Conference PaperPDF Available

A complete mathematical study of a 3D model of heterogeneous and anisotropic glioma evolution

Authors:

Abstract and Figures

Glioma is the most aggressive type of brain cancer. Several mathematical models have been developed towards identifying the mechanism of tumor growth. The most successful models have used variations of the diffusion-reaction equation, with the recent ones taking into account brain tissue heterogeneity and anisotropy. However, to the best of our knowledge, there hasn't been any work studying in detail the mathematical solution and implementation of the 3D diffusion model, addressing related heterogeneity and anisotropy issues. To this end, this paper introduces a complete mathematical framework on how to derive the solution of the equation using different numerical approximation of finite differences. It indicates how different proliferation rate schemes can be incorporated in this solution and presents a comparative study of different numerical approaches.
Content may be subject to copyright.
Abstract Glioma is the most aggressive type of brain
cancer. Several mathematical models have been developed
towards identifying the mechanism of tumor growth. The
most successful models have used variations of the
diffusion-reaction equation, with the recent ones taking into
account brain tissue heterogeneity and anisotropy.
However, to the best of our knowledge, there hasn’t been
any work studying in detail the mathematical solution and
implementation of the 3D diffusion model, addressing
related heterogeneity and anisotropy issues. To this end,
this paper introduces a complete mathematical framework
on how to derive the solution of the equation using different
numerical approximation of finite differences. It indicates
how different proliferation rate schemes can be
incorporated in this solution and presents a comparative
study of different numerical approaches.
I. INTRODUCTION
LIOMAS are the most malignant form of brain
tumor, which differ from other tumors, because of
their highly aggressive and diffusive behavior. Since
90s, many important diffusive models have been
introduced [1-3], with the most recent ones taking brain
tissue heterogeneity and anisotropic cell migration into
account. According to Jbadbi [3], the spatiotemporal
diffusion equation that describes glioma growth is
(1)
where c(x,t) is the tumor concentration in position x at
time t, is the diffusion tensor, i.e. a 3x3 symmetric
matrix that expresses anisotropy of cell migration, and
div are the gradient and divergence operators
respectively and is the net cell proliferation rate.
Variant formalisms of have been proposed [4], with
the main ones following either the exponential law
, (2)
or the Verhulst law
(3)
or Gompertz law
(4)
Manuscript received April 7, 2009. This work was supported in part by
the EC ICT project ContraCancrum, Contract No: 223979.
A. Roniotis, V. Sakkalis, and K. Marias are with the Institute of
Computer Science, Foundation for Research and Technology
(FORTH), Heraklion 71110, Greece (e-mail: {roniotis; sakkalis;
marias}@ics.forth.gr).
G. D. Tsibidis is with the Institute of Electronic Structure and Laser,
Foundation for Research and Technology (FORTH), Heraklion 71110,
Greece (e-mail: tsibidis@iesl.forth.gr).
A. Roniotis and M. Zervakis are with the Dept. of Electronic &
Computer Engineering, Technical University of Crete, 73100, Chania,
Greece (email: roniotis@ics.forth.gr, michalis@display.tuc.gr).
where denotes the proliferation rate constant and is
the maximum value that can reach.
Due to the spatial dependence of D, an analytical
solution of (1) cannot be acquired; therefore, it has to be
numerically approximated. Finite differences (FDs) are
commonly used in diffusive models. By using FDs, a big
system of equations arises, the iterative solution of
which yields the approximated tumor cell concentration
at a desired time point. Different numerical schemes for
approximating partial derivatives are able to differentiate
the emerging system, thus the way it is solved.
Up to now, the various implementations lack a firm
mathematical background on the derivation of the
system, with concrete assumptions on the approximation
scheme. The main objective of this paper is to provide
the direct formalism of the derived linear system, for the
widely used FD schemes, namely forward Euler (FE),
backward Euler (BE), Crank Nikolson (CN) and θ-
methods. These formalisms are designed for 3D,
heterogeneous and anisotropic brain tissue and entail the
general form of , so that one could use any net
proliferation rate. We also present how equations (2),(3)
and (4) are tailored to the model. Finally, we make a
comparative study of the different FD numerical
schemes by analyzing experimental results.
II. METHODS
Equation (1) is a partial 2nd order differential equation.
Before hunting up a direct expression of the linear
system that iteratively solves it, (1) is expanded in three
spatial dimensions. Assume that the diffusion tensor
D(x) at a point x=(x1, x2, x3) is:
(5)
Then, by using definitions of and , (1) becomes:
(6)
The next step is to use FDs so as to approximate the
solution of (6). For approximating the partial derivatives,
i) FE, ii) BE, iii) θ- and iv) CN schemes are used.
A. Forward Euler Method (FE)
At first, assuming that initial and boundary conditions
have been defined, we study how FE approximates the
solution of (6) for a grid. If
c(nΔT,iΔX,jΔY,kΔZ), Dpq(iΔX,jΔY,kΔZ),
i , j , k and
, then the partial derivatives of (6) in point
A complete mathematical study of a 3D model of
heterogeneous and anisotropic glioma evolution
Alexandros Roniotis, Kostas Marias, Vangelis Sakkalis, George D. Tsibidis, and Michalis Zervakis
G
(iΔX,jΔY,kΔZ) can be approximated as
(7)
and similarly for , , , and .
If any of the neighboring is a boundary point, then
its value will be as initially defined. Continuing, by
substituting the approximations (7) to (6), we derive:
+ +
+ + +
+ + +
+ + +
+ + +
+ + +
+ + (8)
or equivalently:
(9)
where
(10)
If the vectorized version of at time m is taken, as
(11)
then, the overall solution of the equation at time ,
for given , can be found by solving iteratively
where A is a
atrix with its elements defined as:
where:
(12)
and F a vectorization operator that vectorizes f( ) as
(13)
A is a symmetric, sparse, 19-
diagonal matrix, with its form being visualized in Fig.1,
where all light gray areas have zero values.
After having acquired A, a direct solution can be found
by iteratively calculating
(14)
where I is the identity matrix. This is
the solution of the FE. This is called forward, because
the next-time approximation of concentration can be
directly estimated as a linear combination of the
previous-time approximation and is easy to implement,
but numerical stability has to be ensured. As proven in
[5], this method is stable giving reliable results when
(15)
B. Backward Euler (BE)
Similarly, the density of glioma cells at time t, using BE,
can be derived by iteratively solving the system:
- (16)
till time is reached, with A and F being the same with
(12) and (13) respectively. In each iteration the system
can be expressed with a big, sparse, symmetric and
positive definite matrix; an iterative method for solving
Fig. 1. Sparse Matrix A: White areas have zero values, thick lines
are 3 diagonals in a row and thin lines are single diagonals.
linear systems, e.g. the conjugate gradient method can be
used. Unconditional stability and accuracy are
advantages of this method, whereas computational and
storage load that has to be considered [6].
C. θ-methods/Crank Nikolson (CN)
The θ-methods use a balancing parameter θ
so as to combine forward and backward numerical
schemes concurrently. By using θ-methods we get
- (17)
where . As in BE, a method for solving linear
systems is required. The θ-methods are flexible due to
choice of parameter θ but higher computational and
storage load are required [6]. CN is a θ-method for θ.
In Table I, the formulation of the iterative linear
systems that produce the approximated solution of (6) is
presented according to various selected methods. Based
on this, we have implemented models for glioma growth,
directly in 3D using C++ (the authors can be contacted
for more details).
D. Vectorization Operator
It is interesting to study how operator F is chosen for
the mainly used proliferation rates of (2),(3) and (4). If
the net proliferation rate is given by (2) holds, then
according to (13):
(18)
or equivalently:
(19)
Next, if the net proliferation rate is given by (3), then:
(20)
or equivalently:
. (21)
The definitions of and are given in Table 2.
Lastly, if the net proliferation rate is given by (4) holds,
then according to (13):
(22)
or equivalently:
(23)
In Table II, a summary of vectorization operator for the
commonly used proliferation rates is presented.
III. RESULTS AND DISCUSSION
In order to study the performance of the different
numerical schemes presented, a simplified test case of
the pure diffusion equation is used, for which there is a
known analytical continuous expression of the solution.
Hence, the magnitude of each numerical scheme
deviation from the real solution can be studied, which
serves here as ‘ground truth’ for validating our numerical
approximations.
A. Spherical homogeneous test tumor
To validate our methodology, it is assumed that tumor
growth in 3D, unbounded, isotropic (i.e. D is constant)
and homogeneous region exhibits a pure diffusion
behavior (i.e. ). The tumor has initially a
concentration and it is constrained in a sphere of
radius (Fig. 2). Due to symmetry, the concentration of
glioma depends only on the distance from the center of
the sphere and it is given by the expression [7]:
(24)
B. Testing and Simulation Description
In our tests, the model is adapted to approximate (24),
by using the following parameters:
, , ,
, points and
. Five different simulations were
run for 200 days, using FE, three θ-methods for θ=0.25,
θ=0.5 (CN) , θ=0.75, and BE performed on a Pentium 4
at 3.8 GHz.
In Fig.3, the evolution of cell concentration in time is
presented, with respect to , with lines corresponding to
results from analytical expression (24), while dots
representing what simulation with CN yields. Fig. 3
shows a very good agreement between these results;
however a more rigorous investigation would require an
error estimation analysis.
Fig. 2. Left: The initial spherical tumor of radius . Right: The
initial tumor concentration according to at time t=0
TABLE I
DIRECT EXPRESSIONS OF THE ITERATIVE LINEAR SYSTEMS
Iterative linear system solving (6)
TABLE II
VECTORIZATION OPERATOR FOR DIFFERENT PROLIFERATION RATES
Proliferation
Rate
Constan
t Rate
f(c)
Vectorization Operator
Exponential
Verhulst
(logistic)
Gompertz
C. Error Estimation
In order to estimate the error that each scheme yields
the normalized mean absolute error is introduced. Due
to symmetry, at time is computed as
100% (25)
In Table III, the estimated for each scheme on day 10,
100 and 200 of simulation is reported. Moreover, the
overall simulation times are also reported. The highest
for all five schemes is on 10th day, while it decreases to
values on the 200th day. FE error is the lowest
on 10th day, i.e. 6.36%, but overcomes all other schemes
on 200th day, reaching 0.57% at the 200th day. CN is the
scheme that reaches the lowest error value at 200th day at
0.23%. In Fig.4, a logarithmic graph of in time is
presented, for each scheme. It is noticeable that all
schemes initially produce higher errors. A possible
explanation is that the initial concentration has
an abrupt step descent at r=10mm, as seen in Fig. 2
(right). This makes the approximation of the partial
derivatives at the edge of this step erroneous for all
schemes. However, as simulation continues, all schemes
tend to significantly decrease . Continuing, one can
observe that decreases to values <1% on day 40, 75,
94, 143 and 156 for FE, 0.25-method, CN, 0.75-method
and BE respectively. FE error has a remarkable descent
till day 42, but later decreases smoothly and finally
overcomes all other schemes. BE produces the highest
error, till day 186, when FE overcomes it. As expected,
approximation with θ-methods is more accurate than BE
throughout all days and more accurate than FE in most
time. θ-method for θ=0.25 initially tends to have a steep
descent to error 0.3%, but later reaches a balancing value
higher than CN. θ-method for θ=0.75 has a smoother
descent, being always higher than CN. Lastly, CN
(θ=0.5) seems to have the best general performance,
reaching the lowest error 0.23% on day 200.
Thus, CN seems to yield more accurate approximations
of . However, if there is room for sacrificing
accuracy for faster model simulations, then FE should be
used, since from Table III it’s derived that FE is 4.63
times faster than CN. Even if one chooses FE, that is the
worst case scenario, error doesn’t overcome 0.6%
IV. CONCLUSION
This paper presents a complete, FDs’ based,
framework for the mathematical solving of the
anisotropic, heterogeneous and 3D diffusion-reaction
equation simulating either the growth of glioma or other
phenomena where diffusive models are applicable.
Moreover, the introduction of the vectorization operator
gives the flexibility to include any proliferation rate into
the model. Lastly, a performance study of different
numerical schemes is presented based on an analytical
expression that can be derived for the pure diffusion
equation.
REFERENCES
[1] P. Tracqui, “From passive diffusion to active cellular migration in
mathematical models of tumor invasion,” Acta Bibliotheoretica,
vol. 43, pp 443-464, 1995.
[2] K. R. Swanson, E. C. Alvord, and J. D. Murray, A Quantitative
Model for Differential Motility of Gliomas in Grey and White
Matter, Cell Proliferation, vol. 33, no. 5, pp. 317-330, 2000.
[3] S. Jbabdi, E. Mandonnet, and H. Duffau, “Simulation of
anisotropic growth of low-grade gliomas using diffusion tensor
imaging,” Magn Reson Med, vol. 54, pp. 616-24, 2005.
[4] M. Marusic, Z. Bajzer, J. P. Freyer, and S. Vuk-Palovic,
“Analysis of growth of multicellular tumour spheroids by
mathematical models,” Cell Prolif, vol. 27, pp. 73-94, 1994.
[5] S. Puwal, and B. J. Roth, Forward Euler Stability of the
Bidomain Model of Cardiac Tissue, IEEE Transactions on
Biom. Eng., vol. 54, no. 5, pp. 951-953, 2005.
[6] S. Teukolsky, W. Vetterling, B. Flannery, Numerical Recipes:
The Art of Scientific Computing (Third Edition), Cambridge
University Press, NY, 2007.
[7] J. Crank, The Mathematics of Diffusion (Second Edition), Oxford
University Press, NY, 1975.
TABLE III
ERROR AFTER 10, 100 AND 200 DAYS
Scheme
10 days
100 days
200 days
Simulation Time
FE
6.36%
0.89%
0.57%
3’53”
θ-method
(θ=0.25)
9.73%
0.43%
0.33%
18’13”
CN
9.83%
0.91%
0.23%
19’01”
θ-method
(θ=0.75)
10.17%
1.43%
0.35%
20’16”
BE
11.33%
1.95%
0.50%
22’31”
Fig. 3. The evolution of after 10, 100 and 200 days
70 80 90 100 110
101
102
103
104
r(mm)
C(r)
cells/mm 3
Analytical Solution
Numerical Solution
Analytical Solution
Numerical Solution
Analytical Solution
Numerical Solution
Day 200
Day 100
Day 10
Fig. 4. Logarithmic visualization of error in time
... Over the last decades, a plethora of mathematical models have been developed, addressing different aspects of tumor complexity ranging from tumor growth to heterogeneity in tumor microenvironment [1][2][3][4][5][6][7][8]. Furthermore, the massive amount of human biological data being produced by multiple and diverse sources (imaging, expression, mutation, clinical, etc.) necessitates the development of integrative approaches able to summarize all this information into answers to specific clinical questions related to treatment responses and prognoses. ...
Article
Full-text available
The massive amount of human biological, imaging, and clinical data produced by multiple and diverse sources necessitates integrative modeling approaches able to summarize all this information into answers to specific clinical questions. In this paper, we present a hypermodeling scheme able to combine models of diverse cancer aspects regardless of their underlying method or scale. Describing tissue-scale cancer cell proliferation, biomechanical tumor growth, nutrient transport, genomic-scale aberrant cancer cell metabolism, and cell-signaling pathways that regulate the cellular response to therapy, the hypermodel integrates mutation, miRNA expression, imaging, and clinical data. The constituting hypomodels, as well as their orchestration and links, are described. Two specific cancer types, Wilms tumor (nephroblastoma) and non-small cell lung cancer, are addressed as proof-of-concept study cases. Personalized simulations of the actual anatomy of a patient have been conducted. The hypermodel has also been applied to predict tumor control after radiotherapy and the relationship between tumor proliferative activity and response to neoadjuvant chemotherapy. Our innovative hypermodel holds promise as a digital twin-based clinical decision support system and as the core of future in silico trial platforms, although additional retrospective adaptation and validation are necessary.
... Several research studies have employed mathematical models to investigate the consequences of different scenarios for decision-making in various medical domains, including both non-communicable diseases like cancer [17,18] and communicable ones. Specifically, for communicable diseases, these models can be broadly classified into three types: statistical, empirical, and state-space models. ...
Article
Full-text available
The regulation policies implemented, the characteristics of vaccines, and the evolution of the virus continue to play a significant role in the progression of the SARS-CoV-2 pandemic. Numerous research articles have proposed using mathematical models to predict the outcomes of different scenarios, with the aim of improving awareness and informing policy-making. In this work, we propose an expansion to the classical SEIR epidemiological model that is designed to fit the complex epidemiological data of COVID-19. The model includes compartments for vaccinated, asymptomatic, hospitalized, and deceased individuals, splitting the population into two branches based on the severity of progression. In order to investigate the impact of the vaccination program on the spread of COVID-19 in Greece, this study takes into account the realistic vaccination program implemented in Greece, which includes various vaccination rates, different dosages, and the administration of booster shots. It also examines for the first time policy scenarios at crucial time-intervention points for Greece. In particular, we explore how alterations in the vaccination rate, immunity loss, and relaxation of measures regarding the vaccinated individuals affect the dynamics of COVID-19 spread. The modeling parameters revealed an alarming increase in the death rate during the dominance of the delta variant and before the initiation of the booster shot program in Greece. The existing probability of vaccinated people becoming infected and transmitting the virus sets them as catalytic players in COVID-19 progression. Overall, the modeling observations showcase how the criticism of different intervention measures, the vaccination program, and the virus evolution has been present throughout the various stages of the pandemic. As long as immunity declines, new variants emerge, and vaccine protection in reducing transmission remains incompetent; monitoring the complex vaccine and virus evolution is critical to respond proactively in the future.
... where div n i,j,k is the divergence of the tumour cell flux at location (i, j, k) and time step n constituting the first term of Equation (6.6). To ensure numerical stability of the explicit Euler method, the time step ∆t must be chosen such that [215]: ...
Thesis
Full-text available
Gliomas are the most common primary brain tumours and are associated with poor prognosis. Among them, diffuse gliomas – which include their most aggressive form glioblastoma (GBM) – are known to be highly infiltrative. The diagnosis and follow-up of gliomas rely on positron emission tomography (PET) and magnetic resonance imaging (MRI). However, these imaging techniques do not currently allow to assess the whole extent of such infiltrative tumours nor to anticipate their preferred invasion patterns, leading to sub-optimal treatment planning. Mathematical tumour growth modelling has been proposed to address this problem. Reaction-diffusion tumour growth models, which are probably the most commonly used for diffuse gliomas growth modelling, propose to capture the proliferation and migration of glioma cells by means of a partial differential equation. Although the potential of such models has been shown in many works for patient follow-up and therapy planning, only few limited clinical applications have seemed to emerge from these works. This thesis aims at revisiting reaction-diffusion tumour growth models using state-of-the-art medical imaging and data processing technologies, with the objective of integrating multi-parametric PET/MRI data to further personalise the model. Brain tissue segmentation on MR images is first addressed with the aim of defining a patient-specific domain to solve the model. A previously proposed method to derive a tumour cell diffusion tensor from the water diffusion tensor assessed by diffusion-tensor imaging (DTI) is then implemented to guide the anisotropic migration of tumour cells along white matter tracts. The use of dynamic [S-methyl-11C]methionine ([11C]MET) PET is also investigated to derive patient-specific proliferation potential maps for the model. These investigations lead to the development of a microscopic compartmental model for amino acid PET tracer transport in gliomas. Based on the compartmental model results, a novel methodology is proposed to extract parametric maps from dynamic [11C]MET PET data using principal component analysis (PCA). The problem of estimating the initial conditions of the model from MR images is then addressed by means of a translational MRI/histology study in a case of non-operated GBM. Numerical solving strategies based on the widely used finite difference and finite element methods are finally implemented and compared. All these developments are embedded within a common framework allowing to study glioma growth in silico and providing a solid basis for further research in this field. However, commonly accepted hypothesis relating the outlines of abnormalities visible on MRI to tumour cell density iso-contours have been invalidated by the translational study carried out, leaving opened the questions of the initialisation and the validation of the model. Furthermore, the analysis of the temporal evolution of real multi-treated glioma patients demonstrates the limitations of the formulated model. These latter statements highlight current obstacles to the clinical application of reaction-diffusion tumour growth models and pave the way to further improvements.
... Mathematical mechanistic models are commonly used to allow deeper understanding and quantification of the underlying biological mechanisms involved in tumor progression [20] and even proposals for clinical translation [21]. A variety of mathematical models have been proposed to describe tumor spheroids and explain their growth dynamics [22][23][24][25]. ...
Article
Full-text available
Glioblastoma is the most malignant brain tumor among adults. Despite multimodality treatment, it remains incurable, mainly because of its extensive heterogeneity and infiltration in the brain parenchyma. Recent evidence indicates dysregulation of the expression of the Promyelocytic Leukemia Protein (PML) in primary Glioblastoma samples. PML is implicated in various ways in cancer biology. In the brain, PML participates in the physiological migration of the neural progenitor cells, which have been hypothesized to serve as the cell of origin of Glioblastoma. The role of PML in Glioblastoma progression has recently gained attention due to its controversial effects in overall Glioblastoma evolution. In this work, we studied the role of PML in Glioblastoma pathophysiology using the U87MG cell line. We genetically modified the cells to conditionally overexpress the PML isoform IV and we focused on its dual role in tumor growth and invasive capacity. Furthermore, we targeted a PML action mediator, the Enhancer of Zeste Homolog 2 (EZH2), via the inhibitory drug DZNeP. We present a combined in vitro–in silico approach, that utilizes both 2D and 3D cultures and cancer-predictive computational algorithms, in order to differentiate and interpret the observed biological results. Our overall findings indicate that PML regulates growth and invasion through distinct cellular mechanisms. In particular, PML overexpression suppresses cell proliferation, while it maintains the invasive capacity of the U87MG Glioblastoma cells and, upon inhibition of the PML-EZH2 pathway, the invasion is drastically eliminated. Our in silico simulations suggest that the underlying mechanism of PML-driven Glioblastoma physiology regulates invasion by differential modulation of the cell-to-cell adhesive and diffusive capacity of the cells. Elucidating further the role of PML in Glioblastoma biology could set PML as a potential molecular biomarker of the tumor progression and its mediated pathway as a therapeutic target, aiming at inhibiting cell growth and potentially clonal evolution regarding their proliferative and/or invasive phenotype within the heterogeneous tumor mass.
... The work ignores the pharmacokinetics, the spatial distribution of nutrients and drugs that is developed in dense configurations, the phenotypic drift and clonal evolutionary dynamics that may alter the proliferative capacity of the daughter cells relative to their parents, the microenvironmental heterogeneity, as well as the variety of microenvironmental dependences and adaptation that may alter the phenotypes. Furthermore, the work also focuses on tumor growth driven solely by cellular proliferation, whereas migration and invasion mechanisms could be added in the future, as well as additional resistant mechanisms and trade-offs (11), more complex evolutionary mechanisms (21), a realistic 3D setting (26), and the exploration of more sophisticated therapeutic schemes, like adaptive therapy (11). We have intentionally chosen a simple starting point in order to better demonstrate the impact of heterogeneity with respect to cell cycle duration before, during, and after treatment. ...
Article
Full-text available
Tumors are complex, dynamic, and adaptive biological systems characterized by high heterogeneity at genetic, epigenetic, phenotypic, as well as tissue microenvironmental level. In this work, utilizing cellular automata methods, we focus on intrinsic heterogeneity with respect to cell cycle duration and explore whether and to what extent this heterogeneity affects cancer cell growth dynamics when cytotoxic treatment is applied. We assume that treatment acts on cancer cells specifically during mitosis and compare it with a (cell cycle-non-specific) cytotoxic treatment that acts randomly regardless of the cell cycle phase. We simulate the spatiotemporal evolution of tumor cells with different initial spatial configurations and different cell length probability distributions. We observed that in heterogeneous populations, strong selection forces act on cancer cells favoring the faster cells, when the death rates are lower than the proliferation rates. However, at higher mitotic death rates, selection of the slower proliferative cells is favored, leading to slower post-treatment regrowth rates, as compared to untreated growth. Of note, random cell death progressively eliminates the slower proliferative cells, consistently, favoring highly proliferative phenotypes. Interestingly, compared to the monoclonal populations that exhibit complete response at high random death rates, emergent resistance arises naturally in heterogeneous populations during treatment. As divergent selection forces may act on a heterogeneous cancer cell population, we argue that treatment plan selection can considerably alter the post-treatment tumor dynamics, cell survival, and emergence of resistance, proving its significant biological and therapeutic impact.
... By predicting the behavior of the system, new targeted experiments can be designed. In that way, the process of mathematical modeling validation is an iterative refinement procedure [25], which terminates when a valid and biologically plausible and concrete description of the system that reproduces the observed cellular behaviors and growth patterns is found. Several mathematical approaches have been proposed to describe the complex, multiscale spatiotemporal tumor evolution. ...
Article
Full-text available
The application of accurate cancer predictive algorithms validated with experimental data is a field concerning both basic researchers and clinicians, especially regarding a highly aggressive form of cancer, such as Glioblastoma. In an aim to enhance prediction accuracy in realistic patient-specific environments, accounting for both inter- and intratumoral heterogeneity, we use patient-derived Glioblastoma cells from different patients. We focus on cell proliferation using in vitro experiments to estimate cell doubling times and sizes for established primary Glioblastoma cell lines. A preclinically driven mathematical model parametrization is accomplished by taking into account the experimental measurements. As a control cell line we use the well-studied U87MG cells. Both in vitro and in silico results presented support that the variance between tumor staging can be attributed to the differential proliferative capacity of the different Glioblastoma cells. More specifically, the intratumoral heterogeneity together with the overall proliferation reflected in both the proliferation rate and the mechanical cell contact inhibition can predict the in vitro evolution of different Glioblastoma cell lines growing under the same conditions. Undoubtedly, additional imaging techniques capable of providing spatial information of tumor cell physiology and microenvironment will enhance our understanding regarding Glioblastoma nature and verify and further improve our predictability.
... Roniotis et al. [113,114] presented mathematical solution for the RD model in 3D with different FDM schemes and FEM. Namely, forward Euler, the backward Euler, the Crank Nikolson, and the θ-method were discussed in terms of accuracy, simulation time, and storage. ...
Article
Full-text available
Mathematical models have been ubiquitously employed in various applications. One of these applications that arose in the past few decades is cerebral tumors growth modelling. Simultaneously, medical imaging techniques such as magnetic resonance imaging, computed tomography, and positron emission tomography have witnessed great developments and become the primary clinical procedure in tumors diagnosis and detection. Studying tumor growth via mathematical models from multimodal images is an important application that is believed to play significant role in cancer treatment by predicting tumor evolution, quantifying the response to therapy, and effective treatment planning of chemotherapy and/or radiotherapy. In this review, we focus on the macroscopic growth modeling of brain tumors, mainly glioma, and highlight the current achievements in the-state-of-the-art methods. In addition, we discuss some challenges and perspectives on this research that can further promote the research of this field.
... There have also been some recent studies accounting for heterogeneity and anisotropy in tumor growth. A finite differences based framework for the anisotropic, heterogeneous and 3D diffusive tumor growth model has been developed [110]. A recently developed heterogeneous 3D avascular tumor model have used lattice gas cellular automata employing a particle lattice structure where the movement and transition of cells are determined by a set of local rules [120]. ...
Article
Full-text available
New directions in medical and biomedical sciences have gradually emerged over recent years that will change the way diseases are diagnosed and treated and are leading to the redirection of medicine toward patient-specific treatments. We refer to these new approaches for studying biomedical systems as predictive medicine, a new version of medical science that involves the use of advanced computer models of biomedical phenomena, high-performance computing, new experimental methods for model data calibration, modern imaging technologies, cutting-edge numerical algorithms for treating large stochastic systems, modern methods for model selection, calibration, validation, verification, and uncertainty quantification, and new approaches for drug design and delivery, all based on predictive models. The methodologies are designed to study events at multiple scales, from genetic data, to sub-cellular signaling mechanisms, to cell interactions, to tissue physics and chemistry, to organs in living human subjects. The present document surveys work on the development and implementation of predictive models of vascular tumor growth, covering aspects of what might be called modeling-and-experimentally based computational oncology. The work described is that of a multi-institutional team, centered at ICES with strong participation by members at M. D. Anderson Cancer Center and University of Texas at San Antonio. This exposition covers topics on signaling models, cell and cell-interaction models, tissue models based on multi-species mixture theories, models of angiogenesis, and beginning work of drug effects. A number of new parallel computer codes for implementing finite-element methods, multi-level Markov Chain Monte Carlo sampling methods, data classification methods, stochastic PDE solvers, statistical inverse algorithms for model calibration and validation, models of events at different spatial and temporal scales is presented. Importantly, new methods for model selection in the presence of uncertainties fundamental to predictive medical science, are described which are based on the notion of Bayesian model plausibilities. Also, as part of this general approach, new codes for determining the sensitivity of model outputs to variations in model parameters are described that provide a basis for assessing the importance of model parameters and controlling and reducing the number of relevant model parameters. Model specific data is to be accessible through careful and model-specific platforms in the Tumor Engineering Laboratory. We describe parallel computer platforms on which large-scale calculations are run as well as specific time-marching algorithms needed to treat stiff systems encountered in some phase-field mixture models. We also cover new non-invasive imaging and data classification methods that provide in vivo data for model validation. The study concludes with a brief discussion of future work and open challenges.
Conference Paper
Full-text available
In this article we review some of the recent developments in mathematical modeling of tumor. Despite internal complexity, tumor growth kinetics follow relatively simple laws that can be expressed as mathematical models. Parabolic partial differential equations with nonlocal boundary conditions arise in modeling of tumor invasion. The 2D diffusion equation allows us to talk about the statistical movements of randomly moving particles in two dimensions. The movement of each individual particle moving in a Brownian (diffuse) way does notfollow the diffusion equation. However, many identical particles each obeying the same boundary and initial conditions share some statistical properties dealing with their spatial and temporal evolution. In this paper, we present the implementation of positivity preserving Padé numerical schemes to the two-dimensional diffusion equation with nonlocal time dependent boundary condition. The goals were threefold: 1) to conclude a mathematical model for description of the measurement error, 2) to establish the descriptive power, using several goodness-of-fit, and 3) to measure the models' ability to estimate future tumor growth. We successfully implemented these numerical schemes and the numerical results show that these Padé approximation based numerical schemes are quite accurate and easily implemented.
Chapter
The proposed method is to analyze brain tumor to identify the grade of glioma from magnetic resonant image and histopathological images. The proposed work includes three phases. The first phase preprocesses the pathological images. This involves enhancement and contrast improvement of the images. Secondly, the processed histopathology image is subjected to feature extraction. Gaussian filters techniques and statistical feature extraction techniques are utilized for feature extraction. In the last phase, classifiers are developed to classify the low-grade and high-grade images based on extracted features. K-mean clustering network and SVD classifier are used for classification of low-grade/high-grade gliomas. MATLAB, a familiar tool, efficiently uses algorithms and techniques for identification of low-grade and high-grade glioma tumors.
Article
Full-text available
The bidomain model describes the anisotropic electrical properties of cardiac tissue. One common numerical technique for solving the bidomain equations is the explicit forward Euler method. In this communication we derive a relationship between the time and space steps that ensures the stability of this numerical method.
Article
Mathematical models of tumour invasion appear as interesting tools for connecting the information extracted from medical imaging techniques and the large amount of data collected at the cellular and molecular levels. Most of the recent studies have used stochastic models of cell translocation for the comparison of computer simulations with histological solid tumour sections in order to discriminate and characterise expansive growth and active cell movements during host tissue invasion. This paper describes how a deterministic approach based on reaction-diffusion models and their generalisation in the mechano-chemical framework developed in the study of biological morphogenesis can be an alternative for analysing tumour morphological patterns. We support these considerations by reviewing two studies. In the first example, successful comparison of simulated brain tumour growth with a time sequence of computerised tomography (CT) scans leads to a quantification of the clinical parameters describing the invasion process and the therapy. The second example considers minimal hypotheses relating cell motility and cell traction forces. Using this model, we can simulate the bifurcation from an homogeneous distribution of cells at the tumour surface toward a nonhomogeneous density pattern which could characterise a pre-invasive stage at the tumour-host tissue interface.
Article
We wished to determine the applicability of previously proposed deterministic mathematical models to description of growth of multicellular tumour spheroids. The models were placed into three general classes: empirical, functional and structural. From these classes, 17 models were applied systematically to growth curves of multicellular tumour spheroids used as paradigms of prevascular and microregional tumour growth. The spheroid growth curves were determined with uniquely high density of measurements and high precision. The theoretical growth curves obtained from the models were fitted by the weighted least-squares method to the 15 measured growth curves, each corresponding to a different cell line. The classical growth models such as von Bertalanffy, logistic and Gompertz were considered as nested within more general models. Our results demonstrate that most models fitted the data fairly well and that criteria other than statistical had to be used for final selection. The Gompertz, the autostimulation and the simple spheroid models were the most appropriate for spheroid growth in the empirical, functional and structural classes of models, respectively. We also showed that some models (e.g. logistic, von Bertalanffy) were clearly inadequate. Thus, contrary to the widely held belief, the sigmoid character of a three or more parameter growth function is not sufficient for adequate fits.
Article
We have extended a mathematical model of gliomas based on proliferation and diffusion rates to incorporate the effects of augmented cell motility in white matter as compared to grey matter. Using a detailed mapping of the white and grey matter in the brain developed for a MRI simulator, we have been able to simulate model tumours on an anatomically accurate brain domain. Our simulations show good agreement with clinically observed tumour geometries and suggest paths of submicroscopic tumour invasion not detectable on CT or MRI images. We expect this model to give insight into microscopic and submicroscopic invasion of the human brain by glioma cells. This method gives insight in microscopic and submicroscopic invasion of the human brain by glioma cells. Additionally, the model can be useful in defining expected pathways of invasion by glioma cells and thereby identify regions of the brain on which to focus treatments.
Article
A recent computational model of brain tumor growth, developed to better describe how gliomas invade through the adjacent brain parenchyma, is based on two major elements: cell proliferation and isotropic cell diffusion. On the basis of this model, glioma growth has been simulated in a virtual brain, provided by a 3D segmented MRI atlas. However, it is commonly accepted that glial cells preferentially migrate along the direction of fiber tracts. Therefore, in this paper, the model has been improved by including anisotropic extension of gliomas. The method is based on a cell diffusion tensor derived from water diffusion tensor (as given by MRI diffusion tensor imaging). Results of simulations have been compared with two clinical examples demonstrating typical growth patterns of low-grade gliomas centered around the insula. The shape and the kinetic evolution are better simulated with anisotropic rather than isotropic diffusion. The best fit is obtained when the anisotropy of the cell diffusion tensor is increased to greater anisotropy than the observed water diffusion tensor. The shape of the tumor is also influenced by the initial location of the tumor. Anisotropic brain tumor growth simulations provide a means to determine the initial location of a low-grade glioma as well as its cell diffusion tensor, both of which might reflect the biological characteristics of invasion.