Sei sulla pagina 1di 31

Reports on Progress in Physics

Related content
- Invited Article
Biophysical models of tumour growth J S Lowengrub, H B Frieboes, F Jin et al.

- The fundamental role of mechanical


To cite this article: P Tracqui 2009 Rep. Prog. Phys. 72 056701 properties in the progression of cancer
disease and inflammation
Claudia Tanja Mierke

- Dynamic interplay between tumour, stroma


and immune system can drive or prevent
View the article online for updates and enhancements. tumour progression
R J Seager, Cynthia Hajal, Fabian Spill et
al.

Recent citations
- Acid-mediated tumor invasion as a
function of nutrient source location
Silvia A. Menchón

- A cell-based model for analyzing growth


and invasion of tumor spheroids
PengCheng Chen et al

- Aggregation dynamics of active cells on


non-adhesive substrate
Debangana Mukhopadhyay and Rumi De

This content was downloaded from IP address 132.248.227.150 on 08/10/2019 at 19:52


IOP PUBLISHING REPORTS ON PROGRESS IN PHYSICS
Rep. Prog. Phys. 72 (2009) 056701 (30pp) doi:10.1088/0034-4885/72/5/056701

Biophysical models of tumour growth


P Tracqui
CNRS UMR 5525, Laboratoire TIMC-IMAG, DynaCell Group, Institut de l’Ingénierie et de
l’Information de Santé (In3S), Faculté de Médecine de Grenoble, 38706, La Tronche Cedex, France
E-mail: Philippe.Tracqui@imag.fr

Received 9 October 2008, in final form 11 March 2009


Published 29 April 2009
Online at stacks.iop.org/RoPP/72/056701

Abstract
Tumour growth is a multifactorial process, which has stimulated in recent decades the
development of numerous models trying to figure out the mechanisms controlling solid
tumours morphogenesis. While the earliest models were focusing on cell proliferation
kinetics, modulated by the availability of supplied nutrients, new modelling approaches
emphasize the crucial role of several biophysical processes, including local matrix
remodelling, active cell migration and traction, and reshaping of host tissue vasculature. After
a brief presentation of this experimental background, this review will outline a number of
representative models describing, at different scales, the growth of avascular and vascularized
tumours. Special attention will be paid to the formulation of tumour–host tissue interactions
that selectively drive changes in tumour size and morphology, and which are notably mediated
by the mechanical status and elasticity of the tumour microenvironment. Emergence of
invasive behaviour through growth instabilities at the tumour–host interface will be presented
considering both reaction–diffusion and mechano-cellular models. In the latter part of the
review, patient-oriented implications of tumour growth modelling are outlined in the context of
brain tumours. Some conceptual views of the adaptive strategies and selective barriers that
govern tumour evolution are presented in conclusion as potential guidelines for the
development of future models.
(Some figures in this article are in colour only in the electronic version)
This article was invited by Professor G Gillies.

Contents
1. Introduction 2 4. Control of tumour growth by mechanical factors:
2. Biological background underlying tumour growth continuum mechanics based models 13
modelling: a brief overview 4 4.1. Modelling the effect of pressure within the
2.1. Tumour growth as a deregulation of the tumour and associated cell velocity field 13
interplay between intrinsic cellular programs 4.2. Modelling the role that stress responsiveness
and environmental constraints 4 may play in controlling tumour growth 15
2.2. Contribution of active biophysical processes 5. Morphological instabilities at the tumour/stroma
to tumour growth: cell migration and cellular interface and tumour invasion 16
forces 5 6. Tumour–host tissue interactions and tumour
vascularization 18
2.3. Transduction of physical signals 6
7. Multiscale modelling of tumour growth 19
2.4. Tumour-associated angiogenesis 8
8. Tumour growth models incorporating anticancer
2.5. In vitro models of tumour growth: MTSph 9 treatments: model-driven evaluation and
3. Modelling avascular tumour growth 10 optimization of therapeutic designs 20
3.1. From temporal to spatially structured models 10 9. Conclusions and perspectives 22
3.2. Modelling tumour growth sustained by cell Acknowledgments 25
migration in response to environmental cues 12 References 25

0034-4885/09/056701+30$90.00 1 © 2009 IOP Publishing Ltd Printed in the UK


Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Used acronyms regulated events in growing tumours (Tlsty and Coussens


2006).
EC endothelial cell Thus, while cancer is still currently viewed as a disease
ECM extracellular matrix involving irreversible genomic changes affecting intrinsic
EGFR epidermal growth factor receptor cellular programs, one has to consider that such genomic
MMP Matrix Metaloproteinase alterations act in combination with modifications of extrinsic
MTSph multicellular tumour spheroids programs taking place in environmental conditions defined
TIMP tissue inhibitor of metaloproteinase by inflammation, immune response, matrix metabolism and
VEGF vascular endothelial growth factor stiffness, gradients of signalling molecules, tissue oxygenation
and vascular status, which all combine and underline tumour
development (Kassis et al 2001, Lunt et al 2008). In the
1. Introduction
case of solid tumours, which are considered in this paper,
these interactions include signalling through cell adhesion
Tumorigenesis, i.e. the process of initiation and progression
molecules, such as cadherins and integrins (Hynes 2002a),
of a tumour, is a multifactorial process, surely emerging
and host tissue remodelling through the action of enzymes,
from molecular and genetic abnormalities (Albertson et al
such as plasmin or metalloproteinases (Egeblad and Werb
2003, Balmain et al 2003), but also strongly dependent on
2002), as well as differential cell response to growth factors
cell–cell and cell–extracellular matrix (ECM) macroscopic
and signalling molecules (Balkwill 2004, Coussens and Werb
regulations and interactions (Bissell et al 2002, Ruiter et al
2002, Kulbe et al 2004), all of which act together to
2002, Vincent and Gatenby 2008). It is known that a
control processes such as proliferation, survival/apoptosis,
tumour (from the Latin verb tumere, meaning to bulge or
migration and invasion. Recent research in cell and tumour
to swell) originates from the transformation of normal cells
biology notably highlighted that a wide range of cellular
into cancer cells (carcinogenesis), which acquire the ability and molecular processes are tightly controlled by mechanical
to become unresponsive to normal physiological controls factors depending on cell cytoskeleton and ECM mechanical
(Radisky et al 2001). But this simple statement embraces the properties (Butcher et al 2009, Tilghman and Parsons 2008,
extremely diverse and complex reciprocal dialogues that cells Zaman et al 2006). It is of particular interest that the expression
and molecules comprising a tumour engage through highly of many of the genes involved in biological processes quoted
regulated interactions favouring malignancy. above can be affected by both the biochemical and biophysical
In the past 25 years, a majority of cancer studies have conditions defining the tumour microenvironment, including
focused on examining functional consequences of activating hypoxia and mechanical stresses (Huang and Ingber 2005,
and/or inactivating mutations in critical genes implicated Liotta and Kohn 2001, Lunt et al 2008, Scheurer et al 2004).
in cell cycle control. These studies have taught us a In this context, modelling of tumour growth is a rather
great deal about the functions of tumour promoter genes old research subject (Araujo and McElwain, 2004, Byrne et al
(oncogenes) and tumour suppressor genes, as well as on 2006), but which neither stops getting increasing relevance
the signalling pathways regulating cell proliferation and because of the challenge arising from considering (i) on the
cell death (apoptosis). However, such studies have often one hand the exponential accumulation of data produced
ignored the fact that cancers are heterogeneous cellular entities by the development of new techniques from molecular
whose growth is dependent upon reciprocal and dynamical biology within all components of the ‘omics’ family, from
interactions between genetically altered cells and the time- genomics, epigenomics and proteomics to metabolomics
varying microenvironment in which they live. Especially, (Jones and Baylin 2007, Nielsen and Oliver 2005) (ii) on the
and in view of further modelling approaches, it is worth other hand, the necessity of conducting models development
underlining that pathological features observed during cancer in an integrative or system biology framework in order
progression are still close enough to crucial physiological to counterbalance the limitation of reductionist approaches
processes. Indeed, these series of multistage events involving relying only on molecular biology studies, since they are
cellular and tissular mutual influences also orchestrate organ insufficient to tackle systemic aspects of tumour development
morphogenesis during embryonic development and tissue (Anderson and Quaranta 2008, Bissell et al 2005, Bizzarri et al
repair occurring in wound healing. Thus, tumour growth 2008, Ingber 2002).
is often presented as a ‘wound that does not heal’ (Bissell Early models of tumour growth were based on kinetic
and Radisky 2001, Chang et al 2004, Dvorak 1986, Kalluri models of cell proliferation, and then mostly investigated from
and Zeisberg 2006), illustrating the paradoxical aspects of a mathematical point of view (Araujo and McElwain 2004).
tumorigenesis and the subtle transition existing between However, an evolution toward a more physical approach of
physiological and pathological contexts. Thus, stromas, i.e. tumour growth has taken place in the last ten years (Preziosi
the arrangement of cells and connective tissue at sites of 2003). Notably, theoretical models have been improved
wound healing and tumour growth, share many characteristics by increasing consideration of (i) the biomechanics of the
(Chang et al 2004, Dvorak 1986). Other biological processes, tumour and its microenvironment, (ii) the coupling of tumour
including cell migration, tissue remodelling and formation growth with the tumour vascularization process, i.e. tumour
of new vasculature, which are crucial and normal features angiogenesis (Chaplain et al 2006, Roose et al 2007). The
utilized during tissues repairing and healing, are also critically evolution of tumour growth models has concomitantly coped

2
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

with the complexity and non-intuitive end products originating


from multifactorial and mostly nonlinear biological processes,
which require the development and renewal of concepts in this Traction
field (Gatenby and Maini 2002, Nagy 2005, Potter 2001, 2007,
Quaranta et al 2008, Vincent and Gatenby 2008).
It is known that one obstacle that biophysical models of
living systems must overcome is the multiplicity of the time and Proliferation
length scales, from molecular and intra-cellular processes (e.g. activated Adhesion
progression through the cell cycle, signal transduction, etc) to stroma
tissular processes, especially microenvironment remodelling Intratumoral
and vascularization. At a microscopic scale, tumour models pressure
would suitably describe malignant transformation of normal
cells and associated alterations of cell–cell interactions and ECM
Tumour
tumour heterogeneity. At a macroscopic scale, models must Deformation
stiffness
consider cell response to gradient fields of diverse origins,
such as concentration gradients of diffusible or non-diffusible Migration
molecules as well as strain and stress gradients generated by
the growing tumour mass. Thus, one might at least expect Extracellular
that biophysical models of tumour growth will be able to stiffness
(i) cope with physical phenomena that differ in nature but are
still highly connected, (ii) gather in a manageable way the
most relevant data provided by multimodal tumour imaging
and by biophysical measurements of tumour and tumour- Figure 1. Schematic representation of avascular tumour/host tissue
dynamical interactions through coupled cellular and biophysical
microenvironment properties, (iii) provide explanations and processes. Notably, all processes contribute to the modulation of the
predictions helpful for the design and optimization of therapies mechanical stresses within the tissue, stroma and tumour. Arrows
targeting tumour development. highlight that both normal and tumour cells dynamically adapt to
Before considering more precisely the set of a priori such stresses and strains by modifying their behaviour and
relevant data in this modelling perspective, it seems worth remodelling their microenvironment. Thus, the indicated circular
feedback loop might increase ECM compaction and density around
starting with simple outlines that may shed light on the the tumour (encapsulation) or conversly contribute to tumour cell
progressive evolution of the models reviewed in this paper. detachment and disease progression.
One should first observe that the growth of solid tumours
is physically a mechanical process according to which a et al 2009, Huang and Ingber 2005, Olson and Sahai 2009,
heterogeneous tissue expands within a surrounding medium. Peyton et al 2007). A third element underlying this schematic
Expansion is controlled by some internal driving stresses, modelling framework of solid tumour growth is the recognition
which are counterbalanced by the mechanical resistance of the rather large range of physical properties exhibited by the
provided by the surrounding tumour environment (figure 1). biological objects themselves, i.e. the diversity and specificity
As a first implication, the interface of the tumour and its of the biomechanical properties of individual cells (Suresh
environment appears as a region of remarkable interest since 2007), of tumours (Netti et al 2000, Venkatesh et al 2008), and
complex phenomena could develop there, especially through of the tumour microenvironment and host tissue (Humphrey
instabilities which may break a uniform and compact tumour 2003). Therefore, a rather large spectrum of mechanical and
front into the wavy and irregular morphologies that can be biological responses to endogenous and exogenous stimuli has
observed experimentally, and which dramatically correlate to be expected.
with increased tumour aggressiveness (Weigelt and Bissell Keeping in mind these introductory remarks, this paper
2008). reviews some of the representative contributions to the
A second modelling guideline is the recognition of the modelling of solid tumour growth. We will focus mainly
biological specificity of the tumour growth, which is driven on continuous approaches, extended to some recent hybrid
by both physical and active cellular processes. Typically, continuous–discrete models coupling physical processes to
internal stresses are mostly generated by cell proliferation cellular automaton or multiagent-based description of cell
dynamics, influenced by the diffusion of nutrients within the behaviour. Owing to the enormous body of journals and
tumour. Similarly, the physical forces existing at the tumour publications devoted to the analysis of solid tumour growth,
front are not just force balance equations involving tumour no such review could be comprehensive. Nevertheless, we
surface tension and the resistive pressure of the surrounding tried to provide a concise overview, illustrating from selected
medium. Active cellular traction forces mediate cell adhesion representative papers and contributions of research groups
and migration in this interface region (figure 1) (Gordon how the field of tumour growth modelling has evolved from
et al 2003, Mierke et al 2008). These mechanical forces earlier mathematical description to recent models more firmly
reshape the tumour microenvironment, either directly by grounded on the physical and mechanical aspects sketched
structural changes enforcing medium anisotropy, or indirectly above. Thus, special attention will be given to most recent
by activating mechanosensitive cellular processes (Butcher studies and their capacity to account for extended sets of

3
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

in vitro and in vivo data, while underlying the continuity or types present in the tumour microenvironment, including
breakthrough with regard to earlier models. The main issues, fibroblasts, immune cells and cells lining blood vessels.
orientations and perspectives will be highlighted without going These cells are embedded in irregular tissue architecture
into the details of the models formulation, which can be found exhibiting an abnormal microcirculation, with a loss of
exhaustively in the corresponding quoted references. vascular hierarchy and large variability in vessel diameter,
The outline of the paper is as follows: in the continuity of length and tortuousness.
this introductory part, section 2 reviews the main components Since cancer is still largely considered as a genetic
of the increasing corpus of biological processes and data that disease rather than as a system biology disease (Bizzarri et al
have supported and inspired the evolution of tumour growth 2008), much research has focused on genetic and molecular
models, from earlier studies to more recent works. Section 3 properties of the tumour cells themselves. Nevertheless, such
presents the leading aspects of these earliest models, largely genomic alterations act in concert with several biological and
devoted to the study of avascular solid tumour growth and biophysical processes, which modify local tissue homeostasis
which assign to the diffusion of nutrients a critical role in by stimulating cell proliferation, motility and differentiation,
determining tumour architecture and growth rate. Recent while simultaneously impairing cell death (apoptosis), cell
extensions of avascular tumour models, taking into account adhesion and microenvironment architecture (Kass et al 2007).
more explicitly the influence of the mechanical resistance Thus, hypoxic microenvironment of a tumour modifies the
introduced by the tumour microenvironment, are presented expression of a number of genes contributing to cancer cells
in section 4. Modelling of the dynamics at the tumour–host survival and growth, and to metastatic efficiency (Keith and
tissue interface is developed in section 5, emphasizing the Simon 2007, Scheurer et al 2004). Tissue vascularization,
prominent role of the morphological instabilities that may immune response and ECM turnover depend on reciprocal
be generated at the tumour front beyond some bifurcation interactions between normal and cancer cells, between cells
threshold. Section 6 extends this analysis by considering and soluble molecules, and between cells and ECM (Tlsty and
architectural modification of the tumour environment due to Coussens 2006, Weigelt and Bissell 2008). The mechanical
tumour angiogenesis. The development of a vascular network stresses experienced by tumour cells and tumour stroma may
within the tumour vascularization is a multiscale problem that equally affect cell phenotype and the expression of several
is presented in section 7, in association with a review of genes through mechanotransduction processes (Butcher et al
hybrid models of tumour growth. Tumour growth models are 2009, Huang and Ingber 2005, Liotta and Kohn 2001, Lunt et al
developed with the hope of being able to predict and optimize 2008, Scheurer et al 2004), as further detailed in section 2.3.
tumour response to treatment. These aspects are reviewed in Globally, solid tumour growth cannot be sustained unless
section 8, where the evolution of tumours in conjunction with the tumour efficiently co-opts inflammatory cells, vascular
different therapies is illustrated in the case of brain tumours. cells and fibroblasts to help reset the balance between ECM
Conclusions and perspectives will be addressed in the final degradation and ECM biosynthesis (Bhowmick et al 2004,
section, focusing on (i) the ability of biophysical models to deal Coussens and Werb 2002, Tlsty 2001).
with alterations of cell and tissue dynamical architecture that Fibroblasts, the predominant cells in the ECM (Bosman
characterize the first critical stages of tumour development, and Stamenkovic 2003), were thought to be passive
(ii) some conceptual evolutions which might stimulate new participants in neoplastic programming of tissues. However,
modelling strategies and (iii) the models clinical value for recent data indicate that they exert an active role: they are able
improvement and new design of therapeutic strategies against to develop significant mechanical forces (Gutierrez-Fernandez
malignancy. et al 2008, Tymchenko et al 2007) and are responsible for
the production of various classes of proteolytic enzymes, their
2. Biological background underlying tumour growth inhibitors, as well as many soluble paracrine growth factors
modelling: a brief overview that regulate cell proliferation, morphology, survival and death
(Kalluri and Zeisberg 2006, Sugimoto et al 2006, Zeisberg
This section reviews some key biological processes and et al 2007). In response to pathologic signals, fibroblasts
experimental data, which give consistence to relevant change their phenotype and function and undergo dynamic
modelling of tumour growth. Without going into excessive changes accompanying tumour progression (Zeisberg et al
detail, the challenge is to provide a concrete background 2007). While multiple cell types contribute to the maintenance
and critical arguments to evaluate the relevance, benefits and of appropriate ECM composition at the invasive front, similar
limitations of the models that will be presented in the following to that observed during wound healing, fibroblasts and vascular
sections. cells are particularly responsible for the new synthesis and
remodelling of ECM components (collagens, fibronectin, etc)
that commonly surround solid tumours.
2.1. Tumour growth as a deregulation of the interplay
It is intuitively clear that inappropriate synthesis
between intrinsic cellular programs and environmental
or degradation of any ECM molecule can alter the
constraints
architecture and the mechanical state of the stroma and
Let us recall that solid tumours are heterogeneous multicellular tumour microenvironment, thus modifying host tissue
entities containing a diverse population of cells, composed integrity. Indeed, the turnover of ECM molecules—synthesis
of genetically mutated cancer cells as well as other cell balanced by degradation— is essential for maintaining tissue

4
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

homeostasis (Wolf et al 2007). When developing neoplasms and its sensitivity to cell–cell biochemical signalling (Fillmore
undergo malignant conversion and acquire the capacity to et al 2003, Chasiotis et al 2003). Since the pioneering
invade normal tissue, increased architectural disorder at the work of Harris et al more than two decades ago imaging the
invasive front of the neoplastic mass is characteristic, with wrinkling of thin elastic substrates by adherent cells (Harris
the balance between ECM synthesis and degradation shifted et al 1981, 1984), analysis of cellular traction forces fosters
in favour of ECM accumulation or fibrosis. Accordingly, the development of a remarkable diversity of experiments.
composition and architecture of ECM varies considerably Indeed, even non-muscle cells, like endothelial cells (ECs)
during tumorigenesis and the newly synthesized peri-tumoral or fibroblasts, remarkably generate active tension through an
stroma is mostly characterized by a disorganized orientation actomyosin filament sliding mechanism similar to the one
of the collagen fibres (Ruiter et al 2002). Importantly, used in the contraction of smooth muscles cells. Importantly,
such increased collagen synthesis and dense accumulation of both normal and cancer cells exert traction forces onto their
fibrillar collagens result in tight encapsulation of malignant own environment (du Roure et al 2005). The qualitative
cells that creates a physical barrier to tumour expansion (Kass and quantitative features of cell traction forces exerted by
et al 2007). different types of cells, including tumour cells, are now
This deregulation toward increased synthesis of extracel- rather well documented (Delvoye et al 1991, Dembo et al
lular components is more or less counterbalanced by enzymatic 1996, Shreiber et al 2003), thanks to the large spectrum
degradation (proteolysis) of pericellular ECM molecules in the of experimental setups developed at different scales. This
tumour neighbourhood, predominantly by matrix metallopro- spectrum includes measurements of isometric tension or
teinases (MMPs). This large family of proteolytic enzymes, compaction rates of viscoelastic biogels contracted by cellular
which normally remodel ECM, are secreted by epithelial forces (Benkherourou et al 2000, Eastwood et al 1996,
cells, activated fibroblasts and inflammatory cells (Coussens Kolodney and Wysolmerski 1992), as well as the tracking
et al 2002, Egeblad and Werb 2002, Sternlicht and Werb of microbeads displacements and of the deformations of
2001). These enzymes are either soluble or plasma membrane- substrates with printed micro-lithography patterns (Balaban
associated and play key roles in wound healing and in cancer et al 2001, Beningo et al 2002, Oliver et al 1999). More
progression (Egeblad and Werb 2002). Interestingly, these recently, several micro and nanotechnology devices, designed
enzymes regulate tumour development in several ways: they as discrete deflectable elements such as micro-cantilevers
were originally described as cleaving ECM substrates, but sev- (Galbraith and Sheetz 1997) or elastic micro-posts (du Roure
eral recent studies indicate they could also have a surprising et al 2005, Tan et al 2003), allow force quantification from
and counterintuitive protective role (Gutierrez-Fernandez et al direct relations between the element deflection and the force
2008). As in many enzymatic networks, specific inhibitors applied on it. The reported magnitude of forces is in the order
control MMPs activity, namely the tissue inhibitors of met- of 10 nN, corresponding to traction stresses in the range 2.5–
alloproteinases (TIMPs) (Lafleur et al 2003). Globally, the 5.5 nN mm−2 (du Roure et al 2005). However, even if such
expression of many MMPs and TIMPs, and thus ECM remod- efficient cellular machinery may potentially promote tumour
elling, is regulated at the transcription level by a variety of growth through tumour cell migration, we still largely ignore
growth factors, cytokines and chemokines, but also at a post- at this stage how it may become effective in the environmental
transcriptional level in specific cases (Clark et al 2008, Fu context prevailing in the tumour neighbourhood.
et al 2008). Thus, tumour growth takes place in a highly dy- In addition to random migration, it is known that
namic landscape that develops a multifactorial environmental cells can exhibit directional migration, or taxis, in response
response to tumour development. to environmental gradients of both soluble chemicals
(chemotaxis) and substrate-attached molecules (haptotaxis)
2.2. Contribution of active biophysical processes to tumour (Hsu et al 2005, Lamalice et al 2007, Lecaudey and Gilmour
growth: cell migration and cellular forces 2006, Weiner 2002). In addition, it has been shown more
recently that ECM mechanical properties also influence cell
Tumour cells migration is obviously a determinant process migration (mechanotaxis) (Kass et al 2007, Zaman et al 2006).
favouring solid tumour expansion. Let us recall that cell Importantly, cells are able to discriminate between soft and
migration involves a series of highly coordinated events, rigid ECM (Alexander et al 2008), then developing a net cell
including the extension of cell membrane protrusions, the motion in the direction of higher ECM rigidity (Zaman et al
formation of new adhesions, the development of traction forces 2006), a phenomenon known as durotaxis (Lo et al 2000).
and the release of previous adhesion to ensure cell body Furthermore, architectural properties of ECM are of critical
translocation (Sheetz et al 1998, 1999). In the context of importance for directional cell migration. Indeed, cellular
cancer cell migration, special attention has been paid to thin shape, orientation, adhesion and migration can be guided by
actin-rich protrusions, termed invadopodia, that colocalize the topography of the microenvironment (figure 2). Such
high proteolytic activity and cytoskeletal signalling pathways topographic guidance of cell migration has been evidenced by
to cell–ECM contact points. the alignment of cells migrating on micro-machined grooves
All these processes received increasing attention from in the substrate or on micropillars (Saez et al 2007, Tzvetkova-
the biologists and physicists. Thus, observations of the Chevolleau et al 2008).
nanodynamics of invadopodium by atomic force microscopy Taken together, these few experimental findings indicate
revealed the remarkable directionality of this cell projection that many possible regulatory loops may differentially affect

5
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

(a) (b) (c)

Figure 2. Compilations of 2D cell trajectories exhibited by 3T3 fibroblasts migrating on polydimethylsiloxane (PDMS) flat substrate (a)
and on PDMS micropatterned with lines oriented horizontally and having an equal width and inter-linear space of 750 nm (c). The
departure of each cell trajectory has been shifted to the origin (0,0), with distance given in micrometers. Cell trajectories are random over
the flat substrate (a), while lines promote a cell guidance phenomenon and induce a preferential cell migration in their direction (c). This
directional migration occurs in pace with a highly polarized morphology of the migrating cells (b). Scale bar in (b) is 20 µm (from
Tzvetkova-Chevolleau et al 2008).

the dynamics of normal and cancer cells, and thus the switch (figure 4) through which control and adaptation of cell
expansion of a solid tumour. For example, cellular forces may response to mechanical stresses and strains will operate (Chen
induce the re-organization of the tumour microenvironment 2008, Geiger and Bershadsky 2002, Geiger et al 2001, Wang
from a diffuse quasi-isotropic to an anisotropic fibrillar ECM et al 2007).
assembly, thus modifying ECM and stroma architecture, and Thus, cellular tension drives cell adhesion and focal
thus exerting feedback onto cell migration and associated adhesions assembly, growth and maintenance depend on
tumour growth (Spencer et al 2007). In this case, mechanical forces applied to them (Smith et al 2008).
mechanosensing processes have a leading role in the Such adaptive behaviour was investigated using extracellular
determination of the cellular response. substrates of different mechanical stiffness, with studies
reporting that cells organize their cytoskeleton and adhesive
2.3. Transduction of physical signals contacts differently on soft and stiff surfaces (Alexander et al
2008, Collin et al 2006, Geiger et al 2001). In the light of the
Transduction of physical signals is known to regulate a large
proposed mechanosensory switch behaviour, the tension acting
array of physiological processes, from hearing and touch to
on the adhesion sites of cells attached to a flexible substrate
bone and vasculature shaping (Jaalouk and Lammerding 2009).
may be smaller than when cells are spread over a rigid surface.
Therefore, it is not surprising that failure of mechanosensitive
Consequently, dimensions of focal adhesions formed on soft
processes contributes to cancer if tumour cells, already
elastic substrates are smaller than those formed when cells are
stimulated by various chemical signals, additionally sense
and respond to the mechanical cues provided by mechanical attached on a stiffer ECM (Balaban et al 2001, Riveline et al
interactions with their extracellular environment (Bissell et al 2001).
2002, Sottile 2004). More precisely, it appears clearly that According to these experimental data, the interplay
ECM provides not only the necessary structural support for between mechanical forces and tissue growth and remodelling
cell migration as presented previously, but is also part of the appears as a central process in the modelling of tumour
deformable continuum able to transmit signals through cell– growth, with mechanotransduction, i.e. the conversion of
surface adhesion sites (Chen 2008, Geiger et al 2001, Li et al mechanical force into biochemical information (Huang et al
2005). Such adhesion regions correspond to different clusters 2004, Li et al 2005, Orr et al 2006), being a key function
of proteins, ranging from dot-like adhesion to larger focal for understanding how tumour cells and tumour growth are
adhesions (Hirata et al 2008). Cell adhesion at these sites affected by mechanical constraints. Since this structural
is accompanied by a remodelling of the cytoskeleton network continuity is ensured up to the cell nucleus and the nuclear
into densely packed straight fibres made of actin filaments. matrix (Wang et al 2009), the tensional state of ECM and
Specific transmembrane receptors like integrins, a large family tumour stroma may influence cell dynamics by modifying
of heterodimers which differ by the combination of their two cellular phenotypes and expression of mechanosensitive genes
subunits (α, β) (Hynes 2002b), ensure a physical continuity (Gieni and Hendzel 2008, Orr et al 2006, Wang et al 2007).
and dynamic links between extracellular environment on one The ECM and tumour mechanical status depends on several
side of the plasmic membrane and the complex molecular factors, originating from intracellular contractile machinery,
bridges that connect the cytoskeleton actin filaments to the from external loads due to mechanical activity of neighbouring,
cytoplasmic domains of integrin subunits on the other side from resistive stresses exerted by the host tissue on the growing
(figure 3) (Berrier and Yamada 2007, Gieni and Hendzel 2008). tumour as well as from movement of fluids within tumour
These scaffolds define a multi-component mechanosensory interstitium (Netti et al 2000).

6
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Transmembrane
receptors

Cytoskeleton
stress fibres
nucleus

ECM Focal adhesion

σcell
Actomyosin
Focal contractility
Integrins
adhesion Transmembrane
Cytoskeleton
receptors
tension

β σext
α
ECM tension

Figure 3. In adhesive microenvironment, here simplified as a 2D substrate (upper figure), normal and cancer cells can generate, transmit and
respond to inside–out and outside–in cues. This signalling process involves the anisotropic distribution of cytoskeleton stress fibres (bundles
of actomyosin) that are anchored into focal adhesions. Fibres contraction promotes the clustering of transmembrane receptors (integrins)
and the accumulation of proteins that make up the focal adhesion complex. Schematic close-up of a focal adhesion (lower figure) illustrates
that the balance of external and internal forces drives the cell response to ECM tension and stiffness cues.

environment through bi-directional (outside–in/inside–out)


signalling feedback schemes. As a mechanical extension
of the homeostasis concept, a cornerstone of physiological
regulations, the tensional homeostasis concept, and its
deregulation in cancer, was invoked more recently as a
relevant background for analysing tumour growth (Butcher
et al 2009, Paszek et al 2005). Both concepts are linked,
the latter one qualifying how the tumour microenvironment
specifies a contextual information framework for both normal
and cancer cells to determine the correct (or incorrect)
response to superimposed biochemical and biomechanical
stimuli (figure 4). Dynamic reciprocity usually involves
autocrine and paracrine regulatory loops (Bussolino et al
2006, Lee et al 2007). For example, when carcinomas
Figure 4. The balance of internal and external stresses at a develop, epithelial cells can initiate incorrect signalling,
mechanosensor, like focal adhesion, drives the tensional resulting in stromal cell production of growth factors that,
homeostasis experienced by the cell and the subsequent up or down in turn, stimulate inappropriate proliferation of the secreting
regulation of fundamental cellular functions such as proliferation, cells themselves (autocrine feedback) or of neighbouring
adhesion, migration or differentiation. The stress balance depends cells (paracrine regulation). Focusing on mechanoreciprocity
on both active elements (intracellular contraction or traction exerted
by neighbouring cells) and passive components (cystoskeleton and (Butcher et al 2009) while keeping for biomechanical
ECM stiffnesses). Then, softening or hardening, i.e. remodelling of regulations a similar conceptual framework, it may be
these viscoelastic components, directly affects the local tensional useful to introduce autobaric and parabaric pathways through
homeostasis. which cells influence themselves and each other through
mechanical signals (Banes et al 1995, Tracqui 2006). Thus,
These considerations underline that the development and autobaric processes define a self-enhancement of the cell
relevance of biophysical models of tumours have to be stress state by the cell’s own contraction, as illustrated in
examined in the light of different concepts. A first one the mechanosensitive development of focal adhesions quoted
is the dynamic reciprocity concept proposed by Bissel and previously. Parabaric regulatory loops may be initiated by a
co-workers (Bissell and Aggeler 1987, Nelson and Bissell cell increasing its mechanical load over neighbours’ cells by
2006) to qualify the mutual interactions of cells and their short-range mechanical deformation transmitted by the ECM.

7
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

VEGF, ...
Angiogenic
vessel

Blood vessel
VEGF,... Avascular
tumour

EC
Endothelial proliferation EC Tumour
cells (EC) and sprouting chemotaxis angiogenesis

Figure 5. Tumours induce blood vessel growth, i.e. angiogenesis, by secreting various growth factors like vascular endothelial growth factor
(VEGF), which binds to receptors present on ECs membrane. ECs then release proteases, escape from the original parent vessel walls
(sprouting) and migrate by chemotaxis toward the tumour where they form new vessels. Single tumour cells can break away from the
tumour mass and enter one of these blood vessels, being then carried to a distant site where they can initiate the growth of a secondary
tumour (metastasis).

To what extent biophysical models of tumour growth already (Gharaee-Kermani and Phan 2001, Gruss et al 2003). These
consider these diverse regulatory loops will be presented in factors are highly expressed in developing tumours, as they are
the following sections. One should however stress that it is during physiologic wound healing. Availability of diffusible
often difficult to isolate and analyse separately such regulatory angiogenic inhibitors and stimulators is however limited, as
loops, since they evolve together within a ECM that changes they are either sequestered to ECM molecules or tethered
temporally as an adaptation to varying cell responses (Nelson to cells via membrane anchorage domains. Extracellular
and Bissell 2006). proteases like MMPs regulate the release of these factors,
rendering them available for interaction with receptors on
2.4. Tumour-associated angiogenesis vascular cells and thus activating the development of tumour-
associated vasculature. Intuitively, MMPs were previously
For maintaining tissue homeostasis and influx of oxygen thought to facilitate tumour expansion by merely degrading
and nutrients, formation and activation of blood vessels are ECM structural components, thereby allowing a cleared
essential during all tissue repair and growth processes as path for migrating tumour or ECs. In fact, recent studies
well as during cancer development (Avraamides et al 2008, indicate that proteolytic degradation of ECM by MMPs
Cavallaro and Christofori 2000, Folkman 2002, Nyberg et al release molecules that act as positive or negative regulators of
2005). When a primary tumour expands, the development of tumour-associated angiogenesis, depending on host tissues and
a new blood vasculature is required to sustain the metabolic development stage of the tumour (Bergers et al 2000, Coussens
needs of this rapidly growing cellular mass and to ensure et al 2002, Lafleur et al 2003).
the elimination of waste products. This is accomplished by Such considerations of biochemical regulatory pathways
activation of pre-existing vascular networks, a process referred do not explicitly account for the morphogenesis of the
to as angiogenesis, by opposition to vasculogenesis that defines tumour-associated angiogenic vasculature, which is distinctly
the de novo formation of new vessels (Carmeliet 2003). tortuous and inherently unstable. In fact, while considerable
During angiogenesis, a well-orchestrated series of events information is known concerning the diverse assortment
occurs, encompassing EC proliferation and further directional of positively and negatively acting soluble and insoluble
migration of these cells through remodelled basement factors regulating angiogenesis, less is known about the
membrane and stroma (figure 5). ECs respond to the role of mechanical factors in the onset and modulation of
angiogenic stimuli released by inflammatory cells and by the vascular network morphogenesis. Significant insights into
expanding populations of cancer cells constituting the tumour the mechanobiology of angiogenesis have come from the
(Folkman 2003). Further recruitment of perivascular support development and analysis of in vitro assays using cultured
cells enables stabilization of nascent vessels, functional ECs (Vailhe et al 2001). These assays, referred to as
lumen formation and appropriate blood flow. Thus, tumour in vitro angiogenesis or tubulogenesis, mimic quite well the
angiogenesis is a multifactorial process, regulated in part by early morphogenesis of pseudo-capillaries networks, during
local changes in the relative balance between soluble and which ECs cultured on ECM may organize as pseudo-
insoluble either pro- or anti-angiogenic mediators, which capillaries around the meshwork of growing areas devoid
impact on EC proliferation, differentiation, migration and 3D of ECM, the lacunae. Such patterning of ECs have in the
organization into tubular structures with functional lumen. last ten years attracted the attention of experimentalist and
Pro-angiogenic polypeptide growth factors comprise most theoreticians by the intriguing fact that they appear to follow
notably different forms of VEGF (Lee et al 2007), basic self-organizing rules (Gamba et al 2003, Namy et al 2004,
fibroblast growth factor (FGF) and tumour necrosis factor TNF Tosin et al 2006, Tracqui 2006). Several types of intervening

8
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

(a)
(b)

Figure 6. Adhesion of transformed EAhY926 ECs onto the surface of Cytodex© microbeads (diameter ∼200 µm) mimics the existence
of outer rim of cells shedding from the tumour surface and interacting with the surrounding ECM. When focusing on one of these
microcarriers (a), significant displacements (arrows (a)) of the viscoelastic fibrin matrix underlying the microcarrier can be reconstructed
from time-lapse videomicroscopy sequences, revealing the field of traction forces generated by the cells. Interestingly, for soft enough fibrin
gels, ECs self-organized into circular cellular chords (arrows (b)). This experimental feature illustrates how vessels morphogenesis could be
sustained and modulated in a tumour microenvironment through stroma stiffness and cell–ECM mechanical interactions.

pathways have been identified, among which are cells–ECM A clear limitation of the microcarrier assay is that cell
mechanical interactions. Indeed, ECM may sustain the dynamics and interactions are limited to the microcarrier
early morphogenesis of this capillary-like structures network surface. This is not the case with multicellular tumour
by providing positional cues, by mechanically influencing spheroids (MTSph), the most common in vitro assay developed
microvessel cell behaviour in a given range of ECM stiffness to mimic solid tumour growth. Since a large number of the
(Ghajar et al 2008, Stephanou et al 2007, Vailhe et al 1997) biophysical models reviewed in this paper have been developed
and by transmitting mechanical loads imposed to neovessels and discussed with respect to this assay, we briefly present in
during angiogenesis (Krishnan et al 2008). the next paragraph the properties and major aspects of this
Interestingly, in vitro assays conducted with microbeads experimental model.
can illustrate how such self-organizing phenomenon could
be induced from the outer rim of cells shedding from 2.5. In vitro models of tumour growth: MTSph
the tumour surface. Following a previously described
in vitro microcarrier-based angiogenesis assay (Nehls and Many types of mammalian cells can aggregate into 3D
Drenckhahn 1995), we cultured transformed ECs (EAhY926 structures named multicellular spheroids (figure 7). Since
cell line) derived from human umbilical vein endothelial monolayer cultures of tumour cells cannot represent the
cells (HUVECs). EAhY926 cells were allowed to migrate characteristics of three-dimensional solid tumours, MTSph
onto microcarrier beads, generating an outer rim of cells. are well established in experimental cancer research as 3D
Then, microcarrier beads are laid down onto a fibrin gel structures of intermediate complexity (Mueller-Klieser 1997,
and the outgrowth of the ECs is recorded by time-lapse 2000, Sutherland 1988). One major advantage of MTSph is
videomicroscopy. Figure 6 illustrates two key features of this their reproducible concentric arrangement of the different cell
experiment. First, the centripetal displacement of the fibrin gel populations found in tumours microregions. MTSph are thus
toward each microcarrier reveals the existence of significant excellent in vitro models for studying cell chemosensitivity, as
cell traction forces generated by cells progressively shedding well as intrinsic and acquired forms of resistance to anticancer
from the microbead (figure 6(a)). Second, the migration of agents. Structurally, the cross-section of MTSph reveals,
cells from the outer bead surface, combined with cell–cell starting from the spheroid centre (figure 8): (i) an inner necrotic
and cell–fibrin matrix mechanical interactions, result in a self- core, which is largely variable depending on the cell type
organized morphogenesis of a lacuna within the fibrin gel, and on the culturing conditions, with a first layer of hypoxic
surrounded by a circular chord of cells (figure 6(b)). quiescent cells, (ii) an intermediate well-oxygenated zone
This morphogenetic process occurs for a given range of with quiescent cells, (iii) a well-oxygenated outer rim with
fibrin gel stiffness, in agreement with data obtained from usual actively proliferating cells (Mueller-Klieser 2000). MTSph
in vitro angiogenesis assays where ECs are initially uniformly are usually classified into three groups according to the
seeded onto the fibrin gel surface (Stephanou et al 2007, Vailhe relative emergence of necrotic and hypoxic regions (Mueller-
et al 1997, Vernon et al 1992). This feature also illustrated Klieser 2000). However, other factors may influence MTSph
how the sensitivity of tumour-derived ECs to the elasticity properties, as illustrated by the fact that not all malignant
of the tumour microenvironment may contribute to normal or cells can form MTSph. Moreover, MTSph can hardly be
abnormal development of the local microvasculature (Ghosh vascularized when co-cultured in vitro with ECs, while they
et al 2008). become vascularized following implantation into animals.

9
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

(a)

Tumour volume (mm3)


(c)
(b)

Figure 7. Growth of multicellular spheroids of liver epithelial cell line proliferating in soft agar gel and visualized at different days Ji after
cells seeding (a). (b) provides a close-up view of one spheroid at day 11. When inoculated subcutaneously into nude mice, these cells grew
as a tumour. The tumour growth curve exhibits a typical shape (c), with a plateau corresponding to the inhibition of tumour expansion (see
section 3.1). (Courtesy of A Bessard and G Baffet, INSERM U522, Rennes.)

R(t) by embedding MTSph in extracellular matrices, like agar


Large (figure 7) or collagen, with tunable composition and stiffness.
proliferating Low oxygen
cells level Thus, Helmlinger et al (1997) investigate the influence
of external mechanical loading on MTSph by conducting
experiments where carcinoma and sarcoma cells were growing
in an agarose matrix. The matrix stiffness was tuned by
varying the volume fraction of the solid component during
nutrients the preparation. The spheroid growth was then constrained by
Necrotic resistive stresses fields with different magnitude. Experimental
centre results showed that the size reached by the growing spheroid
decreases as the stiffness of the gel increases. Concomitantly
High oxygen to the reduction of spheroid size, decreasing apoptosis rate and
level increasing cellular packing were observed.
Biophysical models of tumour growth took the benefit
Small
proliferating cells of such direct experimental analysis of structure to function
waste products
properties, where the proliferative and metabolic properties of
cells in spheroids are related to their radial position. Thanks
Figure 8. Schematic cross-section of a multicellular spheroid. to their well-defined spherical geometry, theoretical analysis
Within the spherical shape, a concentric arrangement of large of MTSph growth may use simple, radially symmetric or
proliferating tumour cells is found in the periphery, while smaller
non-proliferating cells concentrate in deeper regions. Diffusional asymmetric models that will be repeatedly encountered in the
constraints to oxygen and nutrients supply increase with spheroid different following sections, considering notably mechanical
size and a central necrotic core emerges as a result of cell death for a properties of MTSph in section 4 and irregular MTSph
certain spheroid radius R. This value, which largely depends on the morphologies explained by shape instability scenarios in
cell type and on cell culture conditions, is one of the main variables section 5.
considered in MTSph growth models.

3. Modelling avascular tumour growth


This indicates that MTSph properties are determined both
3.1. From temporal to spatially structured models
by intrinsic properties of cells and by cell–cell and cell–
microenvironment interactions. The modelling of avascular tumour growth is a first step
These considerations stimulate the design of experiments toward an integrated description of tumour development and
for mimicking tumour stroma influence. Evolution of the metastasis. The earliest and simplest models described the
above standard MTSph outgrowth assay has been proposed changes over time of the number of cancer cells within a

10
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

solid tumour, with cell proliferation following an unlimited and death. The main assumption (and limitation) of such
exponential growth law or more realistically Verhulst’s models is to consider for simplicity the growth of a spherical
(logistic), von Bertalanffy or Gompertz’s growth laws (Afenya tumour. This symmetry condition allows the determination
and Calderon 2000, Kozusko et al 2007a, Marusic et al 1994a, of the tumour boundary expansion from the one-dimensional
Retsky et al 1990). The latter include slowdown and inhibition movement of the outer tumour radius R(t) in the radial
of tumour’s evolution, due to contact inhibition between direction (0, r) as the solution of the integro-differential
packed cells and deprivation of environmental resources. In equation (Byrne and Chaplain 1996a):
addition to these standard models, alternative descriptions have
been recently proposed (Bru et al 2003, Delsanto et al 2004,  R  R
d 3
Kozusko and Bourdeau 2007b), looking for some universality (R ) = G(n, . . .)r 2 dr − A(n, . . .)r 2 dr, (2)
dt Rnc 0
in the characterization of the growth process. Restricted to
the temporal aspects of tumour growth, these models reduce where the functions G(n, . . .) and A(n, . . .) model indirectly
to a single ordinary differential equation whose solution can
cell proliferation and cell death, respectively, with Rnc being
be successfully compared with experimental growth curves
the radius of the necrotic core. As growth proceeds, a
exhibited by in vitro growing MTSph (figure 7(c)) or in vivo
multilayer architecture emerged within the tumour from the
growing tumours (Marusic et al 1994b, Retsky et al 1990).
imbalance between supply and demand of nutrient. Thus,
However, as pointed out in Araujo and McElwain (2004),
such models are essentially mathematical descriptions, with such models are simple and successfully reproduced the
model parameters hardly related to physical processes. This multilayered architecture of MTSph cultured in vitro, with an
limits their explicative power, even if they can be extended outer rim of nutrient-rich, proliferating cells, a central core of
to deal with tumour heterogeneity and the existence of sub- nutrient-starved, necrotic debris and an intermediate region of
populations of cancer cells (polyclonality) emerging from nutrient-poor, quiescent cells, as described in section 2.5. In
genetic mutations and characterized by different proliferation addition, analytical and quantitative predictions are possible
rates and sensitivity to environmental or chemical cues. regarding (i) the time at which the necrotic core of the
In order to understand more precisely the influence idealized tumour occurs, (ii) the size of the tumour at the
environmental conditions have on tumour growth, notably onset of necrotic core formation, which depends on the model
nutrients delivery and acidity levels, spatially structured parameters values, notably the diffusion coefficient of the
reaction–diffusion models have been developed as moving nutrient.
boundary problems, in which the position of the outer tumour Generalization of this diffusion-consumption scenario to
boundary is unknown (Ferreira et al 2002, Smallbone et al other diffusing molecules (hydrogen ions, glucose, lactate,
2005). The generic physical driving processes that will etc) and growth factors within reaction–diffusion models
be commonly encountered in models are the diffusion of allows a more refined description of the control exerted
nutrients and oxygen, provided by the surrounding host tissue by the host microenvironment onto the growing tumour
and consumed by tumour cells. Since oxygen and nutrient (Casciari et al 1992, Kelly et al 2008, Smallbone et al
availability limits cell proliferation, the expected outcome of 2005). Thus, Gatenby et al (2006) explored an acid-mediated
such models is the definition of conditions leading to the tumour invasion hypothesis, according to which acidification
appearance of necrotic and hypoxic regions. When the tumour of the tumour microenvironment is toxic to normal cells, but
is small, nutrient availability stimulates the cell proliferation. advantageous to cancer cells, thus triggering the transition
As the tumour expands, cells located in the central part of
from benign to malignant tumour growth. Their model
the tumour are progressively starved of nutrient. Cell death
consists of three coupled reaction–diffusion equations for
increases, generating a necrotic core in regions when nutrient
cancer cells, normal cells and pH, and simulates the death
falls below some critical values (figure 8).
of normal cells induced by an increasing acidity generated
As a simplification, tumour cell density is no longer
by neighbouring cancer cells. The latest extension of this
explicitly considered as a variable of these spatial models. The
nutrient concentration n(r, t) and the tumour radius R(t) are model gives a still more accurate description of the role
the generic variables, with tumour cells proliferation assumed of acidosis in tumour growth by considering the respective
proportional to the local nutrient concentration. From a formal contribution of active and quiescent cells (Smallbone et al
point of view, nutrient concentration is given by a reaction– 2008). Additionally, Swietach et al (2008) used diffusion–
diffusion equation. Coupled to the diffusion of nutrient reaction modelling to explore the potential of cell membrane
(diffusion coefficient Dn ), the rate of nutrient consumption enzyme for controlling the gradients of intracellular pH within
depends on the tumour radius R(t) to account for tumour a tumour, a more uniformly alkaline tumour favouring further
heterogeneity (figure 8), leading to the simple mass balance growth.
equation These models, incorporating a limited number of
∂n biological processes and simple geometries, do not account
= Dn n − f (c, R). (1)
∂t for the observed irregular shape of multicellular spheroids or
Tumour growth is given by a differential equation, describing tumours. This consideration stimulated the development of
the rate of change of the tumour volume from the balance models trying to include cell motility processes and cellular
between the nutrient-dependent rates of cell proliferation response to environmental cues.

11
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

3.2. Modelling tumour growth sustained by cell migration in and to the chemical gradient signal ∇g(r, t), with Jg =
response to environmental cues αm∇g. The coefficient α defines the cell’s sensitivity to
the diffusible chemoattractant. In a similar way, active cell
Modelling tumour growth in connection with the invasion
migration by haptotaxis, i.e. up to ligand-density gradients (or
of tumour cells into the host tissue is a major challenge,
adhesivity gradient) is modelled by considering an haptotactic
since the switching on or off of this migratory stage has a
cell flux Jh . In a first approximation, this flux may be
dramatic influence on the induction of the metastasis process.
considered proportional to both the local cell density and
We know from section 2.2 that direction and speed of
the ECM density gradient ∇e(r, t). Assuming that the
cells migration depend on environmental stimuli, especially
sensitivity of the cell response to this gradient is monitored
gradients of soluble chemicals (chemotaxis), gradients of
by a coefficient h (haptotactic cell sensitivity), the associated
substrate-attached molecules (haptotaxis) or ECM mechanical
cellular flux is described by the phenomenological expression
properties (mechanotaxis), including gradient of matrix
Jh = hm∇e. Figure 9 illustrates that such modelling of
rigidity (durotaxis) and ‘substrate guidance’ monitored by the
haptotactic cell migration, in combination with cellular forces,
topography and anisotropy of the microenvironment (figure 2).
can generate a self-enhanced accumulation of cells in specific
However, spatio-temporal evolution of such directional
regions of a deformable medium. More generally, the above
migration processes may become hardly predictable when the
tumour microenvironment continuously varies in composition expressions of cellular fluxes can be modified by considering
as a result of growth factors and ECM turnover, or in that chemotactic or haptotactic coefficients are no longer
organization because of ECM deformations induced by cellular constant parameters, but rather functions that depend on cell
traction forces. A crucial property of biophysical models is density or on microenvironment composition and structure, as
then their ability to determine whether the coupling between illustrated above for cellular diffusion fluxes.
tumour microenvironment evolution, cellular forces and active The above theoretical expression of cellular fluxes has
cell migration will sustain tumour growth or, conversely, might been considered in a large class of continuous differential
prevent its expansion (figure 1). tumour models coupling reaction–diffusion equations with
Theoretical models have incorporated different degrees chemotactic and possibly haptotactic cell migration processes.
of complexity to account for cell motility, either passively in Analysis of travelling-wave solutions exhibited by reaction–
a diffusive manner or actively in response to external cues. migration models provides a theoretical basis for studying
Mathematical differential expressions of some associated in 1D domains tumour cells infiltration in the surrounding
cellular fluxes are briefly introduced below. A rather tissue in different contexts (Marchant et al 2000, Sherratt and
exhaustive presentation of such models can be found in the Chaplain 2001). In 2D domains, consideration of cell motility
seminal books of J D Murray (2002, 2003). Random cell in addition to competition for nutrients among normal and
motility is classically described by cellular diffusion, in which cancer cells was proven to give rise to a wide range of tumour
the diffusive cell flux Jd is proportional to the local cell morphologies. Thus, Ferreira et al (2002) derived from a
gradient ∇m(r, t) at location r, i.e. Jd = −D∇m, where nutrient-limited growth model a phase-like diagram exhibiting
the cellular diffusion coefficient D can be identified from the the transition from compact and circular morphologies to
analysis of cell tracking data (figures 2(a) and (c)) based on papillary and finger-like tumour patterns. Considering a
random walk or persistent random walk models (Rosello et al 3D-geometry, Stein et al (2007) modelled the invasion of
2004, Selmeczi et al 2005). Instead of considering a constant collagen gels by glioblastoma tumour spheroids. The model
diffusion coefficient, some models of tumour growth assumed is designed to describe the dynamics in two distinct regions
that this parameter is a function D(m) of the cell density (self- of the dispersive spheroid: the central core contains cells
reinforcement of cell motility) or depends locally on the ECM that proliferate rapidly but move slowly; the invasive rim
properties. We especially reported in section 2 that cell traction contains cells that proliferate slowly but are highly motile, with
forces induce architectural reorganization and anisotropy of radially directed motility. The model simulates the growth and
the fibrous tumour microenvironment at the invasive front. morphologies of both the core radius and the invasive radius,
In agreement with experimental observations (Korff and which compare favourably to experiments on the patterns of
Augustin 1999), such influence of dynamic reshaping and growth and dispersion conducted with wild type and epidermal
distortion of the microenvironment onto cellular diffusion can growth factor-transfected tumour cell lines. Interestingly,
be modelled by a strain-dependent diffusion tensor D(ε) here the authors reported that in two-dimensional radial migration
ε is the Cauchy’s infinitesimal strain tensor (Cook 1995). The assays, transfected cells exhibit a higher dispersion rate than
cellular diffusive flux Jd then reads Jd = −∇ · [D(ε)m]. wild-type cells, in agreement with observations made in animal
However, the most common directional cell motion invoked models and with the reported implication of epidermal growth
when modelling tumour growth remains cell chemotaxis, factor activity in the increased aggressiveness of many cancers.
with cells moving up (more rarely down) gradients of a But surprisingly, the wild-type cells are more invasive than
diffusible molecule (growth factor, cytokine, etc) like VEGF the transfected ones in the 3D spheroid/collagen assay, which
(Lee et al 2007, Selmeczi et al 2005). Cell chemotaxis points out the different influences of 2D and 3D extracellular
has notably a pivotal role in modelling the reaction of the environments on cell phenotype and behaviour (Martins and
host immune system to tumour cells (Adam and Bellomo Kolega 2006). In this case, wild-type cells would have a better
1997). In the simplest models, the chemotactic flux of ability to reshape the collagen matrix, forming preferential
cells Jg is assumed proportional to the cell density m(r, t) migration tracks which could increase directed cell motility.

12
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Figure 9. Finite-element simulation of the 2D haptotactic migration of cells on an ECM exhibiting a bumpy variation of density.
(Left) Initial spatial profile of ECM density, slightly patterned by the superimposition of initial cell density (quasi-homogeneous distribution
with slight random fluctuations around a normalized unit density value). (Right) Haptotatic migration of cells over the ECM gradient,
combined with ECM deformation generated by cell traction forces, lead to the inhomogeneous steady-state shown on the right, with
accumulation of cells in the central part of the ECM.

Such differences plead for a more accurate modelling of describe constitutive stress/strain relationships within the
directional cell migration. Interestingly, a recent work of tumour.
Gerisch and Chaplain (2008) introduced a refined analysis of
cell–matrix adhesion by considering a sensing region for cells, 4.1. Modelling the effect of pressure within the tumour and
with cell velocity being enhanced in the direction where cells associated cell velocity field
can form the most bonds within the sensing region.
Following the work of Greenspan (1976), the role of cell–
cell adhesion as a physical constraint regulating tumour
4. Control of tumour growth by mechanical factors: compactness was addressed by several groups (Byrne and
continuum mechanics based models Chaplain 1996b, Cristini et al 2003). In these models, cell–
cell adhesion was introduced by considering surface tension
Including mechanical factors as control parameters for tumour at the tumour outer rim as a control parameter for tumour
growth is a conceptual change with respect to the above reshaping during growth. Tumour growth itself is driven by
modelling frameworks focusing on cell population dynamics the intratumoral pressure gradient ∇p generated by localized
modulated by diffusible molecules. As outlined previously, cell proliferation. Such gradients induce motion of tumour
tumour growth is fundamentally a biomechanical process cells, away from the proliferation regions (high pressure) and
with multiple facets, ranging from inherent balance between towards regions where cell death predominates. However, cell
proliferative pressure and resistive force of surrounding host density does not appear explicitly as a model variable and a key
tissue to the far more complex processes of mechanical control variable remains as usual the nutrient concentration delivered
of cells dynamics and tumour vascularization. Then, tumours by the microenvironment. Its evolution still obeys a reaction–
should not be considered as passive mechanical structures, but diffusion equation, assuming additionally that a steady-state
rather as dynamic active media where mechanical signals can distribution has been reached, since the time scale of the
be transduced in different ways to determine the differential growing tumour is much slower than the diffusion time scale
response of normal and cancer cells, respectively (figure 4). of the nutrient. Considering the tumour as an incompressible
Biophysical models that are grounded within a mechanical fluid, it is assumed that cell proliferation and death produce
framework aim at answering questions about the influence a velocity field v linked to the nutrient concentration through
of strains and stresses on the evolution of the tumour the continuity equation ∇ · v = (n), where (n) represents
structural and mechanical properties itself, on the mechanical the net balance between the nutrient-dependent proliferating
interactions between the different types of cells inside and rate and the cell death rate (Cristini et al 2003, Graziano and
outside the tumour, and between these cells and the tumour Preziosi 2007). A constitutive equation is needed to relate the
microenvironment. This section highlights representative velocity field to the internal pressure p. The usual assumption
modelling approaches proposed in this context, from the is to view the tumour as a porous medium through which
earliest models introducing pressure and surface tension of the the tumour cells flow. Then Darcy’s law is used to relate
tumour to more recent approaches dealing with multiphase the velocity v to the pressure gradient, i.e. v = −γ ∇p,
models and introducing diverse strain energy functions to where γ is a positive constant characterizing the hydrostatic,

13
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

viscous-like properties of the tumour cells (Byrne and Preziosi enzymes produced by the tumour itself, the model extension
2003, Macklin and Lowengrub 2007). On the free boundary proposed by Jackson and Byrne (2002) considered that tumour
of the tumour, which moves with velocity v , different types cells produce protease at a constant rate when the pressure they
of conditions expressing tumour compactness are provided experience exceeds a threshold value. In addition, proteases
to close the model formulation. Thus, mean curvature and enhance motility of the cell by weakening cell–cell interactions
surface tension accounting for cell–cell adhesion can be used and adhesion. As expected, protease activity induces capsule
to prescribe boundary conditions on the pressure or on the breakdown and favours tumour expansion.
nutrient concentration (Byrne and Chaplain 1996b). In section 1, we highlighted that the active response of the
A main departure of such models from those assuming host tissue to tumour expansion is similar to a wound-healing
radially symmetric tumour morphologies is their ability to deal response, which, in addition to collagen and extracellular
with the influence of tumour shape and local curvature, and proteins synthesis, can induce differentiation of fibroblasts
thus to cope more realistically with the diversity of irregular into migrating myofibroblasts able to contract the tissue. In
tumour morphologies observed experimentally. Furthermore, an attempt to analyse the role of tumour microenvironment
it becomes theoretically possible to characterize parameter contraction on potential tumour encapsulation in the absence
regions for which the radially symmetric solution becomes of ECM biosynthesis by normal cells, Lubkin and Jackson
unstable to asymmetric perturbations. This represents a (2002) proposed a fluid mechanics approach to discriminate
first basis for understanding the emergence of tumours with between expansive growth and active contraction scenarios.
branching morphologies, as detailed in the next section. On the They consider both the host tissue and the tumour to be
other hand, several limitations of these modelling approaches composed of two interpenetrating material phases, a first
can be pointed out. From a physical point of view, the validity aqueous phase for the interstitial fluid and a second phase
of Darcy’s law to describe cell motion within the tumour consisting of normal and tumour cells embedded in fibrous
may be questionable. Modelling tumours as freely expanding extracellular components. They assume that, on the time
bodies, keeping a relative compactness thanks to cell–cell scale of tumour growth, the second phase behaves as a Stokes
adhesion processes, is also a rather simplified view of the fluid. Still commonly assuming that tumour cell proliferation
real experimental situations, only justified in the case of free is a function of available nutrients concentration, the model
floating growing MTSph. In other cases, a growing mass of introduces two kinds of stresses, the so-called solvation and
tumour cells contends with increasing mechanical resistance contractile stresses, respectively. The solvation stress is a
from the surrounding host tissue, as experienced in vitro by measure of the affinity of one phase for another, with positive
MTSph growing in gels (section 2.5). In vivo, this constrained values if the cell–ECM phase is strongly hydrophilic, and
growth has fundamental implications regarding the formation negative values if the cell–ECM phase is hydrophobic. The
of a dense capsule of extracellular material around the tumour. contractile stress, modelling the potential contraction of the
A crucial issue is then to know under which conditions such host tissue, is proposed as an interaction term proportional to
physical barrier will be sufficient to contain tumour growth and the normal and tumour cells densities. Simulations of this
to prevent dissemination of cancer cells and metastasis. simple three-parameter model indicates that contractility of
In the light of preceding sections, analysis of tumour the surrounding host tissue is not needed for the formation of
encapsulation may invoke two kinds of processes: (i) a passive a capsule, but that this process may be enhanced if host tissue
mechanical response of the surrounding tissue, which is contractility additionally takes place.
simply compressed into a capsule by expansion of the tumour More recently, Guiot et al (2006) developed a rather
(expansive growth hypothesis), (ii) active response of the host different approach by considering a mechanistic description of
tissue, with induction of ECM biosynthesis by normal cells. tumour invasion in the framework of solid inclusion growing
These two processes have been analysed by Jackson and Byrne in an elastic fracturable medium. The transition from a non-
(2002), with spatio-temporal evaluation of cells and ECM invasive growth to invasive patterns takes place when a failure
(the model constituents or phase variables) simulated when stress, corresponding to a threshold for fracture propagation,
both processes are considered to act either independently or is reached. The so-generated cracks provide a mechanistic
synergistically. Stresses inside the tumour include for each analogy for the generation of infiltration paths for tumour
constituent intraphase stresses, depending on the constituent cells within the host tissue. The considered tumour growth
volume fraction, and interphase stresses, including frictional model implements the isotropic growth of a spherical and
drag due to the relative motion between two constituents. linear elastic inclusion inside a linear elastic medium, the
Cell proliferation and matrix production are inhibited at growth rate being inhibited by the interfacial pressure. In the
high cell and ECM densities. The model is furthermore standard case of a linear elastic microenvironment, the model
considered as a closed system, with the sum of all constituents predicts very satisfactorily the time evolution of the volume
being constant. One-dimensional model simulations indicate of non-invasive spheroids of human adenocarcinoma cells
that without ECM production, tumour growth generates thin growing in agarose gels with increasing stiffness (Helmlinger
capsule, without growth suppression. With ECM production, et al 1997). When a fracturable elastic microenvironment
ECM accumulates within the tumour. With both processes is considered, Guiot et al (2007a) also reported a very
acting, tumour is growing and surrounded by a thin and dense good agreement between the model simulations and observed
capsule. Closer to the biological observations, and in order rates of volume growth obtained experimentally with invasive
to analyse how the local host stroma is affected by degrading glioma spheroids (Gordon et al 2003).

14
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

4.2. Modelling the role that stress responsiveness may play in


controlling tumour growth F=Fe G
As underlined previously, solid tumour expansion occurs T=0
T=Tr
within a deformable host medium that mechanically constrains
Initial
the growth process. Quite naturally, biophysical modelling of configuration
tumour growth is thus part of a general framework dealing
with development and influence of stress field on growing G Current
biological tissues (Cowin 2004, Humphrey 2003, Lin and Fe configuration
Taber 1995, Volokh 2006), occurring during both physiological
and pathological morphogenetic processes. T=0
In this context, a rather large diversity of biophysical Virtual
models have been considered, with tumour being considered configuration
as one component of multiphasic medium exhibiting viscous Figure 10. Three states considered in finite growth of an elastic
fluid-like or elastic solid-like properties. Experimental data tumour: the growth tensor G maps the original stress-free (Cauchy
indicate that, according to their cellular origin (carcinoma, stress T = 0) initial configuration into an intermediate virtual
sarcoma, glioma, etc), tumours develop with different configuration (unconstrained growth). Since this stress-free
mechanical properties. Thus, Netti et al (2000) reported configuration may differ from the observed current configuration, an
elastic transformation Fe describes the stress response of the tumour
significant differences in the stress relaxation behaviour of from the natural to the current configuration, tumour mass being
tumours obtained from various human tumour cell lines preserved. The overall deformation gradient F indicates how the
and grafted in mice. When submitted to a sequence of growing tumour is deforming locally when going from the initial to
compressive strain within a confining chamber, the studied the current configuration. The transformation Fe gives rise to a
residual stress Tr in the current configuration.
carcinomas exhibit a viscous behaviour, with almost complete
stress relaxation in between the imposed strains, which
is typical of macromolecular solutions. Conversely, the supplemented by the usual mass balance equations for the
tested glioblastoma and sarcoma display a viscoelastic solid tumour mass and the nutrient, and by the balance of linear
type of behaviour, with sequential increase of stress with momentum, which sums up to divT = 0 when inertial terms
strain. Additionally, a significant nonlinear dependence of the and body forces are neglected. Key points in the model
relaxation phase with strain was observed with glioblastoma formulation are the expression of the growth rate appearing
(Netti et al 2000), which closely resembled the one observed in the tumour mass balance equation and the associated
with extracellular fibrin matrix submitted to similar loading relationship between the growth rate and the growth tensor
conditions (Benkherourou et al 2000). Such tumours thus G. Isotropic growth of a sphere can be modelled with a
behave as structured soft tissues. Thus, these experiments
provide quite relevant quantitative data for modelling tumour growth tensor of the form G = g(t)I , where the identity
growth in a continuum mechanics framework. tensor is multiplied by an explicit function of time g(t). More
In the continuum mechanics approach of Ambrosi and interestingly, and with reference to Helmlinger et al (1997)
Mollica (2004), the tumour spheroid was considered as experiments reporting inhibition of tumour spheroid growth
compressible elastic solid whose increase in mass with time is with increasing stiffness of the surrounding agarose matrix
modulated by nutrients levels. A known modelling difficulty (section 2.5), Ambrosi and Mollica (2004) considered that the
is then to describe simultaneously the change in mass and spherical growth tensor G depends on nutrient concentration
in stresses occurring during growth. The usual approach and on intratumoral stress. Conservation of mass leads
(Rodriguez et al 1994) consists of describing the evolution to a growth model where the function g(t) is given by a
of the mechanical properties of the growing tumour as a differential equation gt = h(trace P , n)g, where n is the
composition of two separate transformations. The first one nutrient concentration and P the first Piola–Kirchoff stress
considers the change in mass due to growth from the initial tensor, introduced to solve the mechanical problem in the
configuration up to an intermediate and virtual, so-called reference configuration. The dependence on stress is modelled
natural configuration, and is described by a growth tensor G through a decreasing exponential function, such that stress
(figure 10). The second transformation describes the stress always inhibits growth. Additional constitutive equations
response of the tumour up to the current configuration. For are prescribed for the mechanical behaviour of the agarose
simplicity, the newly synthesized material is introduced with gel. The mechanical response of this matrix, considered as a
the same stress state as the existing material. A constitutive poroelastic material, is derived from a strain energy function
relationship has to be provided to model the stress response depending both on the third invariant and, exponentially, on the
from the natural configuration. first invariant of the right Cauchy–Green deformation tensor.
By analogy with polymeric foams, Ambrosi and Mollica Corresponding simulations of tumour spheroid growth
(2002) considered the tumour as an isotropic nonlinear showed that in the absence of external loads (floating spheroid),
elastic and compressible material characterized by a strain the change in growth rate was essentially due to the reduced
energy function, from which the Cauchy stress tensor T in availability of nutrient that occurs when the diameter of
the tumour can be derived. This constitutive equation is the spheroid increases. A significant nutrient concentration

15
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

only subsists in a thin layer below the spheroid surface, 5. Morphological instabilities at the tumour/stroma
which may be interpreted as the outer rim of proliferative interface and tumour invasion
cells observed experimentally with MTSph assays. When
the spheroid is growing within the surrounding poroelastic Tumour invasion involves a variety of processes that
medium, its size increases linearly from the very beginning, ultimately lead to cell detachment from the primary tumour
until it tends to plateau. In this case, growth is essentially and infiltration into adjacent tissue and before possible
inhibited by extracellular stresses. Let us remark that an dissemination to secondary metastatic sites with the aid of
additional complexity in the description of MTSph growth the bloodstream system (figure 5). We already approached
is the intrinsic generation of residual stresses, a rather in the preceding section a challenge in modelling tumour
common feature observed with different physio-pathological growth, namely understanding how the propensity of limiting
processes (Alford et al 2008, Ohayon et al 2007), but which tumour expansion by compression and fibrosis of the host
may significantly influence the tensional homeostasis of the tissue may be redirected to the capacity of triggering invasive
growing and remodelled tissue (Alford et al 2008, Ambrosi progression and further metastasis capabilities of tumour cells
et al 2008, Lin and Taber 1995). (figure 1). Considering the invasive branching morphology
While the tumour spheroid model of Ambrosi and Mollica commonly observed in vitro with growing MTSph, a promising
(2004) is one–component, the growth of MTSph embedded view consists of approaching this pattern formation process
in a compressible elastic matrix was alternatively modelled as the result of the amplification of growth instabilities at
by considering a growing structure composed of solid and the tumour/host tissue interface. Thus, the landmark of
fluid phases in interaction. Following Landman and Please tumour aggressiveness would not be solely a localized tumour
(2001), Chen et al (2001) considered the tumour spheroid as a cell proliferation, modifying the tumour’s surface-to-volume
mixture of cells and extracellular fluid characterized by their ratio, but a global switch or bifurcation between ‘smooth
volume fraction and velocity, related through force balance margin’ and ‘fingering protrusions’ surface patterns. In
equations involving hydrodynamic drag force, hydrostatic this framework, biophysical models are expected to shed
pressure and intercellular forces. Growth takes place through light on the selection of invasive patterns by the tumour
cell proliferation and mass exchange between phases, with microenvironment characteristics, i.e. nutrient availability,
dead cells assumed to transform into fluid phase. The growth factors activity and ECM properties.
spherically symmetric spheroid then consists of a necrotic
We outlined in the preceding section that 1D models
region, resulting as usual from diffusion-limited nutrient
focusing on surface tension (Greenspan 1976) or cell–cell
supply, and of a compact but active outer proliferative rim.
adhesion (Byrne and Chaplain 1996b) as the biophysical
The spheroid’s expansion is restrained by the surrounding
processes maintaining the tumour’s compactness exhibit
matrix, still modelled as an infinite hyperelastic isotropic
parameter regions for which the radially symmetric solution
and compressible medium characterized by a strain energy
becomes unstable to asymmetric perturbations. Both
function, which is a slight modification of the strain energy
processes yielded similar qualitative predictions: regular
function proposed by Helmlinger et al (1997): it depends on
tumour growth occurs if surface tension or cell–cell adhesion is
the first and third strain invariants and is similar to the one
strong enough, but irregular, infiltrative structures resembling
used in (Ambrosi and Mollica 2004). As intuitively expected,
the ECM inhibits the tumour spheroid’s growth, but the 1D irregular outer boundaries of tumours emerge if these
simulations of the model allow a quantitative tracking of the parameters are smaller.
sigmoı̈dal pattern exhibited by the evolution with time of the Alternatively, the emergence of non-equilibrium steady
spheroid matrix interface, together with the associated rapidly states as dissipative structures is well documented in reaction–
varying displacement of the necrotic core. diffusion models or in reaction–diffusion–migration models
The biomechanical models presented above can be of pattern formation (Murray 2002, Nicolis and Prigogine
certainly extended by taking into account additional properties 1977). In the original model proposed by Turing in 1952
characterizing tumours mechanical response to extracellular (Turing 1990), the so-called Turing’s instability arises from the
strains and stresses. One may notably consider more precisely coupling between nonlinear reaction kinetics and the relative
how the mechanical confinement surrounding the tumour diffusion of two competing (inhibitor and activator) chemical
mass feeds back onto tumour remodelling through variation species. In reaction–diffusion–migration models, simplest
of mechanical moduli characterizing the tumour properties. scenarios involve cells that produce their own chemoattractant.
Such analyses can be conducted in a multiphase modelling Then a small perturbation in an initially homogeneous
framework (Preziosi and Tosin 2009) or transposed from distribution of cells gives rise to a gradient of chemoattractant
studies developed in other fields, e.g. when analysing stress that enhances the heterogeneity of cell density. On the
adaptation of bone or heart tissue: here tissue stiffness is a other hand, cellular diffusion, as well as chemoattractant
function of microstructures which evolve in time depending diffusion, tends to eliminate such heterogeneities. Clearly,
on the external loading conditions (Cowin 2004, Taber and spatial patterns will be generated if the aggregative process
Chabert 2002, Tozern and Skalak 1988). Aside from pure is greater than the dispersive effects. A similar self-enhance
tumour mass expansion, the mechanical effects of cell traction aggregative effect can be driven by adhesivity gradients
forces onto the alignment and anisotropy of the fibrous tumour (haptotaxis), exhibited by inhomogeneous ECM densities
microenvironment also appear as critical active controls of (figure 9) (Murray 2002, Tracqui 2006). This aggregative
tumour expansion, as developed in the following section. effect is still counterbalanced by cellular diffusion, but since

16
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

u as the driving nonlinearity of the reaction terms. Given


a set of parameter values, which satisfy the conditions for
Turing’s instability, different spatially heterogeneous steady-
state distributions of the two chemicals u and v can be obtained
over the tumour surface. As in rectangular geometries, higher
order unstable modes become excited as tumour grows and
tumour surface becomes larger. This leads to a series of
spatial pre-patterns of growth-inhibitor and growth-activator
concentrations (figure 11) that may be associated with the
progression and asymmetries of tumour morphologies.
Another conceptual approach, based on the principle
of ‘least resistance, most permission and highest attraction’
has been proposed by Deisboeck and coworkers (Deisboeck
et al 2001, Habib et al 2003). In this scenario, cells
follow each other because of reduced mechanical resistance
of the ECM, enhanced haptotactic gradient, and increased
chemical attraction, all as part of a self-organizing adaptive
system. Following these guidelines, Sander and Deisboeck
(2002) analysed and modelled the emergence of branching
pattern around the central core of muticellular glioma
Figure 11. Illustrative mixture of spots and stripes obtained at the spheroids, generated from a mutant cell line in which the gene
surface of a sphere as an asymptotic spatial pattern emerging beyond coding for the epidermal growth factor receptor (EGFR) was
a Turing’s instability. This conceptual background for tumour amplified. The spheroid was cultured between two layers of
morphological instabilities and branching is illustrated here from the
ECM. In addition to random cell motion, the model implements
finite-element simulation of a simple two-variable
reaction–diffusion model including a nonlinear autocatalytic two types of directional migration for tumour cells, called
reaction term. Then, colour scale would correspond to concentration heterotype and homotype chemotaxis, respectively. The first
variations of a potential growth-promoting chemical diffusing on the one refers to the usual chemoattraction of cells in response
surface of a spherical tumour. to gradient of a diffusible nutrient supplied by the tumour
environment. The second one refers to paracrine stimulation,
ligands are bounded to the ECM and do not diffuse, it is only according to which tumour cells produce soluble growth
the ECM viscoelastic properties which can act as a stabilizing factors during their migration. Neighbouring cells are attracted
factor in this mechanical instability scenario (figure 9). by the so-created growth factor gradient, with motile cells
In view of these many possibly different sources of following each other because of this chemotactic process. The
instabilities, several theoretical and computational approaches 2D variation of the concentration of motile tumour cells is thus
have been developed to sustain the view that nonlinear and given by an extended diffusion–chemotaxis transport equation,
self-organizing processes may drive tumour morphological which reads
instabilities, with clusters of cells organized and distributed
non-homogeneously at the tumour edge as a response to a ct = ∇ · (Dc ∇c) − ∇ · (c∇χ (n)n) − ∇ · (cη∇h). (4)
biochemical pre-patterning (figure 11). Thus, in the spirit
Here c(r, t) is the concentration of cells, and n(r, t) is the
of Turing’s pre-patterning theory, Chaplain et al (1995,
nutrient concentration. The coefficient χ (n) is a nonlinear
2001) studied the evolution of the concentrations of growth-
function of n, which defines the sensitivity of cells to
inhibiting and growth-promoting diffusive chemicals, which
heterotype chemotaxis. The last term is the homotype
are produced by the tumour cells on the surface of a growing
chemotaxis, mediated by the concentration h(r, t) of the
spherical tumour. This surface is defined as a thin layer of
paracrine mediator produced by motile cells. Proliferation
proliferative cells, within which the production of the chemical
of cells is assumed negligible in the invasive region, but cell
factors is restricted. By assuming that the two chemical
generation counterbalancing cell shedding is implemented as
species u and v interact nonlinearly through cross activator–
a boundary condition on the spheroid surface. Diffusion–
inhibitor mechanisms, it is known theoretically that linearly
reaction equations are additionally provided for nutrient
stable steady-state values of u and v in the absence of diffusion
and homotype factor concentration, assuming a steady-state
may bifurcate toward unstable solutions when diffusion comes
distribution for the nutrient due to its much faster diffusion.
into play. The growing tumour is modelled as a sphere of radius
A linear stability analysis of a schematic 2D portion of the
R(t), and the reaction–diffusion model is defined as a moving-
region surrounding the spheroid shows that the steady-state
boundary problem that reads
concentration of the growth factor is unstable. Then, a branch
1 1 of invasive tumour cells may extend from the spheroid surface
ut = u+f (u, v), vt = dv+g(u, v), as a result of this intrinsic instability due to the combination
[R(t)]2 [R(t)]2
(3) of heterotype and homotype chemotaxis.
where f (u, v) and g(u, v) are polynomial functions of u and v One should notice that formally, the last term in
incorporating an order-2 autocatalytic formation of chemical equation (4) looks like a haptotactic flux, except that molecules

17
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

of h can diffuse. Tumour cell haptotaxis at the tumour divergence of the tumour cell velocity field v , related to the
surface was proposed in Tracqui (1995) as a possible driving pressure in the tissue by Darcy’s law. Tumour cells and
mechanism for surface pre-patterning of invasive tumours, ECM are treated as comprising porous media with hydraulic
in conjunction with deformation of the tumour–host tissue conductivity, while cell adhesion forces are modelled using
interface by cellular forces. Considering the few layers of an equivalent surface tension at the tumour/tissue interface.
active cells at the tumour surface, the model assumption is Diffusing nutrient and oxygen are supplied by neovasculature
that haptotactic cell movement initiates local cell aggregation, and captured by tumour cells. The onset of morphologic
with an associated local increase in cellular forces amplitude instability and tumour fragmentation is based on the 3D linear
and tumour stroma deformations which positively feedback stability analysis of the reaction–diffusion model, where mass
onto cell motion. If haptotaxis is sufficiently large compared growth and shape evolution of the tumour depend on four
to cellular diffusion, this autocatalytic process can be strong microphysical variables: cell mitosis rate, diffusion length
enough to overcome the mechanical resistance of the tumour L, ratio A of cell death rate over proliferation rate and
stroma, thus initiating and stabilizing the formation of pre- ratio Gm of tumour cell mitosis rate over relaxation rate,
invasive spurs of cancer cells at the tumour surface. where the relaxation rate is associated with cell–cell adhesive
Quite recently, Macklin and Lowengrub (2007) proposed forces and cell motility coefficient introduced in Darcy’s law.
a more refined analysis of the crucial role played by the The bifurcation diagram associated with the model defines
tumour microenvironment. In particular, they show that, in transitions between tumour morphologies as a function of A
a nutrient-poor microenvironment, the only viable invasion and Gm. For small Gm, the spherical shape is stable and
strategy for the tumour is to break into small fragments, which the balance between cell proliferation and death regulates the
penetrate the surrounding tissue. Conversely, under nutrient- tumour size. When too weak cell adhesion forces lead to larger
rich conditions, the tumour behaviour depends mostly on the values of Gm, shape instability may develop. These theoretical
biomechanical property of the medium: a soft, mechanically results are in agreement with observations of MTSph invasive
responsive medium enhances ‘smooth margin’ invasion while, morphologies and support a diffusional instability scenario
in the opposite situation, long invasive fingers are produced, where emergence of irregular tumour morphologies is driven
whose characteristics largely depend on cell-to-cell adhesion. by the differential cell proliferation induced by gradients of
Alternatively, the instability scenario explored by Khain and nutrients and growth factors in the tumour microenvironment
Sander (2006) is based on a dynamical switch of the tumour (Frieboes et al 2006). However, extrapolation to in vivo
cells phenotype. They compared the invasive patterns obtained situations is not straightforward, since tumours in vivo possess
with mutant MTSph, where mutant cells were also obtained by varying degrees of vascularization and exhibit a complex
amplification of the EGFR gene, to the spherically symmetric interface with normal cells and the ECM. This provides a clear
pattern observed with MTSph composed of wild-type, i.e. incitation to develop more complex models including explicitly
non mutated, cells. The model assumptions are that cells tumour vascularization.
have high motility but slow proliferation in the spheroid
neighbourhood, whereas rapid proliferation occurs in the inner 6. Tumour–host tissue interactions and tumour
spheroid region. Thus, Khain and Sander (2006) introduced an vascularization
autocatalytic-type of density-dependent proliferation, leading
to a two-variable nonlinear reaction–diffusion model similar The switch from the slow and relatively harmless avascular
to the one used in Chaplain et al (2001). As known from tumour growth phase modelled above to a rapid vascular
2D-analysis of such nonlinear systems, plane wave solutions growth phase occurs because of tumour vascularization by
can become transversally unstable if the ratio of the nutrient angiogenesis (figure 5). Therefore, biophysical modelling
and cell diffusion coefficients exceeds a threshold value. of tumour growth should be ideally able to couple tumour
Bifurcation of spherical spheroid shape toward developing mass growth to the well differentiated stages of vascularization
branches of tumour cells is then associated with this instability presented in section 2 and which build up tumour vasculature
phenomenon. When considering additionally that ECM from the derivation of existing host tissue vessels (figure 5).
reorganization due to cell/ECM interaction enhances tumour Thus, several approaches, based on continuous (Holmes
cell migration in the radial direction, the simulated developing and Sleeman 2000, Levine et al 2001) or discrete formalisms
branches appear much thinner than in the isotropic diffusion (Anderson and Chaplain 1998, Bauer et al 2007, Chaplain et al
case. Thus, this simple reaction–diffusion model captures 2006, Merks et al 2006), tried to provide a global description of
different in vitro morphologies of mutant and wild-type successive events leading to tumour vascularization, from loss
spheroids. It deserves nevertheless further experimental of interconnection between the ECs and release of proteolytic
validation of the associated model predictions, i.e. that enzymes in response to diffusible angiogenic factors secreted
wild-type cells have a larger diffusion coefficient but a by tumour cells, to capillary sprouts, tips formation and
smaller proliferation rate than mutant cells in the spheroid interconnection into closed loops (anastomosis), until new
microenvironment. capillaries penetrate the tumour (figure 5). In models of
A diffusional instability scenario that takes place during capillary network morphogenesis and adaptation to blood
tumour growth was also considered by Cristini and co- circulation, tumour is usually only viewed as a source of
workers (Cristini et al 2003, Frieboes et al 2006). In their angiogenic factors. We will concentrate here on models
model, the local specific mass growth rate is given as the explicitly coupling the influence of tumour vasculature on

18
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

tumour growth, which have been significantly improved in Interactions between tumour and vasculature take place via
recent years. two concentration fields: the oxygen supplied by the vessel
Cristini et al (2005) related tumour shape and network and growth factors secreted by tumour cells. Each
microvascular density to provide evidence that spatially edge of the generated graph represents a tubular vessel carrying
homogeneous oxygen and nutrient act by suppressing a hydrodynamic blood flow that exerts a shear force upon the
morphological instability. Their model incorporates an vessel walls. Vessel dilatation takes place in regions of large
angiogenesis component as reinforced random walk of new growth factor concentrations. Conversely, weakly perfused
blood vessels drawn into the tumour, and blood vessel vessels can collapse when the hydrodynamic shear force is too
will collapse when the blood-to-tumour pressure difference low. As a result, geometry of the tumour vasculature emerges
approaches zero. The two-dimensional simulations of the in the model as a combination of dense and void regions that
model were interpreted and summarized into a two-parameter might possess fractal properties.
phase diagram summarizing possible tumour morphologies: By coupling tumour growth to tumour angiogenesis,
one parameter is associated with cell adhesion forces, while such models have obviously strong medical implications for
the other one quantifies microvascular density. Model understanding potential failure of anticancer chemotherapy
computations show that even after angiogenesis, the tumour due to insufficient drug delivery to the tumour. They also
maintains a compact morphology because of high cell reinforced the multiscale properties of tumour growth and
adhesion. However, with a smaller value of the cell adhesion the benefit that could be expected by developing models
parameter, a morphologic instability of the tumour mass arises addressing the specificity of biological and biophysical
as the tumour develops. Such computational results can be processes expressed at each level. The next section
discussed in the light of the tortuousness and loss of vascular briefly reviews some representative models developed in this
hierarchy observed in solid tumours (Fukumura and Jain 2007). perspective.
Indeed, ideal angiogenesis will lead to a compact and spherical
tumour mass confined in a limited volume of tissue. In this 7. Multiscale modelling of tumour growth
case, further tumour growth requires additional angiogenesis
to increase nutrient access. In contrast, disordered vascularity A major issue in cancer research, as in other domains
leads to instability and tumour branching since the same of physiopathology, is to handle the enormous amount
tumour mass does not critically require angiogenesis for further of data produced at the molecular, cellular and tissular
growth and thus might extend across a larger region within the levels, both in normal and pathological contexts. Thus,
host tissue. multiscale biophysical models of tumour growth are especially
Recently, the same group (Frieboes et al 2007) important as formal and explicative tools for relating molecular
proposed an extended 3D model of tumour growth processes and gene expression to global tumour morphologies
still focusing on the influence on tumour angiogenesis (figure 12).
and associated nutrients diffusion gradients onto tumour In the Jiang et al (2005) model, avascular tumour
invasion. Their hybrid continuum–discrete model of tumour growth spans three distinct scales. At the cellular level,
angiogenesis is based on convection–reaction–diffusion a lattice Monte Carlo model describes cellular dynamics
equations governing the dynamics of tumour cells, vital (proliferation, adhesion and viability). At the subcellular
nutrients and cytokines. Mechanical interactions of the level, a Boolean network regulates the expression of
ECM with evolving neovasculature are modelled by adhesion proteins that control the cell cycle. At the extracellular
energy and momentum equations based on Darcy’s law and level, reaction–diffusion equations describe the chemical
equilibrium among pressure and elastic forces. The model dynamics (nutrient, waste, growth promoter and inhibitor
predicts gross morphological features of growing tumours, concentrations). This model produces an avascular tumour
with regions of viable cells, necrotic areas and fractal-like that quantitatively mimics experimental measurements in
neovasculature. Interestingly, the model accounts for the multicellular spheroids. Based on the simulations, the model
tumour engulfing and compressing the vessels, thus increasing predicts (i) the microenvironmental conditions required for
necrosis. This result highlights the parabaric feedback tumour cell survival and (ii) the pattern of spheroid growth
loop that affects the angiogenic pathway described above: curves under different external nutrient supply conditions.
growing around capillaries that are usually immature, the Later, Anderson et al (2006) analysed the evolution of
tumour compresses them and might cause their collapse. But tumour morphology driven by selective pressure from the
in turn, this increases hypoxia of the surrounding tissue, microenvironment, thanks to a multiscale model of cancer
which stimulates secretion of angiogenic factors and thus the invasion where cell mutations and interaction with different
formation of new capillaries. types of microenvironment, referred to as homogeneous,
Additionally, adaptation of the vessel network to ‘bumpy’ and ‘grainy’, are considered. Sharing some analogies
circulating hydrodynamic blood flow, with local vessel with the granular solid considered by Guiot et al (2007a),
dilation, is a regulation process increasingly considered both bumpy and grainy media represent density variations of the
during host tissue vascularization and during tumour growth microenvironment taken at a cellular scale or at a macroscale,
(Chaplain et al 2006, Stephanou et al 2005). Thus, Lee respectively. The approach of Anderson et al (2006) is
et al (2006) considered the tumour–vessel system as a based on a hybrid discrete–continuum model in which cells
dynamically evolving graph interacting with tumour growth. are treated as discrete entities, while microenvironmental

19
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

of virtual tumours some practical guidelines based on a


model-driven optimisation of the tumour response to different
types of anticancer treatments, including chemotherapy,
radiotherapy, surgery, etc, while diminishing the unavoidable
side effects of such treatments. Successes and limitations
of theoretical models explicitly including and comparing
anticancer therapies are exemplified here, with special
consideration of brain tumours.
Briefly, the development of brain tumours, after
diagnosis, is routinely recorded by different and accurate
medical imaging techniques (computerized tomography (CT),
magnetic resonance imaging (MRI), positron emission
tomography (PET) (Spence et al 2008), etc). Typically,
serial scans can lead to a quantification of the expensive or
treatment-induced recessive dynamics of the brain tumour
for each patient. Different models have been developed to
analyse the different stages of a brain tumour growth (Murray
2003 chapter 11, Harpold et al 2007). Their biological
Figure 12. Illustration of a multiscale approach of tumour growth
and physical bases do not differ significantly from those
based on a mixed continuous–discrete (hybrid) model. considered in the models reviewed previously. All models
Spatio-temporal variations of continuous extracellular gradient are consider cell proliferation and cell infiltration as fundamental
combined to the probabilities P1–P4 of a single tumour cell located processes, possibly together with the compression of the
at the lattice coordinates (i, j ) to move in one of four directions. brain tissue surrounding the tumour. In addition, geometrical
Each probability may depend on the steady-state value of a protein
or gene network describing the controlled expression of proteins
constraints to brain tumour growth can be accurately taken into
regulating tumour cells adhesion and migration. account in the modelling approaches, thanks to the different
modality of image acquisition than can be used. These
factors are modelled as concentrations. Tumour morphologies constraints encompass anatomical boundaries, either fixed as
emerge as a consequence of the cell collective behaviour, the skull, or displaced and deformed by tumour expansion,
controlled by microenvironmental factors that influence cell like the brain ventricles. Such geometric confinements
mutations. In the model, these mutations occur at the time significantly affect the size and shape of the tumour, notably
of cell division, either linearly through four increasingly if the tumour initiates close to these boundaries. These
aggressive phenotypes, or randomly through a mutation properties were highlighted with continuous tumour growth
jumping process in which cells are allowed to acquire one models, in which tumour growth-permitting regions versus
of one hundred predefined phenotypes. A striking result tumour growth-prohibiting regions are defined through space-
of the random mutation scheme is that, even though each dependent diffusion coefficients (Tracqui et al 1995, Tracqui
phenotype is equally likely to be chosen, only a few, aggressive and Mendjeli 1999). Alternatively, similar conclusions can
phenotypes, are actually selected. Thus, the simulated be drawn using the cellular automaton approach, as recently
growing tumour evolves toward increased aggressiveness, reported by Gevertz et al (2008), with automaton cells defined
characterized by tumour cells exhibiting the shortest lag from the Voronoi tessellation of the considered tissue. A
time to proliferation, the highest production of ECM- benefit of this latter approach is to consider intercellular
degrading proteases and the highest haptotactic migration mechanical stress between cells as a control parameter for the
rate. Interestingly, these phenotypic differences correlate tumour expansion, thus incorporating the mechanical feedback
with emergent tumour morphologies that are affected by the exerted by the environmental confinement on the tumour
microenvironment structural properties: under homogeneous growth.
conditions, tumour morphology is non-invasive with smooth Brain tissue heterogeneity also provides anatomical
margins, as opposed to the invasive morphology with fingering tracks favouring tumour cell dissemination, leading to model
margins simulated when tumour develops in the heterogeneous formulations that explicitly consider the larger cell motility
matrices. Thus, heterogeneous tumour microenvironment in white matter compared with grey matter (Swanson et al
seems to drive cancer cells mutations toward phenotypes 2000), as well as differences in the mechanical resistance
characterized by higher motility and higher haptotactic between both tissues. In the 3D model developed by Frieboes
sensitivity. et al (2007), brain structural properties play a critical role in
the formation of rapidly developing tumour buds. Tumour
8. Tumour growth models incorporating anticancer buds invading the white matter encounter less mechanical
treatments: model-driven evaluation and resistance, leading to the formation of relatively slender
optimization of therapeutic designs invasive fingers, compared with the blunt bulbs invading the
grey matter. The tumour buds rapidly elongate and grow
A major application of tumour growth model development is along the folds of the cortex where the tumour cells are
the expected ability to derive from the dynamics and behaviour highly mobile. Interestingly, the overall tumour shape is also

20
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

(a) (b)

Figure 13. Example of a case study where clinical data were obtained from serial MRI scans taken during the terminal year of a patient with
a glioblastoma and who received radiotherapy treatment. (a) Patient’s MRI showing the brain tumour location, close to the neighbouring
ventricles, (b) simulated 3D constrained growth of the treated tumour (b insert) considering density dependent diffusion model with logistic
(dotted line) or Gompertzian (solid line) nonlinear proliferation rates (Tracqui and Mendjeli 1999). Data points correspond to the tumour
volume measured from patient scans at the time of diagnosis and in course of therapy (black rectangles along the time axis). In each case,
the left part of the curves, i.e. from the time of diagnosis back to the appearance of the first cancer cell, has been obtained by reversing time
in the simulations, thus giving access to a predictive description of the silent (non-observable) phase of tumour growth.

determined by the distribution of vessels within the tumour, interestingly provides a basis for reconstructing pre-diagnosis
with the considered multiscale model providing simulations tumour morphology and associated tumour growth history.
of growing glioma and neovascular morphologies that reflect Indeed, by reversing time in model simulation starting from
histopathology data. diagnosis, it becomes possible to track back the temporal
The severity and poor prognosis of brain cancers is pattern of the glioblastoma development up to the appearance
associated with a still limited understanding of the large of the first cancer cell (figure 13). Such estimations may be
diversity of the kinetics and growth patterns exhibited by used as a quantitative basis to define a range of patient ages
the different types of brain tumours. Thus gliomas, highly where early detection of primary tumour can be planed.
invasive primary brain tumours accounting for almost 50% An additional benefit of brain tumour growth models is
of all brain tumours, are known to grow at a wide range of their ability to evaluate virtually, but on quantitative bases,
possible proliferation and diffusion rates (Alvord 1995). This the comparative effects of different and independent or more
stimulates modelling approaches trying to integrate in a single often combined therapies, in a patient-dedicated way. Special
description tumour growth before and after diagnosis and attention has been paid to post-surgical behaviour of gliomas
therapy. Indeed, once detected, tumours are not left untreated after resection through in silico experiments conducted with
in humans, and the imaged tumour growth results from the virtual gliomas (Swanson et al 2003, 2008a, Woodward
combined effect of initial endogenous tumour development et al 1996). Even if the resection seems optimal from a
and its response to therapy. The ability to distinguish between surgical point of view, i.e. presumably removes almost all
these two components of tumour growth is clearly a modelling the cancer cells, it is indeed well known that the small
challenge, especially when considering that therapy is not number of cells left behind could be sufficient for inducing
only translated into an additional cell death rate, but also a complete re-appearance of the tumour some months later.
modifies intrinsic tumour growth, for example by inducing The recurrence of the tumour can start from the tumour cells
mutations and emergence of new, usually therapy resistant, left behind, and this minority of surviving tumour cells is
mutated subpopulations of cancer cells (Tracqui et al 1995). aggressive enough to drastically reduce patient life expectancy
Since the time of onset of the brain tumour is not known (figure 14) (Swanson et al 2008a, Tracqui and Mendjeli 1999).
and thus the duration of the pre-diagnosis tumour growth Similarly, Alvord and colleagues investigated the response
is not clinically measurable, model simulation starts from a to radiation therapy, focusing on the effect of radiation dose
more or less well-defined initial tumour morphology, which fractionation on gliomas growth (Rockne et al 2009, Swanson
determines a subsequent growth pattern against which the et al 2008b). Thus, simple biophysical models, specified by a
clinical data extracted from the analysis of serial scans may be very limited number of parameters, allow a coherent qualitative
compared. However, if assuming as a first simplification that and quantitative description of the wide spectrum of gliomas
the endogenous tumour dynamical features remain unchanged growth and invasiveness patterns. Moreover, their ability to
after therapy, modelling tumour growth pattern after diagnosis predict quite successfully many aspects of real gliomas growth

21
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

growing tumour, also pointed out unexpected results: collapse


and vessel regression did not decrease drug delivery to the
tumours in their computations, a result implying that this
mechanism would not be the major reason for diminishing
drug delivery in real tumours.
Taken together, these different results highlight the ability
of models in providing predictive guidelines for therapeutic
strategies that, solely or in combination, and in the light of
accurate estimations of expectable survival times, are geared
toward optimization of patient-dedicated therapy.

9. Conclusions and perspectives

It is now becoming evident that modelling tumour growth


is much more complex than it was thought not so long
ago. Earliest approaches tended to consider that models,
developed with regard to the multicellular spheroid paradigm,
already address some key issues on cell proliferation and
Figure 14. Predictive power of models simulating virtual tumour death controlled by availability of diffusible nutrients, with
growth under different therapies. The grey line corresponds to the progressive development of necrosis in solid tumours. It turns
tumour evolution already reported in figure 13 when a radiation out that it is not that simple, as stressed by Mueller-Klieser
therapy was used. The hatched line corresponds to the re-growth of
(2000): the onset of necrosis is not caused by simply one factor,
the tumour after a simulated surgical resection, which had left some
cancer cells behind. With such aggressive brain tumours, such as hypoxia, as it had been widely accepted for many
proliferation and diffusion of these cells from the border of the years, and the development of necrosis in tumour spheroids
resected brain region (insert) lead rather rapidly to a lethal tumour is a complex phenomenon involving other factors, such as
volume (horizontal dashed line), in agreement with clinical data. cell–cell and cell–matrix interactions, expression of growth
Estimation of the expected life expectancy of the patient can be read
factor receptors and adhesion molecules, etc. Thus, even if
directly from the intersection of the simulated growth curve with the
lethal tumour volume. more detailed models of MTSph are still actively developed
invoking diffusion-limited phenomena, other recent models
favours their systematic use as tumour grading tools enabling explore alternative views, allowing necrotic regions to develop
a dynamic identification of biologically distinct groups of under conditions of adverse mechanical stress rather than in
gliomas. regions of low nutrient concentrations (Landman and Please
Biophysical models of brain tumours can also support 2001). In parallel, tumour spheroids models can be revisited
investigation of rather different therapeutic strategies. Cristini and improved for mimicking tumour stroma and vasculature
et al (2005) analysed targeting of brain tumours by (Alajati et al 2008, Lin and Chang 2008, Mueller-Klieser
anti-angiogenic therapy. Whereas it seems clear that 2000).
a direct action against tumour growth was to reduce We highlighted in this review how a transition toward
angiogenesis, the modelling approach they developed suggests a more mature approach of solid tumour modelling has
that such intuitive consideration has to be modulated by been taken this last decade, especially with the increasing
more subtle considerations. Indeed, a properly working consideration of biophysical factors in the control exerted
tumour microvasculature can help maintain compact non- by the tumour microenvironment onto the growth process.
infiltrating tumour morphologies by minimizing oxygen and Indeed, the microenvironment is no longer modelled only as
nutrient gradients. In contrast, anti-angiogenic therapy, by a provider of diffusible nutrients and growth factors, or as a
increasing microenvironmental heterogeneity, may promote medium in which inflammatory and immune processes take
morphological instability, leading to invasive patterns even place, but as a deformable and remodelled tissue, with time-
under conditions in which the overall tumour mass shrinks. varying structural and mechanical properties which participate
The model of Cristini et al (2005) suggests that anti- to the ‘selective pressure’ imposed to cell dynamics during
angiogenesis solely with an end to eradicating a tumour tumour development.
may not be the most prudent course. These conclusions This transition is correlated with the increasing
modify the conventional view on angiogenesis leading to development, in this post-genome sequencing era, of
invasion and metastasis: a well-vascularized tumour may be two research domains at the interface between physics,
morphologically stable due to homogeneous distribution of mathematics and cell biology. The first one is the recognized
oxygen and nutrient supply, while local hypoxia or lack of limitation of the reductionist approach, which, despite
cell nutrients due to inefficient neovasculature leads to regions outstanding progresses made in normal and cancer cells
of heterogeneous cell proliferation and migration, which, in molecular biology, is inadequate when used alone to capture
turn, promotes an invasive tumour morphology (Macklin and the complexity of tumour progression and the onset of
Lowengrub 2007). The model proposed by Welter et al (2008), metastasis (Bizzarri et al 2008). In addition, the overwhelming
implementing a dynamically evolving vessels network within a amount of data collected by molecular biology techniques

22
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

has highlighted the need for a systemic approach of cellular indicate that the development path of a tumour is not unique for
and tissular biology. In this system biology framework, the a given set of genomic parameters, but depends on the initial
tumour microenvironment is now a crucial element of the condition and environmental perturbations experienced by the
cancer puzzle. Thus, models of tumour growth are no longer growing tumour.
limited to an inside transformation in cellular organization, We reviewed in this paper another systemic property of
resulting from mutations and controlled by external supply, several tumour models, namely the emergence of specific
but more globally as an altered signalling context in which instability modes in the spatial distribution of cancer cells
dramatic modifications of the signalling-regulatory pathways at the tumour surface and its interpretation as a bifurcation
originating from the tumour stroma and surrounding host tissue process sustained by intrinsic instabilities. Such a bifurcation
may markedly affect the structure, functioning and growth of scenario is clearly very helpful to predict whether a particular
the tumour (Bissell and Radisky 2001, Bissell et al 2005). tumour type in a given host tissue would exhibit at some
These considerations introduce a second facet of the developmental stage a transition from a compact shape with
system biology of tumour growth, namely the increasingly smooth margins toward a stellate morphology with fingering
recognized and documented importance of the biomechanical or filopodial extensions, the latter being the signature of an
contexts as determinants of the cell signalling pathways. invasive phenotype. The problem is that a rather large spectrum
Notably, mechanotransduction processes profoundly influence of instability mechanisms can trigger the onset of finger-
all aspects of normal and cancer cells behaviour, including like invasive patterns. Existing models mostly considered
proliferation apoptosis, adhesion, migration, secretion, etc diffusive instabilities, either as a simple Turing-like pre-
(Huang and Ingber 2005, Ingber 2002). These elements patterning of the tumour surface alone (Chaplain et al 2001),
highlight the need for tumour models development considering or alternatively nonlinear interactions and development of
the superimposition of both biochemical and biomechanical cellular forces at the tumour–host tissue interface. In these
regulatory pathways. models, the irregular tumour morphology results from a global
In this context, relevant representations of cell, tumours and synchronized patterning of the tumour surface, but one
and host tissues as mechanical entities are necessary for the may ask whether the invasive branching rather results from
evaluation of stress and strains experienced and transmitted a desynchronized activity that takes place at different sites
by the tumour cells. We illustrated in section 4 the diversity across the tumour surface. This pleads for a refined modelling
of models used to describe tumour mechanical properties, of microenvironment influence on transition toward invasive
with tumours visualized as amorphous solids, viscous fluids or types of tumour morphologies. It is known that hypoxia
granular solid (Guiot et al 2007a). Central to an understanding or nutrient deprivation leads to invasive tumour behaviour,
of tissue stress evolution is also the necessity of identifying the but the work of Anderson et al (2006) additionally showed
precise constitutive nature of growing tumour tissues, taking that alterations of the microenvironment, from homogeneous
into account tumour heterogeneity. Different modelling choice to inhomogeneous structure, could produce a change from
can be made, either trying to define a homogenized continuum, smooth, non-invasive margins, to morphologically invasive,
or taking into account specifically tumour heterogeneity, fingering margins.
anisotropy and multiphasic nature. Such requirements for refined biophysical models of
As reviewed here, such biomechanical models already tumour growth feedbacks onto the integrative modelling
help to increase our understanding of necrosis formation in strategy quoted in the introductory part of this section, which
tumour and MTSph development. But improved mechanical tends to consider cancer as a systemic disease and more
models of tumours and cancerous tissue also give qualitative specifically to provide an all-comprehensive model of tumour
and quantitative insights into the regulation of cell adhesion and growth (Anderson and Quaranta 2008). A clear implication
migration. This includes both the subcellular levels, in terms of of this perspective is to address the fundamental problem of
stress transmitted by the cells and controlling cell adhesion, as coupling different phenomena at different scales, attempting to
well as the cellular level, where we underlined how haptotaxis integrate detailed subcellular information to make predictions
can be a key determinant of tumour cell migration, controlled at the tumour scale. Filling the gap between microscopic
by both the gradients of adhesion molecules and gradients of and macroscopic descriptions is a challenge of fundamental
rigidity created within the surrounding matrix by proteases importance for tumour growth modelling, which leaves an
activity and ECM biosynthesis. We additionally highlighted open space for the development of models that has just
that accurate modelling of tumours mechanical properties begun to be explored (Ingber 2008b). One may think
has a profound impact on our understanding of tumour to a juxtaposition of models or to intermediate, so-called
vascularization and on the collapse of tumour blood vessels, mesoscopic models (Anderson and Quaranta 2008) in order
which is of fundamental importance for the administration of to present a unified and integrative model of the entire process
anticancer agents. of tumour development. In this system biology framework,
In a broader modelling perspective, considering the most promising models are likely to be based on hybrid
superimposition, at different scales, of both biochemical and continuous–discrete descriptions and formalisms. Indeed,
biomechanical regulatory pathways enforces the consideration continuum descriptions of tumour growth are supported by
of a tumour as a complex biological system governed the existing bodies of phenomenological and constitutive
intrinsically by nonlinear processes. The potential existence of laws elaborated in agreement with fundamental physical
multiple asymptotic solutions exhibited by nonlinear systems principles, and thus take benefit from the knowledge gained

23
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

in other fields of physics. This aspect has been especially mechanomics as the discipline analysing the variation of
emphasized in the above sections with the various approaches the complete set of mechanical interactions at the cell–
proposed to describe a solid tumour in the framework of ECM interface, i.e. the mechanome, in a context dependent
continuum mechanics, with tumour description based on way. The point is that, to increase their relevance,
analogies with fluid dynamics or well-known mechanisms of biophysical models of tumours should tell us something
fracture mechanics (Guiot et al 2006, 2007b). Additionally, about how genome, transcriptome, proteome, metabolome
the spatio-temporal evolution of the associated vector fields is and mechanome networks and charts evolve with variation
also partly amenable to theoretical analysis, with emerging of physical fields defining macroscopically the environmental
simulated behaviours of the models controlled by a rather context. However, such multiscale models are likely to
limited number of parameters or parameter groupings, as become more and more complicated, with large increase in
illustrated with diffusion-instabilities predictions. Thus, Guiot the model parameters. Then, a risk is that they may succeed in
et al (2007a) defined an invasion parameter (IP) through a supporting partly intuition through computational results, but
formula that identifies the most relevant physical parameters, without giving real understanding and insight into underlying
e.g. the tumour’s surface tension, its radius and the confining biological principles, and without ensuring to get relevant
host tissue pressure, whose control should be the target predictions and exploration of all possible model behaviours.
of dedicated therapies. Eventually, a cancer type, organ A guard against building over complicated models might
site and patient-specific IP may be of significant value for be to keep a functional view of tumorigenesis and malignancy,
diagnostic purposes, as most of this multicellular behaviour as a drift from a morphofunctional field, defined as the
occurs well below the current non-invasive imaging resolution physiological spatio-temporal coherence and regulation of
limits. In the same perspective, Chaplain and Sleeman (1993) cells and tissues organization. This view is closely connected
originally proposed earlier a classification and grading of solid to the earlier concept of epigenetic landscape introduced by
tumours based on strain energy functions. However, we are C H Waddington to deal with cell fate in embryogenesis and
still looking for appropriate constitutive laws for modelling morphogenesis (Waddington 1968). Thus, the appearance
tumours mechanical properties, knowing that these laws may of a tumour indicates a breakdown of this morphofunctional
change with tumour types and grading. field, the organizing properties of the tissue homeostasis
being gradually lost as the tumour develops. Within this
Despite these advantages, the averaging over space
framework, tumorigenesis is no longer studied as a merely
realized in continuum formalisms can hardly account for
deterministic consequence of altered gene function, but rather
the diversity of cellular and subcellular dynamical features
as an emerging response of network-structured interactions.
and genetic or epigenetic regulatory mechanisms exhibited at
For nonlinear systems, these interactions would define the
the individual cell level. Alternatively, discrete models are
number and topologies of each attraction bassin of the phase
perfectly adapted for modelling internal signalling networks
portrait associated to the considered model (Huang and Ingber
within each cell. Thus, hybrid models, in which individual
2006, Huang et al 2005). The dynamic of the model variables
cells are treated discretely but interact with other chemical
in response to the different environmental and endogenous
and mechanical continuum fields, are likely to combine the
stimuli experienced by the tumour cells may correspond to
benefits of both descriptions. Under appropriated formalisms,
transitions between attraction basins (domains), i.e. to the
such models will be able to couple the different spatial crossing of the boundaries (or barriers) that delineate each
scales impacted by the growth process with biochemical and of them. In this spirit, Gatenby and Gillies (2008) recently
biomechanical information passed between multiple scales. developed the concept of adaptive landscape to qualify the
Especially, these multiscale biophysical models should help microenvironmental forces and selection mechanisms acting
to understand how the emerging shift of architectural and at work on tumour cells population during carcinogenesis.
tensional homeostasis of the host tissue is shifted toward Considering that the tumour stroma is modified at critical
malignancy by bifurcation processes taking place within steps during cancer development, they analyse the sequence
interactive complex signalling networks. In addition to the of multiple and self-interacting proliferation barriers that
diffusive or mechanochemical instabilities quoted here, these are overcome in carcinogenesis, emphasizing that multiple
modelling approaches will put into a broader perspective strategies may be used to overcome a tissue barrier to
and larger basis the consideration of tumour branching and proliferation, while adaptation to one barrier may decrease or
invasiveness as instability phenomena. increase the height of subsequent barriers. In this perspective,
Nevertheless, this is clearly an enormous challenge relevance and explanatory power of biophysical models of
when considering the increasing number of components tumorigenesis might be evaluated through their capacity to
to consider within the ‘omics family’ which characterize include one adaptation to every barrier, while exploring
complex functions of many cellular components. In different strategies serving as adaptations to the considered
addition to genomics, transcriptomics and proteomics (Doucet barrier. At the same time, the breakdown of the normal
and Overall 2008), metabolomics has been added more morphofunctional field becomes predominantly explainable
recently, as the discipline that describes ‘the complete set from the analysis of the processes and functions ensuring
of metabolites/low-molecular-weight intermediates, i.e. the the adaptation to the barrier (e.g. adhesion or proteolysis),
metabolome, that vary according to the developmental or rather than from the detailed knowledge of the biological actors
pathological state of the cell, tissue, organ or organism’ coming into play (e.g. the precise integrin or metaloprotease
(Nielsen and Oliver 2005). Similarly, let us add by analogy at work).

24
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

In conclusion, the complexity of the system biology of References


cancer, even restricted to the development of solid tumours,
still overwhelms us, leaving a huge gap between the enormous Adam J A and Bellomo N 1997 A Survey of Models for
amount of experimental information that has been collected, Tumor-immune System Dynamics (Boston: Birkhaüser)
and the explicative and predictive power of theoretical models. Afenya E K and Calderon C P 2000 Diverse ideas on the growth
kinetics of disseminated cancer cells Bull. Math. Biol.
But even if open problems in the study of tumour development 62 527–42
are still numerous, biophysics models have still a lot to offer to Alajati A et al 2008 Spheroid-based engineering of a human
improve our understanding of tumour morphodynamics. We vasculature in mice Nat. Methods 5 439–45
outlined the different ways recent models provide mechanistic Albertson D G, Collins C, McCormick F and Gray J W 2003
Chromosome aberrations in solid tumors Nat. Genet.
insights on tumour growth and vascularization through 34 369–76
quantitative simulations that determine how combinations of Alexander N R, Branch K M, Parekh A, Clark E S, Iwueke I C,
microenvironmental and cellular parameters trigger invasive Guelcher S A and Weaver A M 2008 Extracellular matrix
tumour morphologies. Consideration of the many possible rigidity promotes invadopodia activity Curr. Biol. 18 1295–9
Alford P W, Humphrey J D and Taber L A 2008 Growth and
feedback loops, either autobaric or parabaric, which regulate remodeling in a thick-walled artery model: effects of spatial
biomechanical interactions between normal and cancer cells variations in wall constituents Biomech. Modelling
and the tumour extracellular environment is still largely Mechanobiol. 7 245–62
lacking, but analysing how mechanical stresses acting onto cell Alvord E C, Jr 1995 Patterns of growth of gliomas AJNR Am. J.
Neuroradiol. 16 1013–7
secretory activity come into play through mechanotransduction Ambrosi D and Mollica F 2002 On the mechanics of a growing
processes is a promising orientation for new integrative models tumor Int. J. Eng. Sci. 40 1297–316
(Butcher et al 2009, Macklin and Lowengrub 2007). A long- Ambrosi D and Mollica F 2004 The role of stress in the growth of a
term goal of the modelling effort is the ability to predict which multicell spheroid J. Math. Biol. 48 477–99
Ambrosi D, Guillou A and Di Martino E S 2008 Stress-modulated
combination of the key functional switches (proliferation,
remodeling of a non-homogeneous body Biomech. Modelling
death, degradation, biosynthesis, migration, traction, etc) Mechanobiol. 7 63–76
might reverse the invasive behaviour of individual and specific Anderson A R and Chaplain M A 1998 Continuous and discrete
tumours, based on actual measurements of molecular, cellular mathematical models of tumor-induced angiogenesis Bull.
Math. Biol. 60 857–99
and microenvironmental parameters (Ingber 2008a). A more
Anderson A R and Quaranta V 2008 Integrative mathematical
accessible, short-term goal, of the modelling effort, enhanced oncology Nat. Rev. Cancer 8 227–34
by the percolation of biophysical models in biology and Anderson A R, Weaver A M, Cummings P T and Quaranta V 2006
medicine, is to actively participate to the design of new Tumor morphology and phenotypic evolution driven by
selective pressure from the microenvironment Cell
experimental models. Progress in tissue engineering is
127 905–15
encouraging for the larger development of assays targeting Araujo R P and McElwain D L 2004 A history of the study of solid
tumour–stroma interactions, interactions between tumour and tumour growth: the contribution of mathematical modelling
immune cells and tumour vascularization (Ghajar et al 2007, Bull. Math. Biol. 66 1039–91
Avraamides C J, Garmy-Susini B and Varner J A 2008 Integrins in
Ingber 2008a). Another short-term goal for biophysical
angiogenesis and lymphangiogenesis Nat. Rev. Cancer
models development, with strong implications in health 8 604–17
care, is their use as predictive tools in drug discovery Balaban N Q et al 2001 Force and focal adhesion assembly: a close
and in the management of cancer patients. In addition to relationship studied using elastic micropatterned substrates
Nat. Cell. Biol. 3 466–72
molecular or biochemical studies focusing on cell sensitivity
Balkwill F 2004 Cancer and the chemokine network Nat. Rev.
and chemoresistance, they will contribute to explain from a Cancer 4 540–50
more systemic and virtual approach why some therapies fail Balmain A, Gray J and Ponder B 2003 The genetics and genomics
while others prove to be effective in locally controlling tumour of cancer Nat. Genet. 33 238–44
Banes A J, Tsuzaki M, Yamamoto J, Fischer T, Brigman B, Brown T
expansion. Models already appeared as efficient tools for
and Miller L 1995 Mechanoreception at the cellular level: the
gathering the information on tumour morphology, biology and detection, interpretation, and diversity of responses to
metabolism in conjunction with multimodal high-resolution mechanical signals Biochem. Cell Biol. 73 349–65
imaging techniques. As an adjunctive aid to customize Bauer A L, Jackson T L and Jiang Y 2007 A cell-based model
exhibiting branching and anastomosis during tumor-induced
therapy in individual patients, such practical successes will be
angiogenesis Biophys. J. 92 3105–21
highly encouraging, since even if an increasing effort is being Beningo K A, Lo C M and Wang Y L 2002 Flexible polyacrylamide
made for the understanding complexity of tumorigenesis, substrata for the analysis of mechanical interactions at
elaborating new modelling paradigms that integrate all cell-substratum adhesions Methods Cell Biol. 69 325–39
Benkherourou M, Gumery P Y, Tranqui L and Tracqui P 2000
available knowledge remains a challenge facing us.
Quantification and macroscopic modeling of the nonlinear
viscoelastic behavior of strained gels with varying fibrin
concentrations IEEE Trans. Biomed. Eng. 47 1465–75
Acknowledgments Bergers G et al 2000 Matrix metalloproteinase-9 triggers the
angiogenic switch during carcinogenesis Nat. Cell Biol.
2 737–44
We thank Dr. Bruno Vailhé for assistance with the Berrier A L and Yamada K M 2007 Cell-matrix adhesion J. Cell.
microcarrier-based angiogenesis assay. Physiol. 213 565–73

25
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Bhowmick N A, Neilson E G and Moses H L 2004 Stromal Chen C S 2008 Mechanotransduction—a field pulling together?
fibroblasts in cancer initiation and progression Nature J. Cell Sci. 121 3285–92
432 332–7 Chen C Y, Byrne H M and King J R 2001 The influence of
Bissell M J and Aggeler J 1987 Dynamic reciprocity: how do growth-induced stress from the surrounding medium on the
extracellular matrix and hormones direct gene expression? development of multicell spheroids J. Math. Biol. 43 191–220
Prog. Clin. Biol. Res. 249 251–62 Clark I M, Swingler T E, Sampieri C L and Edwards D R 2008 The
Bissell M J and Radisky D 2001 Putting tumours in context Nat. regulation of matrix metalloproteinases and their inhibitors Int.
Rev. Cancer 1 46–54 J. Biochem. Cell Biol. 40 1362–78
Bissell M J, Kenny P A and Radisky D C 2005 Microenvironmental Collin O, Tracqui P, Stephanou A, Usson Y, Clement-Lacroix J and
regulators of tissue structure and function also regulate tumor Planus E 2006 Spatiotemporal dynamics of actin-rich adhesion
induction and progression: the role of extracellular matrix and microdomains: influence of substrate flexibility J. Cell Sci.
its degrading enzymes Cold Spring Harb. Symp. Quant. Biol. 119 1914–25
70 343–56 Cook J 1995 Mathematical models for dermal wound healing:
Bissell M J, Radisky D C, Rizki A, Weaver V M and Petersen O W wound contraction and scar formation PhD Thesis Department
2002 The organizing principle: microenvironmental influences of Applied Mathematics, University of Washington, Seattle
in the normal and malignant breast Differentiation 70 537–46 Coussens L M and Werb Z 2002 Inflammation and cancer Nature
Bizzarri M, Cucina A, Conti F and D’Anselmi F 2008 Beyond the 420 860–7
oncogene paradigm: understanding complexity in Coussens L M, Fingleton B and Matrisian L M 2002 Matrix
Cancerogenesis Acta Biotheor. 56 173–96 metalloproteinase inhibitors and cancer: trials and tribulations
Bosman F T and Stamenkovic I 2003 Functional structure and Science 295 2387–92
composition of the extracellular matrix J. Pathol. 200 423–8 Cowin S C 2004 Tissue growth and remodeling Annu. Rev. Biomed.
Bru A, Albertos S, Luis Subiza J, Garcia-Asenjo J L and Bru I 2003 Eng. 6 77–107
The universal dynamics of tumor growth Biophys. J. Cristini V, Lowengrub J and Nie Q 2003 Nonlinear simulation of
85 2948–61 tumor growth J. Math. Biol. 46 191–224
Bussolino F, Valdembri D, Caccavari F and Serini G 2006 Cristini V, Frieboes H B, Gatenby R, Caserta S, Ferrari M and
Semaphoring vascular morphogenesis Endothelium 13 81–91 Sinek J 2005 Morphologic instability and cancer invasion Clin.
Butcher D T, Alliston T and Weaver V M 2009 A tense situation: Cancer Res. 11 6772–9
forcing tumour progression Nat. Rev. Cancer 9 108–22 Deisboeck T S, Berens M E, Kansal A R, Torquato S,
Byrne H and Preziosi L 2003 Modelling solid tumour growth using Stemmer-Rachamimov A O and Chiocca E A 2001 Pattern
the theory of mixtures Math. Med. Biol. 20 341–66 of self-organization in tumour systems: complex growth
Byrne H M and Chaplain M A J 1996a Growth of necrotic tumors in dynamics in a novel brain tumour spheroid model Cell Prolif..
the presence and absence of inhibitors Math. Biosci. 34 115–34
135 187–216 Delsanto P P, Guiot C, Degiorgis P G, Condat C A, Mansury Y and
Byrne H M and Chaplain M A J 1996b Modeling the role of Deisboeck T S 2004 Growth model for multicellular tumor
cell–cell adhesion in the growth and development of spheroids Appl. Phys. Lett. 85 4225–27
carcinomas Math. Comput. Modelling 24 1–17 Delvoye P, Wiliquet P, Leveque J L, Nusgens B V and Lapiere C M
Byrne H M, Alarcon T, Owen M R, Webb S D and Maini P K 2006 1991 Measurement of mechanical forces generated by skin
Modelling aspects of cancer dynamics: a review Phil. Trans. A fibroblasts embedded in a three-dimensional collagen gel
Math. Phys. Eng. Sci. 364 1563–78 J. Invest Dermatol. 97 898–902
Carmeliet P 2003 Angiogenesis in health and disease Nat. Med. Dembo M, Oliver T, Ishihara A and Jacobson K 1996 Imaging the
9 653–60 traction stresses exerted by locomoting cells with the elastic
Casciari J J, Sotirchos S V and Sutherland R M 1992 Variations in substratum method Biophys. J. 70 2008–22
tumor cell growth rates and metabolism with oxygen Doucet A and Overall C M 2008 Protease proteomics: revealing
concentration, glucose concentration, and extracellular pH protease in vivo functions using systems biology approaches
J. Cell. Physiol. 151 386–94 Mol. Aspects Med. 29 339–58
Cavallaro U and Christofori G 2000 Molecular mechanisms of tumor du Roure O, Saez A, Buguin A, Austin R H, Chavrier P, Siberzan P
angiogenesis and tumor progression J. Neurooncol. 50 63–70 and Ladoux B 2005 Force mapping in epithelial cell migration
Chang H Y, Sneddon J B, Alizadeh A A, Sood R, West R B, Proc. Natl Acad. Sci. USA 102 2390–5
Montgomery K, Chi J T, van de Rijn M, Botstein D and Dvorak H F 1986 Tumors: wounds that do not heal. Similarities
Brown P O 2004 Gene expression signature of fibroblast serum between tumor stroma generation and wound healing N. Engl.
response predicts human cancer progression: similarities J. Med. 315 1650–9
between tumors and wounds PLoS Biol. 2 E7 Eastwood M, Porter R, Khan U, McGrouther G and Brown R 1996
Chaplain M A and Sleeman B D 1993 Modelling the growth of solid Quantitative analysis of collagen gel contractile forces
tumours and incorporating a method for their classification generated by dermal fibroblasts and the relationship to cell
using nonlinear elasticity theory J. Math. Biol. 31 431–73 morphology J. Cell. Physiol. 166 33–42
Chaplain M A, Ganesh M and Graham I G 2001 Spatio-temporal Egeblad M and Werb Z 2002 New functions for the matrix
pattern formation on spherical surfaces: numerical simulation metalloproteinases in cancer progression Nat. Rev. Cancer
and application to solid tumour growth J. Math. Biol. 2 161–74
42 387–423 Ferreira S C Jr, Martins M L and Vilela M J 2002
Chaplain M A, Giles S M, Sleeman B D and Jarvis R J 1995 A Reaction–diffusion model for the growth of avascular tumor
mathematical analysis of a model for tumour angiogenesis Phys. Rev. E Stat. Nonlinear Soft Matter Phys. 65 021907
J. Math. Biol. 33 744–70 Fillmore H L, Chasiotis I, Cho S W and Gillies G T 2003 Atomic
Chaplain M A, McDougall S R and Anderson A R 2006 force microscopy observations of tumour cell invadopodia:
Mathematical modeling of tumor-induced angiogenesis Annu. novel cellular nanomorphologies on collagen substrates
Rev. Biomed. Eng. 8 233–57 Nanotechnology 14 73–6
Chasiotis I, Fillmore H L, and Gillies G T 2003 Atomic force Folkman J 2002 Role of angiogenesis in tumor growth and
microscopy measurement of cytostructural elements involved metastasis Semin. Oncol. 29 15–8
in the nanodynamics of tumour cell invasion Nanotechnology Folkman J 2003 Fundamental concepts of the angiogenic process
14 557–61 Curr. Mol. Med. 3 643–51

26
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Frieboes H B, Zheng X, Sun C H, Tromberg B, Gatenby R and Guiot C, Pugno N and Delsanto P P 2006 Elastomechanical model
Cristini V 2006 An integrated computational/experimental of tumor invasion Appl. Phys.Lett. 89 233901–3
model of tumor invasion Cancer Res. 66 1597–604 Guiot C, Pugno N, Delsanto P P and Deisboeck T S 2007a Physical
Frieboes H B, Lowengrub J S, Wise S, Zheng X, Macklin P, aspects of cancer invasion Phys. Biol. 4 P1–6
Bearer E L and Cristini V 2007 Computer simulation of glioma Guiot C, Delsanto P P and Deisboeck T S 2007b Morphological
growth and morphology Neuroimage 37 S59–70 instability and cancer invasion: a ‘splashing water drop’
Fu X, Parks W C and Heinecke J W 2008 Activation and silencing analogy Theor. Biol. Med. Modelling 4 4
of matrix metalloproteinases Semin. Cell Dev. Biol. 19 2–13 Gutierrez-Fernandez A et al 2008 Matrix metalloproteinase–8
Fukumura D and Jain R K 2007 Tumor microvasculature and functions as a metastasis suppressor through modulation of
microenvironment: targets for anti-angiogenesis and tumor cell adhesion and invasion Cancer Res. 68 2755–63
normalization Microvasc. Res. 74 72–84 Habib S, Molina-Paris C and Deisboeck T S 2003 Complex
Galbraith C G and Sheetz M P 1997 A micromachined device dynamics of tumors: modeling an emerging brain tumor
provides a new bend on fibroblast traction forces Proc. Natl system with coupled reaction–diffusion equations Physica A
Acad. Sci. USA 94 9114–8 327 501–04
Gamba A, Ambrosi D, Coniglio A, de Candia A, Di Talia S, Harpold H L, Alvord E C Jr and Swanson K R 2007 The evolution
Giraudo E, Serini G, Preziosi L and Bussolino F 2003 of mathematical modeling of glioma proliferation and invasion
Percolation, morphogenesis, and Burgers dynamics in blood J. Neuropathol. Exp. Neurol. 66 1–9
vessels formation Phys. Rev. Lett. 90 118101–4 Harris A K, Stopak D and Wild P 1981 Fibroblast traction as a
Gatenby R A and Gillies R J 2008 A microenvironmental model of mechanism for collagen morphogenesis Nature 290 249–51
carcinogenesis Nat. Rev. Cancer 8 56–61 Harris A K, Stopak D and Warner P 1984 Generation of spatially
Gatenby R A and Maini P 2002 Modelling a new angle on periodic patterns by a mechanical instability: a mechanical
understanding cancer Nature 420 462 alternative to the Turing model J. Embryol. Exp. Morphol.
Gatenby R A, Gawlinski E T, Gmitro A F, Kaylor B and Gillies R J 80 1–20
2006 Acid-mediated tumor invasion: a multidisciplinary study Helmlinger G, Netti P A, Lichtenbeld H C, Melder R J and Jain R K
Cancer Res. 66 5216–23 1997 Solid stress inhibits the growth of multicellular tumor
Geiger B and Bershadsky A 2002 Exploring the neighborhood: spheroids Nat. Biotechnol. 15 778–83
adhesion-coupled cell mechanosensors Cell 110 139–42 Hirata H, Tatsumi H and Sokabe M 2008 Mechanical forces
Geiger B, Bershadsky A, Pankov R and Yamada K M 2001 facilitate actin polymerization at focal adhesions in a
Transmembrane crosstalk between the extracellular zyxin-dependent manner J. Cell Sci. 121 2795–804
matrix–cytoskeleton crosstalk Nat. Rev. Mol. Cell Biol. Holmes M J and Sleeman B D 2000 A mathematical model of
2 793–805 tumour angiogenesis incorporating cellular traction and
Gerisch A and Chaplain M A 2008 Mathematical modelling of viscoelastic effects J. Theor. Biol. 202 95–112
cancer cell invasion of tissue: local and non-local models and Hsu S, Thakar R, Liepmann D and Li S 2005 Effects of shear stress
the effect of adhesion J. Theor. Biol. 250 684–704 on endothelial cell haptotaxis on micropatterned surfaces
Gevertz J L, Gillies GT and Torquato S 2008 Simulating tumor Biochem. Biophys. Res. Commun. 337 401–9
growth in confined heterogeneous environments Phys. Biol. Huang H, Kamm R D and Lee R T 2004 Cell mechanics and
5 36010–19 mechanotransduction: pathways, probes, and physiology Am.
Ghajar C M, Suresh V, Peyton S R, Raub C B, Meyskens F L Jr, J. Physiol. Cell Physiol. 287 C1–11
George S C and Putnam A J 2007 A novel three-dimensional Huang S and Ingber D E 2005 Cell tension, matrix mechanics, and
model to quantify metastatic melanoma invasion Mol. Cancer cancer development Cancer Cell 8 175–6
Ther. 6 552–61 Huang S and Ingber D E 2006 A non-genetic basis for cancer
Ghajar C M, Chen X, Harris J W, Suresh V, Hughes C C, Jeon N L, progression and metastasis: self-organizing attractors in cell
Putnam A J and George S C 2008 The effect of matrix density regulatory networks Breast Dis. 26 27–54
on the regulation of 3-D capillary morphogenesis Biophys. J. Huang S, Eichler G, Bar-Yam Y and Ingber D E 2005 Cell fates as
94 1930–41 high-dimensional attractor states of a complex gene regulatory
Gharaee-Kermani M and Phan S H 2001 Role of cytokines and network Phys. Rev. Lett. 94 128701
cytokine therapy in wound healing and fibrotic diseases Curr. Humphrey J D 2003 Continuum biomechanics of soft biological
Pharm. Des. 7 1083–103 tissues Proc. R. Soc. Lond. A 459 3–46
Ghosh K, Thodeti C K, Dudley A C, Mammoto A, Klagsbrun M and Hynes R O 2002a Integrins: bidirectional, allosteric signaling
Ingber D E 2008 Tumor-derived endothelial cells exhibit machines Cell 110 673–87
aberrant Rho-mediated mechanosensing and abnormal Hynes R O 2002b A reevaluation of integrins as regulators of
angiogenesis in vitro Proc. Natl Acad. Sci. USA 105 11305–10 angiogenesis Nat. Med. 8 918–21
Gieni R S and Hendzel M J 2008 Mechanotransduction from the Ingber D E 2002 Cancer as a disease of epithelial-mesenchymal
ECM to the genome: are the pieces now in place? J. Cell interactions and extracellular matrix regulation Differentiation
Biochem. 104 1964–87 70 547–60
Gordon V D, Valentine M T, Gardel M L, Andor-Ardo D, Ingber D E 2008a Can cancer be reversed by engineering the tumor
Dennison S, Bogdanov A A, Weitz D A and Deisboeck T S microenvironment? Semin. Cancer Biol. 18 356–64
2003 Measuring the mechanical stress induced by an Ingber D E 2008b Tensegrity-based mechanosensing from macro to
expanding multicellular tumor system: a case study Exp. Cell micro Prog. Biophys. Mol. Biol. 97 163–79
Res. 289 58–66 Jaalouk D E and Lammerding J 2009 Mechanotransduction gone
Graziano L and Preziosi L 2007 Mechanics in tumor growth awry Nat. Rev. Mol. Cell Biol. 10 63–73
Modelling of Biological Materials ed F Mollica et al (Basel: Jackson T L and Byrne H M 2002 A mechanical model of tumor
Birkhäuser) pp 267–328 encapsulation and transcapsular spread Math. Biosci.
Greenspan H P 1976 On the growth and stability of cell cultures and 180 307–28
solid tumors J. Theor. Biol. 56 229–42 Jiang Y, Pjesivac-Grbovic J, Cantrell C and Freyer J P 2005 A
Gruss C J et al 2003 Stroma formation and angiogenesis by multiscale model for avascular tumor growth Biophys. J.
overexpression of growth factors, cytokines, and proteolytic 89 3884–94
enzymes in human skin grafted to SCID mice J. Invest. Jones P A and Baylin S B 2007 The epigenomics of cancer Cell
Dermatol. 120 683–92 128 683–92

27
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Kalluri R and Zeisberg M 2006 Fibroblasts in cancer Nat. Rev. Lunt S J, Chaudary N and Hill R P 2008 The tumor
Cancer 6 392–401 microenvironment and metastatic disease Clin. Exp. Metastasis
Kass L, Erler J T, Dembo M and Weaver V M 2007 Mammary 26 19–34
epithelial cell: influence of extracellular matrix composition Macklin P and Lowengrub J 2007 Nonlinear simulation of the effect
and organization during development and tumorigenesis Int. J. of microenvironment on tumor growth J. Theor. Biol.
Biochem. Cell Biol. 39 1987–94 245 677–704
Kassis J, Lauffenburger D A, Turner T and Wells A 2001 Tumor Marchant B P, Norbury J and Perumpanani A J 2000 Traveling
invasion as dysregulated cell motility Semin. Cancer Biol. shock waves arising in a model of malignant invasion SIAM J.
11 105–17 Appl. Math. 60 463–76
Keith B and Simon M C 2007 Hypoxia-inducible factors, stem cells, Martins G G and Kolega J 2006 Endothelial cell protrusion and
and cancer Cell 129 465–72 migration in three-dimensional collagen matrices Cell Motil.
Kelly C, Smallbone K and Brady M 2008 Tumour glycolysis: the Cytoskeleton 63 101–15
many faces of HIF J. Theor. Biol. 254 508–13 Marusic M, Bajzer Z, Freyer J P and Vuk-Pavlovic S 1994a
Khain E and Sander L M 2006 Dynamics and pattern formation in Analysis of growth of multicellular tumour spheroids by
invasive tumour growth Phys. Rev. Lett. 96 1881031–4 mathematical models Cell Prolif. 27 73–94
Kolodney M S and Wysolmerski R B 1992 Isometric contraction by Marusic M, Bajzer Z, Vuk-Pavlovic S and Freyer J P 1994b Tumor
fibroblasts and endothelial cells in tissue culture: a quantitative growth in vivo and as multicellular spheroids compared by
study J. Cell Biol. 117 73–82 mathematical models Bull. Math. Biol. 56 617–31
Korff T and Augustin H G 1999 Tensional forces in fibrillar Merks R M, Brodsky S V, Goligorksy M S, Newman S A and
extracellular matrices control directional capillary sprouting Glazier J A 2006 Cell elongation is key to in silico replication
J. Cell Sci. 112 3249–58 of in vitro vasculogenesis and subsequent remodeling Dev.
Kozusko F and Bourdeau M 2007b A unified model of sigmoid Biol. 289 44–54
tumour growth based on cell proliferation and quiescence Cell Mierke C T, Rosel D, Fabry B and Brabek J 2008 Contractile forces
Prolif. 40 824–34 in tumor cell migration Eur. J. Cell Biol. 87 669–76
Kozusko F, Bourdeau M, Bajzer Z and Dingli D 2007a A Mueller-Klieser W 1997 Three-dimensional cell cultures: from
microenvironment based model of antimitotic therapy of molecular mechanisms to clinical applications Am. J. Physiol.
Gompertzian tumor growth Bull. Math. Biol. 69 1691–708 273 C1109–23
Krishnan L, Underwood C J, Maas S, Ellis B J, Kode T C, Mueller-Klieser W 2000 Tumor biology and experimental
Hoying J B and Weiss J A 2008 Effect of mechanical boundary therapeutics Crit. Rev. Oncol. Hematol. 36 123–39
conditions on orientation of angiogenic microvessels Murray J D 2002 Mathematical Biology 3rd edn vol I (Berlin:
Cardiovasc. Res. 78 324–32 Springer)
Kulbe H, Levinson N R, Balkwill F and Wilson J L 2004 The Murray J D 2003 Mathematical Biology 3rd edn vol II (Berlin:
chemokine network in cancer—much more than directing cell Springer)
movement Int. J. Dev. Biol. 48 489–96 Nagy J D 2005 The ecology and evolutionary biology of cancer: a
Lafleur M A, Handsley M M and Edwards D R 2003 review of mathematical models of necrosis and tumor cell
Metalloproteinases and their inhibitors in angiogenesis Expert diversity Math. Biosci. Eng. 2 381–418
Rev. Mol. Med. 5 1–39 Namy P, Ohayon J and Tracqui P 2004 Critical conditions for
Lamalice L, Le Boeuf F and Huot J 2007 Endothelial cell migration pattern formation and in vitro tubulogenesis driven by cellular
during angiogenesis Circ. Res. 100 782–94 traction fields J. Theor. Biol. 227 103–20
Landman K A and Please C P 2001 Tumour dynamics and necrosis: Nehls V and Drenckhahn D 1995 A novel, microcarrier-based in
surface tension and stability IMA J. Math. Appl. Med. Biol. vitro assay for rapid and reliable quantification of
18 131–58 three-dimensional cell migration and angiogenesis Microvasc.
Lecaudey V and Gilmour D 2006 Organizing moving groups during Res. 50 311–22
morphogenesis Curr. Opin. Cell Biol. 18 102–7 Nelson C M and Bissell M J 2006 Of extracellular matrix, scaffolds,
Lee D S, Rieger H and Bartha K 2006 Flow correlated percolation and signaling: tissue architecture regulates development,
during vascular remodeling in growing tumors Phys. Rev. Lett. homeostasis, and cancer Annu. Rev. Cell Dev. Biol.
96 058104 22 287–309
Lee S, Chen T T, Barber C L, Jordan M C, Murdock J, Desai S, Netti P A, Berk D A, Swartz M A, Grodzinsky A J and Jain R K
Ferrara N, Nagy A, Roos K P and Iruela-Arispe M L 2007 2000 Role of extracellular matrix assembly in interstitial
Autocrine VEGF signaling is required for vascular homeostasis transport in solid tumors Cancer Res. 60 2497–503
Cell 130 691–703 Nicolis G and Prigogine I 1977 Self-organization in
Levine H A, Pamuk S, Sleeman B D and Nilsen-Hamilton M 2001 Non-equilibrium Systems (New York: Wiley)
Mathematical modeling of capillary formation and Nielsen J and Oliver S 2005 The next wave in metabolome analysis
development in tumor angiogenesis: penetration into the Trends Biotechnol. 23 544–6
stroma Bull. Math. Biol. 63 801–63 Nyberg P, Xie L and Kalluri R 2005 Endogenous inhibitors of
Li S, Huang N F and Hsu S 2005 Mechanotransduction in angiogenesis Cancer Res. 65 3967–79
endothelial cell migration J. Cell Biochem. 96 1110–26 Ohayon J, Dubreuil O, Tracqui P, Le Floc’h S, Rioufol G,
Lin I E and Taber L A 1995 A model for stress-induced growth in Chalabreysse L, Thivolet F, Pettigrew R I and Finet G 2007
the developing heart J. Biomech. Eng. 117 343–9 Influence of residual stress/strain on the biomechanical
Lin R Z and Chang H Y 2008 Recent advances in three-dimensional stability of vulnerable coronary plaques: potential impact for
multicellular spheroid culture for biomedical research evaluating the risk of plaque rupture Am. J. Physiol. Heart
Biotechnol. J. 3 1172–84 Circ. Physiol. 293 H1987–96
Liotta L A and Kohn E C 2001 The microenvironment of the Oliver T, Dembo M and Jacobson K 1999 Separation of propulsive
tumour-host interface Nature 411 375–9 and adhesive traction stresses in locomoting keratocytes J. Cell
Lo C M, Wang H B, Dembo M and Wang Y L 2000 Cell Biol. 145 589–604
movement is guided by the rigidity of the substrate Biophys. J. Olson M F and Sahai E 2009 The actin cytoskeleton in cancer cell
79 144–52 motility Clin. Exp. Metastasis 26 273–87
Lubkin S R and Jackson T 2002 Multiphase mechanics of capsule Orr A W, Helmke B P, Blackman B R and Schwartz M A 2006
formation in tumors J. Biomech. Eng. 124 237–43 Mechanisms of mechanotransduction Dev. Cell 10 11–20

28
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Paszek M J et al 2005 Tensional homeostasis and the malignant Smallbone K, Gatenby R A and Maini P K 2008 Mathematical
phenotype Cancer Cell 8 241–54 modelling of tumour acidity J. Theor. Biol. 255 106–12
Peyton S R, Ghajar C M, Khatiwala C B and Putnam A J 2007 The Smallbone K, Gavaghan D J, Gatenby R A and Maini P K 2005 The
emergence of ECM mechanics and cytoskeletal tension as role of acidity in solid tumour growth and invasion J. Theor.
important regulators of cell function Cell Biochem. Biophys. Biol. 235 476–84
47 300–20 Smith A S, Sengupta K, Goennenwein S, Seifert U and Sackmann E
Potter J D 2001 Morphostats: a missing concept in cancer biology 2008 Force-induced growth of adhesion domains is controlled
Cancer Epidemiol. Biomarkers Prev. 10 161–70 by receptor mobility Proc. Natl Acad. Sci. USA 105 6906–11
Potter J D 2007 Morphogens, morphostats, microarchitecture and Sottile J 2004 Regulation of angiogenesis by extracellular matrix
malignancy Nat. Rev. Cancer 7 464–74 Biochim. Biophys. Acta 1654 13–22
Preziosi L 2003 Cancer Modelling and Simulation (London: Spence A M et al 2008 Regional hypoxia in glioblastoma
Chapman and Hall, CRC Press) multiforme quantified with [18F]fluoromisonidazole positron
Preziosi L and Tosin A 2009 Multiphase modelling of tumour emission tomography before radiotherapy: correlation
growth and extracellular matrix interaction: mathematical tools with time to progression and survival Clin. Cancer Res.
and applications J. Math. Biol. 58 625–56 14 2623–30
Quaranta V, Rejniak K A, Gerlee P and Anderson A R 2008 Spencer V A, Xu R and Bissell M J 2007 Extracellular matrix,
Invasion emerges from cancer cell adaptation to competitive nuclear and chromatin structure, and gene expression in normal
microenvironments: quantitative predictions from multiscale tissues and malignant tumors: a work in progress Adv. Cancer
mathematical models Semin. Cancer Biol. 18 338–48 Res. 97 275–94
Radisky D, Hagios C and Bissell M J 2001 Tumors are unique Stein A M, Demuth T, Mobley D, Berens M and Sander L M 2007
organs defined by abnormal signaling and context Semin. A mathematical model of glioblastoma tumor spheroid
Cancer Biol. 11 87–95 invasion in a three-dimensional in vitro experiment Biophys. J.
Retsky M W, Swartzendruber D E, Wardwell R H and Bame P D 92 356–65
1990 Is Gompertzian or exponential kinetics a valid description Stéphanou A, McDougall S R, Anderson A R A and Chaplain M A J
of individual human cancer growth? Med. Hypotheses 2005 Mathematical modelling of flow in 2D and 3D vascular
33 95–106 networks: applications to anti-angiogenic and
Riveline D, Zamir E, Balaban N Q, Schwarz U S, Ishizaki T, chemotherapeutic drug strategies Math. Comput. Modelling
Narumiya S, Kam Z, Geiger B and Bershadsky A D 2001 Focal 41 1137–56
contacts as mechanosensors: externally applied local Stephanou A, Meskaoui G, Vailhe B and Tracqui P 2007 The
mechanical force induces growth of focal contacts by an rigidity in fibrin gels as a contributing factor to the dynamics of
mDia1-dependent and ROCK-independent mechanism J. Cell in vitro vascular cord formation Microvasc. Res. 73 182–90
Biol. 153 1175–86 Sternlicht M D and Werb Z 2001 How matrix metalloproteinases
Rockne R, Alvord E C Jr, Rockhill J K and Swanson K R 2009 A regulate cell behavior Annu. Rev. Cell Dev. Biol. 17 463–516
mathematical model for brain tumor response to radiation Sugimoto H, Mundel T M, Kieran M W and Kalluri R 2006
therapy J. Math. Biol. 58 561–8 Identification of fibroblast heterogeneity in the tumor
Rodriguez E K, Hoger A and McCulloch A D 1994 Stress-dependent microenvironment Cancer Biol. Ther. 5 1640–6
finite growth in soft elastic tissues J. Biomech. 27 455–67 Suresh S 2007 Biomechanics and biophysics of cancer cells Acta
Roose T, Chapman S J and Maini P K 2007 Mathematical models of Biomater. 3 413–38
avascular tumor growth SIAM Rev. 49 179–208 Sutherland R M 1988 Cell and environment interactions in tumor
Rosello C, Ballet P, Planus E and Tracqui P 2004 Model driven microregions: the multicell spheroid model Science
quantification of individual and collective cell migration Acta 240 177–84
Biotheor. 52 343–63 Swanson K R, Alvord E C Jr and Murray J D 2000 A quantitative
Ruiter D, Bogenrieder T, Elder D and Herlyn M 2002 model for differential motility of gliomas in grey and white
Melanoma-stroma interactions: structural and functional matter Cell Prolif. 33 317–29
aspects Lancet Oncol. 3 35–43 Swanson K R, Bridge C, Murray J D and Alvord E C Jr 2003 Virtual
Saez A, Ghibaudo M, Buguin A, Silberzan P and Ladoux B 2007 and real brain tumors: using mathematical modeling to
Rigidity-driven growth and migration of epithelial cells on quantify glioma growth and invasion J. Neurol. Sci. 216 1–10
microstructured anisotropic substrates Proc. Natl Acad. Sci. Swanson K R, Rostomily R C and Alvord E C Jr 2008a A
USA 104 8281–6 mathematical modelling tool for predicting survival of
Sander L M and Deisboeck T S 2002 Growth patterns of individual patients following resection of glioblastoma: a proof
microscopic brain tumors Phys. Rev. E Stat. Nonlinear Soft of principle Br. J. Cancer 98 113–9
Matter Phys. 66 051901 Swanson K R, Harpold H L, Peacock D L, Rockne R, Pennington C,
Scheurer S B, Rybak J N, Rosli C, Neri D and Elia G 2004 Kilbride L, Grant R, Wardlaw J M and Alvord E C Jr 2008b
Modulation of gene expression by hypoxia in human umbilical Velocity of radial expansion of contrast-enhancing gliomas and
cord vein endothelial cells: a transcriptomic and proteomic the effectiveness of radiotherapy in individual patients: a proof
study Proteomics 4 1737–60 of principle Clin. Oncol. (R. Coll. Radiol.) 20 301–8
Selmeczi D, Mosler S, Hagedorn P H, Larsen N B and Flyvbjerg H Swietach P, Wigfield S, Cobden P, Supuran C T, Harris A L and
2005 Cell motility as persistent random motion: theories from Vaughan-Jones R D 2008 Tumor-associated carbonic anhydrase
experiments Biophys. J. 89 912–31 9 spatially coordinates intracellular pH in three-dimensional
Sheetz M P, Felsenfeld D P and Galbraith C G 1998 Cell migration: multicellular growths J. Biol. Chem. 283 20473–83
regulation of force on extracellular-matrix–integrin complexes Taber L A and Chabert S 2002 Theoretical and experimental study
Trends Cell Biol. 8 51–4 of growth and remodeling in the developing heart Biomech.
Sheetz M P, Felsenfeld D, Galbraith C G and Choquet D 1999 Cell Modelling Mechanobiol. 1 29–43
migration as a five-step cycle Biochem. Soc. Symp. 65 233–43 Tan J L, Tien J, Pirone D M, Gray D S, Bhadriraju K and Chen C S
Sherratt J A and Chaplain M A 2001 A new mathematical model for 2003 Cells lying on a bed of microneedles: an approach to
avascular tumour growth J. Math. Biol. 43 291–312 isolate mechanical force Proc. Natl Acad. Sci. USA 100 1484–9
Shreiber D I, Barocas V H and Tranquillo R T 2003 Temporal Tilghman R W and Parsons J T 2008 Focal adhesion kinase as a
variations in cell migration and traction during regulator of cell tension in the progression of cancer Semin.
fibroblast-mediated gel compaction Biophys. J. 84 4102–14 Cancer Biol. 18 45–52

29
Rep. Prog. Phys. 72 (2009) 056701 P Tracqui

Tlsty T D 2001 Stromal cells can contribute oncogenic signals Vernon R B, Angello J C, Iruela-Arispe M L, Lane T F and
Semin. Cancer Biol. 11 97–104 Sage E H 1992 Reorganization of basement membrane
Tlsty T D and Coussens L M 2006 Tumor stroma and regulation of matrices by cellular traction promotes the formation of cellular
cancer development Annu. Rev. Pathol. 1 119–50 networks in vitro Lab. Invest. 66 536–47
Tosin A, Ambrosi D and Preziosi L 2006 Mechanics and chemotaxis Vincent T L and Gatenby R A 2008 An evolutionary model for
in the morphogenesis of vascular networks Bull. Math. Biol. initiation, promotion, and progression in carcinogenesis Int. J.
68 1819–36 Oncol. 32 729–37
Tozern A and Skalak R 1988 Interaction of stress and growth in a Volokh K Y 2006 Stresses in growing soft tissues Acta Biomater.
fibrous tissue J. Theor. Biol. 130 337–50 2 493–504
Tracqui P 2006 Mechanical instabilities as a central issue for in Waddington C H 1968 The basic ideas of biology Towards a
silico analysis of cell dynamics Proc. IEEE 94 710–24 Theoretical Biology ed C H Waddington (Edinburgh:
Tracqui P 1995 From passive diffusion to active cellular migration Edinburgh University Press) pp 1–41
in mathematical models of tumour invasion Acta Biotheor. Wang J H, Thampatty B P, Lin J S and Im H J 2007
43 443–64 Mechanoregulation of gene expression in fibroblasts Gene
Tracqui P, Cruywagen G C, Woodward D E, Bartoo G T, Murray J D 391 1–15
and Alvord E C Jr 1995 A mathematical model of glioma Wang N, Tytell J D and Ingber D E 2009 Mechanotransduction at a
growth: the effect of chemotherapy on spatio-temporal growth. distance: mechanically coupling the extracellular matrix with
Cell Prolif. 28 17–31 the nucleus Nat. Rev. Mol. Cell Biol. 10 75–82
Tracqui P and Mendjeli M 1999 Modelling three-dimensional Weigelt B and Bissell M J 2008 Unraveling the microenvironmental
growth of brain tumours from time series of scans Math. influences on the normal mammary gland and breast cancer
Models Methods Appl. Sci. 9 581–98 Semin. Cancer Biol. 18 311–21
Turing A M 1990 The chemical basis of morphogenesis 1953 Bull. Weiner O D 2002 Regulation of cell polarity during eukaryotic
Math. Biol. 52 153–97 (discussion 19–52) chemotaxis: the chemotactic compass Curr. Opin. Cell Biol.
Tymchenko N, Wallentin J, Petronis S, Bjursten L M, Kasemo B and 14 196–202
Gold J 2007 A novel cell force sensor for quantification of Welter M, Bartha K and Rieger H 2008 Emergent vascular network
traction during cell spreading and contact guidance Biophys. J. inhomogeneities and resulting blood flow patterns in a growing
93 335–45 tumor J. Theor. Biol. 250 257–80
Tzvetkova-Chevolleau T, Stephanou A, Fuard D, Ohayon J, Wolf K, Wu Y I, Liu Y, Geiger J, Tam E, Overall C, Stack M S and
Schiavone P and Tracqui P 2008 The motility of normal and Friedl P 2007 Multi-step pericellular proteolysis controls the
cancer cells in response to the combined influence of the transition from individual to collective cancer cell invasion Nat.
substrate rigidity and anisotropic microstructure Biomaterials Cell. Biol. 9 893–904
29 1541–51 Woodward D E, Cook J, Tracqui P, Cruywagen G C, Murray J D and
Vailhe B, Vittet D and Feige J J 2001 In vitro models of Alvord E C Jr 1996 A mathematical model of glioma growth:
vasculogenesis and angiogenesis Lab. Invest. 81 439–52 the effect of extent of surgical resection Cell Prolif. 29 269–88
Vailhe B, Ronot X, Tracqui P, Usson Y and Tranqui L 1997 In vitro Zaman M H, Trapani L M, Sieminski A L, Mackellar D, Gong H,
angiogenesis is modulated by the mechanical properties of Kamm R D, Wells A, Lauffenburger D A and Matsudaira P
fibrin gels and is related to alpha(v)beta3 integrin localization 2006 Migration of tumor cells in 3D matrices is governed by
In Vitro Cell Dev. Biol. Anim. 33 763–73 matrix stiffness along with cell–matrix adhesion and
Venkatesh S K, Yin M, Glockner J F, Takahashi N, Araoz P A, proteolysis Proc. Natl Acad. Sci. USA 103 10889–94
Talwalkar J A and Ehman R L 2008 MR elastography of liver Zeisberg E M, Potenta S, Xie L, Zeisberg M and Kalluri R 2007
tumors: preliminary results AJR Am. J. Roentgenol. Discovery of endothelial to mesenchymal transition as a source
190 1534–40 for carcinoma-associated fibroblasts Cancer Res. 67 10123–8

30

Potrebbero piacerti anche