prev., 13, 1375–1384. 99, 16226–16231.
Stephan-Otto Attolini & Michor: Evolutionary Theory of Cancer 51

Nowak, M. A., Michor, F., & Iwasa, Y. (2003). The linear Kirsten-ras, and p53–alternative genetic pathways
process of somatic evolution. Proc. Natl. Acad. Sci. USA, to colorectal cancer. Proc. Natl. Acad. Sci. USA, 99,
100, 14966–14969. 9433–9438.
Nowak, M. A., Michor, F., Komarova, N. L., & Iwasa, Solé, R. V., & Deisboeck, T. (2004). An error catastrophe
Y. (2004). Evolutionary dynamics of tumor suppres- in cancer? J. Theor. Biol., 228, 47–54.
sor gene inactivation. Proc. Natl. Acad. Sci. USA, 101, Taylor, B., Barretina, J., Socci, N. D., DeCarolis, P., &
10635–10638. Sander, C. (2008). Functional Copy-number Alter-
Nowell, P. C. (1976). The clonal evolution of tumor cell ations in Cancer. PLoS ONE, 3, e3179.
populations. Science, 194, 23–28. TCGA, T. C. G. A. R. N. (2008). Comprehensive genomic
Nunney, L. (2003). The population genetics of multistage characterization defines human glioblastoma genes
carcinogenesis. Proc. Biol. Sci., 270, 1183–1191. and core pathways. Nature, 455, 1061–1068.
Parmigiani, G., Lin, J., Boca, S. M., Sjoblom, T., & Tomlinson, I. P., Novelli, M. R., & Bodmer, W. F. (1996).
Jones, S. (2007). Response to Comments on “The The mutation rate and cancer. Proc. Natl. Acad. Sci.
Consensus Coding Sequences of Human Breast and USA, 93, 14800–14803.
Colorectal Cancers.” Science, 317, 1500. Vincent, T. L., & Gatenby, R. A. (2008). An evolutionary
Pepper, J., Sprouffske, K., & Maley, C. (2007). Animal cell model for initiation, promotion, and progression in
differentiation patterns suppress somatic evolution. carcinogenesis. Int. J. Oncol., 32, 729–737.
PLoS Comput. Biol., 3, e250. Vogelstein, B., & Kinzler, K. W. (2002). The genetic basis of
Perucho, M. (1996). Cancer of the microsatellite mutator human cancer. New York: McGraw-Hill.
phenotype. Biol. Chem., 377, 675–684. von Heydebreck, A., Gunawan, B., & Füzesi, L. (2004).
Prioreschi, P. (2002). Al-Kindi, A Precursor Of The Sci- Maximum likelihood estimation of oncogenetic tree
entific Revolution. ISHIM, 1, 17–20. models. Biostatistics (Oxford, England), 5, 545–556.
Rajagopalan, H., Nowak, M. A., Vogelstein, B., & Weinberg, R. A. (2007). The biology of cancer. London: Gar-
Lengauer, C. (2003). The significance of unstable land Science, Taylor and Francis.
chromosomes in colorectal cancer. Nat. Rev. Cancer, Weinstein, I. B. (2002). CANCER: Addiction to
3, 695–701. oncogenes–the achilles heal of cancer. Science, 297,
Ramaswamy, S., Ross, K. N., Lander, E. S., & Golub, 63–64.
T. R. (2003). A molecular signature of metastasis in Weir, B., Woo, M., Getz, G., Perner, S., Ding, L.,
primary solid tumors. Nat. Genet., 33, 49–54. Beroukhim, R., et al. (2007). Characterizing the can-
Reva, B., Antipin, Y., & Sander, C. (2007). Determinants cer genome in lung adenocarcinoma. Nature, 450,
of protein function revealed by combinatorial en- 893–898.
tropy optimization. Genome Biol., 8, R232. Wodarz, D. (2001). Viruses as antitumor weapons defining
Reya, T., Morrison, S. J., Clarke, M. F., & Weissman, I. conditions for tumor remission. Cancer Research, 61,
L. (2001). Stem cells, cancer, and cancer stem cells. 3501–3507.
Nature, 414, 105–111. Wodarz, D. (2007). Effect of stem cell turnover rates on
Rubin, A. F., & Green, P. (2007). Comment on “The protection against cancer and aging. J. Theor. Biol.,
Consensus Coding Sequences of Human Breast and 245, 449–458.
Colorectal Cancers.” Science, 317, 1500c. Wodarz, D., & Komarova, N. (2007). Can loss of apop-
Sarkar, S. (1991). Haldane’s Solution of the Luria- tosis protect against cancer? Trends Genet., 23, 232–
Delbruck Distribution. Genetics, 127, 257–261. 237.
Sjöblom, T., Jones, S., Wood, L. D., Parsons, D. W., Lin, Yuen, Leung, S. Y., Wooster, R., Futreal, P. A., & Stratton,
J., Barber, T. D., et al. (2006). The consensus coding M. R. (2007). Patterns of somatic mutation in human
sequences of human breast and colorectal cancers. cancer genomes. Nature, 446, 153–158.
Science, 314, 268–274. Zhang, L., Strouthos, C. G., Wang, Z., & Deisboeck, T. S.
Skipper, H. E. (1983). The forty-year-old mutation theory (2009). Simulating brain tumor heterogeneity with
of Luria and Delbruck and its pertinence to cancer a multiscale agent-based model: Linking molecular
chemotherapy. Adv. Cancer Res., 40, 331–363. signatures, phenotypes and expansion rate. Mathe-
Skipper, H. E., Schabel, Jr., F. M., & Wilcox, W. matical and Computer Modelling, 49, 307–319.
(1964). Experimental evaluation of potential anti- Zheng, Q. (1999). Progress of a half century in the study
cancer agents. XIII. On the criteria and kinetics as- of the Luria–Delbrück distribution. Mathematical Bio-
sociated with “curability” of experimental leukemia. sciences, 162, 1–32.
Cancer Chemother. Rep., 35, 1–111. Zheng, Q. (2008). On Bartlett’s formulation of the Luria–
Smith, G., Carey, F. A., Beattie, J., Wilkie, M. J., Lightfoot, Delbrück mutation model. Mathematical Biosciences,
T. J., Coxhead, J., et al. (2002). Mutations in APC, 215, 48–54.

Das könnte Ihnen auch gefallen