Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902.

The copyright holder for this preprint (which was not


peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

Addressing current challenges in cancer immunotherapy with


mathematical and computational modeling
Anna Konstorum1 , Anthony T. Vella2 , Adam J. Adler2 and Reinhard Laubenbacher∗3
1
Center for Quantitative Medicine, UConn Health, Farmington, CT
2
Department of Immunology, UConn Health, Farmington, CT
1,3
Jackson Laboratory for Genomic Medicine, Farmington, CT

Abstract
The goal of cancer immunotherapy is to boost a patient’s immune response to a tumor. Yet, the design of
an effective immunotherapy is complicated by various factors, including a potentially immunosuppressive
tumor microenvironment, immune-modulating effects of conventional treatments, and therapy-related
toxicities. These complexities can be incorporated into mathematical and computational models of can-
cer immunotherapy that can then be used to aid in rational therapy design. In this review, we survey
modeling approaches under the umbrella of the major challenges facing immunotherapy development,
which encompass tumor classification, optimal treatment scheduling, and combination therapy design.
Although overlapping, each challenge has presented unique opportunities for modelers to make contri-
butions using analytical and numerical analysis of model outcomes, as well as optimization algorithms.
We discuss several examples of models that have grown in complexity as more biological information has
become available, showcasing how model development is a dynamic process interlinked with the rapid
advances in tumor-immune biology. We conclude the review with recommendations for modelers both
with respect to methodology and biological direction that might help keep modelers at the forefront of
cancer immunotherapy development.

1 Introduction immunotherapy, i.e. therapy that boosts the func-


tion of the patient’s own immune system in target-
ing the cancer. The development of a knowledge-
base of tumor-immune interactions and basic and
The involvement of the immune system in all stages clinical work on cancer immunotherapy has been
of the tumor lifecycle, including prevention, mainte- paralleled by the development of mathematical and
nance, and response to therapy is now recognized as computational models that utilize this knowledge-
central to understanding cancer development from base to design in silico model systems upon which
a systemic point of view. Therefore, it is not sur- immune-based and other treatments can be mod-
prising that one of the most rapidly developing and eled. In the best case scenario, these models can
exciting fields in cancer treatment is that of cancer
∗ Corresponding author: laubenbacher@uchc.edu

1
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

serve to guide clinicians and developers of clinical take these antigens and, if properly activated by
trials towards optimizing mono- and combination other factors in the tumor microenvironment, differ-
therapies and basic scientists in understanding the entiate to present these antigens to lymphoid cells
underlying mechanisms of the effectiveness (or, in- via their MHC class I and II molecules. Activa-
effectiveness) of therapy combinations. Therefore, tion of cytotoxic CD8+ T cells results in their pro-
in a field as rapidly developing and clinically impor- liferation and trafficking to the tumor site in or-
tant as cancer immunotherapy, mathematical and der to kill the tumor cells, leading to the release
computational modeling can play a central role in of more tumor-associated antigens and thus repeat-
helping to guide the direction the field takes. ing the cycle. This process by which the immune
In this review, we survey the mathematical system keeps a tumor in check is defined as cancer
modeling work in cancer immunotherapy organized immunosurveillance. Via a counter-process termed
by the ’major challenges’ that the immunotherapy immunoediting, a tumor can evolve and strengthen
community is currently grappling with. We will also various mechanisms to escape immunosurveillance
outline strategies that modelers can use to further [25, 59, 49].
enhance their contribution to addressing these chal- The goal of cancer immunotherapy is to boost
lenges. the response of the immune system to the tumor by
This review is organized as follows. In Section 2, intervening at one or several points of the cancer-
we give a brief overview of tumor immunology and immunity cycle. The antigen-based activation of
cancer immunotherapy. We also outline ‘major chal- dendritic cells can be achieved by administration
lenges’ that have been posed in the immunotherapy of a therapeutic vaccine harboring one or multi-
community. In Sections 3-5, we survey how each ple tumor-associated antigens, with or without ad-
challenge has been addressed by the modeling com- ditional factors that prime dendritic cells for acti-
munity; in Section 6, we provide recommendations vation. Another approach has been to extract pe-
as to what techniques the community can employ ripheral blood monocytes and stimulate them to
to further progress on each challenge and to address become DCs via presentation of antigens plus cos-
other rising challenges, and in Section 7, we sum- timulatory molecules ex vivo before reintroducing
marize our findings. them into the patient. An FDA-approved exam-
ple of the latter is the drug Provenge (sipuleucel-T;
2 The major challenges for can- Valeant Pharmaceuticals), which improves median
survival time in advanced prostate cancer by ∼ 4%
cer immunotherapy [38]. Therapies that directly boost anti-tumor T
cell activity include adoptive T cell therapy and in-
A tumor can, in principle, be recognized and con- hibition of T cell checkpoint molecules. In adoptive
trolled by the patient’s immune system via a coor- cell therapy (ACT), T cells are collected from a pa-
dinated process that has recently been summarized tient, expanded ex vivo, and reintroduced into the
as the ‘cancer-immunity’ cycle [14]. Dying tumor patient. This method has been highly successful in
cells release proteins that contain unique tumor- melanoma, and is being explored for other cancers
specific antigens that are either mutated or differen- [56]. Checkpoint therapy involves antagonizing T
tially modified post-translationally relative to nor- cell inhibitory receptors (such as CTLA-4 or PD-
mal cells [29]. Tumors also release inflammatory 1). Reciprocally, there are a number of activating
signals in the form of cytokines and other factors costimulatory receptors on T cells (such as OX40,
(such as heat-shock proteins) that lead to a lo- 4-1BB, GITR, and CD27) that can be engaged to
cal innate inflammatory response. Dendritic cells boost T cell clonal expansion and acquisition of tu-
(DCs), which form a part of this response, can up- moricidal effector functions [46]. Major successes

2
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

using this method include the FDA-approved Yer- include:


voy (ipilimumab; Bristo-Myers Squibb), a mono- 1. Tumor classification for treatment and predic-
clonal antibody (mAb) to CTLA-4 that is used for tion of response.
patients with melanoma [66, 32]. In 2016, the FDA
approved Tecentriq (atezolizumab; Genentech), an 2. Optimal scheduling and dosage of treatment.
inhibitor of the ligand for PD-1 (PD-L1) that is ex-
pressed on tumor cells and that would otherwise 3. Design and identification of combination
engage PD-1 on tumor-infiltrating T cells, for treat- treatment regimes.
ment of locally advanced or metastatic bladder can- In this review, we use these ‘major challenges’ as a
cer [63]. For a more extensive discussion of cancer means by which to present how mathematical mod-
immunotherapies and drugs in clinical development, eling can help to address each challenge and thereby
see [46, 14, 35]. help to progress the field of immunotherapy research
and application. A summary of the tumor-immune
There are particular challenges that present and immunotherapy interactions consistently ad-
themselves in almost all applications of cancer im- dressed by the models across these challenges is
munotherapy, and have been addressed by the depicted in Figure 1.
mathematical community. These major challenges

Figure 1: Summary of modeling efforts in immunotherapy. Generally, models (both mathematical and computational) are
at the cell-population level and consist of tumor and immune cells (immune cells may be represented cumulatively as effector
cells or more specifically by cell-types), cytokines, and other immune-activating or inhibiting factors such as VEGF or TGFβ,
with therapies (in green) targeting any of these factors. Models can assume spatial homogeneity (ordinary differential equation,
ODE models) or be spatially heterogeneous (agent-based and partial differential equation (PDE) models). For addressing tumor
classification for treatment (Challenge 1, Section 3), analytical, numerical, and/or parameter sensitivity analysis is performed
on the models to determine which system parameters contribute most strongly to prediction of treatment response. Control
theory and evolutionary algorithms (depicted by cogwheels) are applied to modeled treatments to optimize scheduling and
dosage (Challenge 2, Section 4), and all these methods can be applied to multiple treatments, including non-immune therapy
such as chemo- and molecular therapy, to model combination treatment regimes (Challenge 3, Section 5). Abbreviations: CTL,
cytotoxic T cells; DC, dendritic cells; CD4, CD4+ T cells, M, macrophages.

3
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

3 Challenge: Tumor classifica- negative effect of E-induced tumor destruction on


T , and the positive effect of tumor-resident E on IL
tion for treatment and pre- production. Tumor growth is modeled using the lo-
diction of response gistic growth function. The authors show that with
no therapy, if c is below a critical c0 , the only stable
steady state is a large tumor. As c increases, the
Tumor classification, usually based on histopatho- tumor size oscillates between large and small, with
logical grading, does not serve as a strong predic- the time it spends in its ’large’ state (and the magni-
tive tool for post-treatment outcome. More sophis- tude of the ’large’ state) decreasing with increasing
ticated methods of tumor classification, such as im- c. Adding therapy s1 creates a tumor-free equilib-
mune profiling [30], inclusion of markers of the tu- rium that is stable if s1 > s1crit where s1crit depends
mor microenvironment [51], and incorporation of on several parameters of the model. A bifurcation
high-throughput data [62], can improve the predic- diagram of s1crit vs. c shows that there exist regions
tive strength of the classifications. Nevertheless, where the tumor will either die or survive depending
due to both the increasing availability of patient on c, hence providing a mathematical basis for tu-
data and treatment options, it can become diffi- mor classification for treatment. The authors show
cult to predict how a patient with a specific set of similar results for s2 > 0 and both s1 and s2 > 0.
tumor characteristics will respond to a given treat-
Models with a relatively small number of equa-
ment. Modeling efforts in this regard have been used
tions can be analyzed in this way and have con-
to identify which patient parameters may be most
sistently shown that knowledge of system parame-
important to predicting therapy outcomes. Since
ters can help lead to tumor classification for treat-
mathematical models, unlike statistical models, are
ment via bifurcation and stability analysis (e.g.
typically mechanistic, they can be used to predict
[50, 8, 6, 41]). Identification of parameters where
the effect of therapy or therapy combinations that
thresholds exist with respect to system response to
have not yet been tried in the clinic (i.e. for which
therapy can aid in the identification of effective ther-
patient data is as yet unavailable). We begin with
apies. For example, for the system modeled in [40],
the Panetta-Kirschner (PK) model [40], one of the
a therapy that increases the antigenecity, c, of the
first to layer immunotherapy into a tumor-immune
tumor can push the tumor past the critical point
model. The PK model consists of system of three
necessary for response to other therapies and/or re-
ordinary differential equations (ODEs) that model
duction in size to a small, stable steady state. An
the dynamics of effector (E) and tumor (T ) cells,
excellent review of simpler tumor-immune models
and the cytokine IL-2 (IL ). For the sake of expe-
and their associated analyses, which may or may not
diency and since one of the parameters is especially
incorporate immunotherapy, can be found in [26]. A
important in the analysis of model behavior, we only
meta-modeling approach was taken by d’Onofrio et
show the equation for E,
al. [22] to synthesize the analytical results of such
dE p1 EIL models. These models can serve as baseline models
= cT − µ2 E + + s1 . (1)
dt g1 + IL that can be extended to incorporate immunother-
The antigenecity of the tumor is modeled by the apy.
parameter c, µ2 represents the death rate of E, and Numerical analysis of models has also focused
the proliferative effect of IL-2 on E is in Michaelis- on the concept of thresholds for predicting patient
Menton form to model saturation of response. Ther- response. For example, Kronik et al. [42] set out to
apies are considered by terms s1 and s2 added to the answer the question whether ‘mathematical model-
right-hand side of dE/dt and dIL /dt, respectively. ing would help to define the prerequisites of an ef-
Michaelis-Menton terms are also used to model the fective immunotherapy approach’. They extended a

4
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

previously published model of T cell transfer im- promoting’) macrophages, and live and dead tu-
munotherapy for glioblastoma [43] to a model of mor cells. In addition to secreted factors, oxy-
ex vivo expanded tumor-specific T cell transfer for gen diffusion was also implemented in the model,
melanoma and used clinical data to retroactively thereby creating oxygen-rich and hypoxic regions
validate it. The model consists of a system of ODEs in the tumor and its microenvironment. The au-
with 5 equations representing tumor and immune thors used a multiparametric sensitivity analysis
cells, and critical signaling molecules produced by (MPSA) to observe that the ratio of M2 to other
the two cell populations (TGFβ, IFNγ, and MHC cell types (termed the Macrophage Polarization In-
Class I molecules). The authors varied initial tumor dex, MPI) early in the simulation is predictive of
size and growth rate to imitate a virtual population tumor survival. Another key predictive outcome of
of patients with heterogeneous tumor profiles. Four the model was that increased tumor heterogeneity
different T-cell therapy regimens were then simu- is associated with tumor survival. Tumor hetero-
lated over this population that corresponded to four geneity was associated with locally elevated stim-
different clinical trials. For one such trial, the model ulated macrophages, leading to local peaks of M2
was able to offer an explanation for the lack of a differentiation which increased overall MPI. It fol-
dose-response relationship with therapy: the model lowed that a decrease in secretion of differentiation
showed that for patients with large enough tumors, factors for M2 cells abrogated the association be-
the therapy would have no effect, whereas in smaller tween tumor heterogeneity and survival in the sim-
tumors a dose-response relationship could be iden- ulation. Therefore, the model provided a novel hy-
tified, again pointing to a threshold for therapy ef- pothesis for tumor-induced immunosuppression via
fectiveness, this time via simulation in lieu of a bi- increases in tumor-heterogeneity, but also a poten-
furcation analysis. Indeed, the authors suggested tial new prognostic tool for tumors where biopsies
that volumetric tumor analysis is a better predictor and cell-specific stains are available. This observa-
for clinical effectiveness of T cell therapy than the tion was further used by the authors to hypothesize
traditional staging method. The authors also called and test in silico that introduction of genetically
for large doses of T cell therapy due to presence of engineered macrophages that could block the M2
thresholds for efficacy of T cell killing with respect transition would be effective in killing the tumor by
to T cell concentrations, although this recommenda- blocking the feedback loop that generates localized
tion should be considered in light of potential toxi- sites of highly concentrated immunosuppressive M2
cities. More broadly, parameter sensitivity analysis cells, thereby constructing a novel treatment using
has played a critical role in the numerical analysis their observations of the most critical contributors
of models of immunotherapy (e.g., [20, 5, 37, 3]) by to tumor growth.
helping to elucidate which tumor characteristics are Lattice- and/or agent-based models (ABMs) can
most predictive of therapy success. provide additional information regarding spatially
To investigate how spatial organization of dif- explicit parameters influencing tumor-immune in-
ferent cell types can impact tumor-immune evolu- teractions and response to immunotherapies. In
tion and response to therapy, Wells et al. [67] de- addition to [67], this approach has been used by
veloped a hybrid discrete-continuous (HDC) agent- Papalardo et al. [53, 52] to develop an agent-based
based model. These models treat cells as agents that simulator of the Triplex vaccine, SimTriplex, which
sit on a lattice and can interact with and respond to is effective in preventing mammary carcinoma in
other cells in a stochastic manner through growth HER-2/neu transgenic mice, and Dréau et al. [24]
factors and cytokines that can diffuse throughout to examine the interactions between a vascularized
the lattice. The cell types in this model were tumor and the immune response. While partial dif-
naive, M1 (’tumor-suppressive’), and M2 (’tumor- ferential equation (PDE) models have been much

5
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

more sparsely employed to model immunotherapy, on x and u via calculation of an extremal of an ob-
they can be especially useful when modeling a large jective functional
number of interacting cells in a tissue, in which case Z T
building a corresponding ABM would be too compu- J(u) = r(x(t), u(t))dt + g(x(T )), (3)
0
tationally costly. For example, Eikenberry et al. [27]
developed a PDE of melanoma with immune infil- where r is termed the running payoff and g the
trate, and showed that surgical removal of primary terminal payoff [28]. This formulation is termed
tumors with high levels of immune infiltrate could the Bolza form. The optimal u∗ found using this
promote growth of satellite metastases, as was ob- method then represents the best possible therapy
served clinically, thereby providing a model-based given conditions on the control parameters (such
hypothesis for tumor classification with respect to as minimization of tumor cells, minimization of
responsiveness to therapy (in this case, surgery). therapy dosage, maximization of immune response,
etc.).
Optimal control theory has been applied exten-
sively to the Panetta-Kirschner (PK) model [40], de-
4 Challenge: Optimal schedul- scribed in Section 3. Burden et al. [7] made ACI
ing and dosage of treatment therapy in the PK model into a control parameter,
u, and built an objective functional
Z T
1
JB (u) = [x(t) − y(t) + z(t) − B · u(t)2 ]dt, (4)
0 2
For any given treatment, an exhaustive experimen-
tal search for optimal dosage and/or scheduling is where x(t) are activated effector cells, y(t) are the
unrealistic. There are several techniques which de- tumor cells, z(t) the local concentration of IL-2,
velopers of mathematical models for immunother- and B the strength of patient tolerance to treat-
apy have used to identify optimal treatment sched- ment (i.e., B is inversely correlated to side effects of
ules in silico, including optimal control and genetic treatment). For t ∈ [0, T ], The class of admissible
algorithms. controls was set to
U = {u(t) piecewise continuous |0 ≤ u(t) ≤ 1}
4.1 Optimal control theory and u∗ was found such that max0≤u≤1 JB (u) =
Optimal control theory has played a prominent J(u∗ ), which minimized tumor mass and therapy
role for using mathematical models of cancer im- administration, and maximized effector cell and IL-
munotherapy for design of optimal therapy regimes. 2 concentration. A down-side to the model was
We can consider a system of ordinary differential that at the end of treatment, tumor mass tended
equations, to start regrowth. Ghafferi and Naserifar [31] im-
( proved upon the model of Burden at al. [7] by in-
ẋ = f (x(t), u(t)) (t > 0) cluding a linear penalty, −ωy(tf ), where ω is con-
(2) stant, y is the quantity of cancer cells and tf is the
x(0) = x0
final time-point of observation, such that their ob-
where x ∈ Rn and u ∈ U ⊂ Rm is a control parame- jective functional is JG (u) = −ωy(tf ) + JB (u) (note
ter (e.g. x(t) can represent concentrations of tumor that −ωy(tf ) is the terminal payoff for JG (u), as
cells, immune cells and cytokine and u(t) can rep- defined above). The updated objective functional
resent an immunotherapy treatment) and an initial allowed for identification of a treatment that stops
condition x0 . The optimal u, u∗ , is chosen from a tumor regrowth and also acts faster than the treat-
space U of admissible controls based on constraints ment identified with JB (u).

6
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

Similarly, Castiglione and Piccoli [11] used opti- of the system, indicating that the parametrization
mal control for a model they developed to investi- of the system from patient characteristics or other
gate dendritic cell vaccine (DCV) therapy for solid relevant knowledge should be performed carefully.
avascular tumors. For N total number of vaccina- Castiglione and Piccoli [10] further expanded the
tions and η total time for one vaccination, the space cost functional in [54] to include, in addition to the
of admissible controls, U , was taken to be previous constraints, drug holidays and minimiza-
tion of vaccine injected to lower toxicity. The au-
U = {us : S ∈ S },
thors also considered different types of cost func-
where S = {ti : i = 0, ..., N − 1, 0 ≤ t0 ≤ T − η} tions: continuous, impulsive (similar to Equation 5,
is the space of DCV injection schedules, S , and for but with η = 0), or hybrid (Equation 5, η 6= 0), to
every S in the space of schedules US , u was taken take into account the different scales between du-
to be ration of drug injection and tumor growth. The
N
X −1 hybrid method was found to be most effective for
us (T ) = ū(t − ti )χ[ti ,ti+η ] , (5) their system and the optimal treatment schedule
i=0 that was identified consisted of a high initial dose
where ū : [0, η] 7→ [0, V̄ ], with η the total time of and repeated smaller follow-up doses.
vaccination, V̄ maximum vaccine amount, and χ the These examples of optimal control theory ap-
indicator function. For every us , the therapy con- plied to cancer immunotherapy show that the con-
sisted of vaccine injections, modeled by ū, at time- trols are developed slowly, with complex control
intervals [ti − η, ti ] for all ti in S. The objective functions often built upon more simple and estab-
functional (here termed the cost functional since it lished ones.
was minimized) was taken to be the final value of
the tumor mass M , and the optimal control prob- 4.2 Genetic algorithms
lem was stated as follows: for initial condition x0 ,
what is the schedule S ∈ US such that M attains For computational agent-based models, function op-
a minimum? The problem stated as such is known timizers such as genetic algorithms (GAs) are more
as the Mayer form, which can be used when the suitable. GAs are part of a class of evolutionary
running payoff, r, is zero in Equation 3. The au- algorithms that work as follows: a set of n ini-
(1)
thors took T =6 months, N = 10 DCV injections, tial binary strings, {Si }ni=1 , sometimes termed
and η = 0. An initial schedule S0 was chosen ran- ‘chromosomes’, representing possible solutions to
domly and 2000 optimization steps were taken to the problem of interest are processed via an evalu-
find the optimal S̄, which resulted in almost com- ation function, Ei = Ei (Si ), which gives a measure
plete clearance of a tumor. Nevertheless, resurgence of the string’s performance, and a fitness function,
of tumor cells occured between vaccinations and the Fi = Fi ({Ei }), that compares the performance of all
final value of the tumor mass was highly dependent the strings to each other. In the intermediate selec-
(1)
on the timing of the last vaccination. tion phase, strings Si are kept and duplicated with
In order to correct for this effect, Piccoli and probability proportional to their respective fitness
Castiglione [54] expanded the cost functional in [11] functions, Fi . The strings of this intermediate popu-
(1∗)
to also minimize the time during which the tumor lation, {Si } are then recombined with each other
is above a maximum mass, M max , necessitating a by cross-over and mutated at a low rate to obtain
(2)
switch back to the Bolza form for their objective a new generation of strings, {Si } (note that the i
functional. The authors emphasized that the opti- do not represent the same string between the gener-
mal injection schedule u∗ obtained is highly depen- ations (1), (1*), and (2)). This process is repeated
dent on the parameter values and initial conditions until an ‘optimal‘ string is found, either via running

7
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

the algorithm for a fixed number of generations, or mutated), an optimal string (i.e. vaccination sched-
until a string is found that surpasses a threshold set ule) was selected. Also, in this case, Equation 6 was
by the fitness function. This optimal string, like × referred to as the ‘fitness’ function, which is not to
u∗ in optimal control theory, represents an optimal be confused with our definition above.
therapy solution given constraints imposed by the An extension of this fitness function was ap-
evaluation and/or fitness functions. Importantly, plied in the algorithm in [44] which incorporated
an agent-based or other computational model can several instances of in silico mice with varying im-
be run with any possible therapy (’string’), and mune backgrounds to choose the optimal vaccina-
the results can be used to determine the output of tion schedule, S ∗ . This schedule was applied to two
the evaluation function, thus the optimal string will sets of 100 virtual HER-2/neu positive mice and re-
give the optimal therapy vis-a-vis the computational sulted in 90% survival rate (this is in comparison
model. We have described the ‘canonical genetic al- to a chronic protocol, which requires 4 vaccinations
gorithm’ [68] originally developed in [33], and there in a 2-week ’on’ and 2-week ’off’ protocol, over the
exist many modifications that will not be presented course of at least one year, and has 100% success
here [68, 48]. rate, but is not realistic for clinical development).
Lollini et al. [44] set out to use a GA to de- A previous ’trial and error’ schedule was found to
velop an optimal vaccine schedule for the agent- reduce the number of injections by ∼ 27% over the
based SimTriplex model [53] described in Section chronic schedule, whereas the one identified by the
3. The Triplex vaccine is a cell-based vaccine GA reduced it by ∼ 42%, yielded excellent survival,
that stimulates an immune response via engineered and generated similar immune dynamics to the cur-
HER-2/neu positive cells that express allogenic rent protocol. In a follow-up study, Palladini et al.
MHC Class-1 molecules (for increased recognition [52] showed excellent agreement between the in sil-
by CD8+ T cells) and IL-12, which boosts both hu- ico predictions and in vivo experiments, and made
moral and adaptive immunity [16]. The GA was additional observations, such as the importance of
initialized with 80 binary 1200-bit strings, where vaccination density in the early phase of the im-
each bit represents a time-step where a vaccine in- mune response and dependence of vaccine success on
jection is/is not (1/0) given. For each string, the age of the mouse. GAs and evolutionary algorithms
SimTriplex simulator was used to determine mouse have also been used to identify potential epitopes for
survival time (s) and maximum number of cancer vaccine development [4], to develop an initial guess
1 2
cells in the transient (Ncc ) and steady (Ncc ) tumor for a discrete optimal control problem based on the
growth phases. The evaluation function was then model in [10], [47], and to solve a multi-objective
taken to be, optimization problem for a model of combination
chemo- and immunotherapy [39].
n2
f (n, s, β) = · β, (6) By example of optimal control theory and ge-
s
netic algorithms, we have shown how mathematical
where n is the number of injections specified by the methods in optimization used in conjunction with
1 2
test string, and β = β(Ncc , Ncc ) is proportional to appropriate models yield treatment schedules de-
the maximum number of cancer cells. Note that in rived in a systematic, rather than experimental or
this implementation, the aim was to minimize the computational ‘trial and error’ method, that can be
fitness function. Via tournament selection (the fit- used not only to create optimal treatment schedules,
ness functions of two or more strings are compared but to also better understand immune response to
and the probability of selecting a string is propor- treatment, such as the observations obtained from
tional to the fitness ranking of the string), followed both methods above to the relative importance of
by mutation and elitism (the best strings are not early vs. late immunization protocols.

8
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

5 Challenge: Design and iden- of combination vs. mono-therapy differed based on


initial patient characteristics. or example a patient
tification of combination with higher CD8+ T cell efficacy would benefit more
treatment regimes strongly from immuno- or combination therapy than
a patient with low efficacy, since an injection of IL-2
for the latter group would result in more CD8+ T
Just as combination therapy using non- cells, but not enough to increase overall anti-tumor
immunotherapeutic approaches is a mainstay in response. Notably, had the authors modeled an im-
cancer treatment, combination immunotherapy ei- munotherapy that altered these variables directly
ther with just immunotherapeutic agents or with (such as injection of CD8+ T cell costimulator ago-
immune- and non-immunotherapeutic agents can nists that boost CD8+ effector activity), they would
and should be designed rationally to maximize have obtained different success rates for the therapy
treatment response [46, 51]. Modeling can con- combinations. Nevertheless, the model can serve as
tribute to this process as it can aid in the develop- a forerunner to models parametrized with patient-
ment of a mechanistic understanding for effective- specific parameters that can then be used to identify
ness of combination vs. mono-therapies and make optimal therapy combinations.
the search for an optimal combination therapy more
efficient. For example, de Pillis et al. developed a Targeted therapies, such as antibodies against
system of six ODEs for combination chemo- and the breast cancer cell receptor HER2 and EGFR,
immunotherapy that included tumor, NK, CD8+ , elicit a strong, adaptive immune response, raising
and white blood cells, as well as bloodstream con- the implication that combination immunotherapy
centrations of chemotherapy (doxorubicin) and im- with targeted therapy can achieve a synergistic anti-
munotherapy drugs, the latter of which include tumor immune response [64, 70]. The humanized
tumor-infiltrating-lymphocyte (TIL) therapy and antibody to VEGF, Avastin (bevacizumab), is FDA-
IL-2 stimulation [17]. The model extended an ear- approved for a number of different cancers, and in
lier model by de Pillis et al. [18] to include more re- addition to blocking the pro-angiogenic activity of
cent biological findings. The effect of chemotherapy, tumor-produced VEGF, also blocks the immuno-
M , was modeled using a saturation term, 1 − e−δM , suppressive activity of VEGF, which includes sup-
where δ represents the efficacy of M , and the boost pression of DC maturation [36]. Soto-Ortiz et al.
to CD8+ activity by addition of IL-2 was modeled [61] hypothesized that a combination therapy of an
using a Michaelis-Menten interaction term, similar anti-VEGF antibody followed by administration of
to what we see in Equation 1 from [40]. Cell-cell DC cells would result in a strong anti-tumor re-
interaction terms were modeled using previously fit sponse by the immune system. The authors built
equations, often of Michaelis-Menten type. For ex- upon a model that focuses on tumor-immune inter-
ample, the CD8+ T cell stimulation by the tumor actions [55] to develop a system of 18 ODEs that in-
was taken to be clude tumor, immune, and vascular endothelial cells,
and a number of cytokines and growth factors in-
dL T cluding IL-2, TGFβ, and VEGF (TGFβ and VEGF
∝j L, (7)
dT k+T are both considered to be immunosuppressive). The
where T and L represent the tumor and authors simulated treatment with injection of anti-
CD8+ populations, respectively, and j and k are pa- VEGF antibody and/or unstimulated DC cells and
rameters. Following [18], several parameters of the analyzed the results numerically. The simulations
model, which collectively determine CD8+ T cell showed that for tumors with low immunosuppres-
efficacy at tumor cell killing, were fitted to patient- sion and high antigenecity, which is represented by
specific data. The authors found that the success a parameter that models the strength of matura-

9
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

tion of DCs after encounter with a tumor antigen, 6 Recommendations


the immune system can keep the tumor at a small
size and administration of either therapy can kill the
entire tumor. When tumor antigenecity was low- 6.1 Intracellular and multi-scale
ered and immunosuppression increased, the simu-
modeling
lated tumor grew and vascularized without therapy.
The authors observed that for tumors with high im- Most current immunotherapy models are developed
munosuppression, DC therapy alone was not suffi- at the resolution level of cell populations. How-
cient to kill the tumor without additional modifica- ever, modeling intracellular signaling cascades and
tion of the immunosuppressive microenvironment, metabolic responses in specific cell types can give
which is an example of a prediction for effectiveness insight into how therapies can act at the intracel-
of a mono-therapy vs. combination when given ini- lular level, and what the relative contribution of
tial system parameters. The authors made a num- different therapies is to cell response: intracellu-
ber of other observations regarding the interdepen- lar vs. cell-cell. As in the case of population-
dencies of tumor growth, antigenecity, and immuno- level models of immunotherapy, where the therapy
suppression that would not have been possible in a component is often added to an existing, validated
simpler model. A prediction arising from this anal- model of tumor-immune interactions, the intracel-
ysis was that there exists a therapeutic window for lular models of therapy can build on known sig-
optimal effectiveness of different immunotherapies naling pathways that interact with proposed ther-
that is dependent on the tumor size and growth rate, apies in tumors and T cells. Intracellular signal-
and thus knowledge of these tumor characteristics ing models have been developed for cancer signal-
can be used to design the optimal mono- or com- ing pathways for a variety of tumor types[34, 2],
bination therapy for a given tumor/patient. More- and can be modified to include subpathways of in-
over, synergy in combination therapy was predicted terest, such as induction of PD-L1 or secreted im-
to occur when immunotherapy followed anti-VEGF munosuppressive factors. Saez-Rodriguez et al.[58]
treatment, thus the model was able to address ques- built a Boolean network model of a signaling net-
tions of efficacy prediction and optimal time and se- work activated upon T cell receptor and corecep-
lection of treatment, and hence effectively address tor activation and validated it using both literature
all the major challenges we have discussed. and experimental wild-type and knock-out data that
Models for various combinations of traditional matched in silico predictions. This network could
and immunotherapies have been developed [9, 23, be made more specific for different T cells (CD4+,
69, 60], which generally build on earlier mono- CD8+, Treg), receptors of therapeutic interest (e.g.
therapy, tumor-immune, or even combination ther- CTLA-4, PD-1) and their downstream signaling tar-
apy models. For example, Chareyron and Alamir gets, which can be added to the model in order to
[12] extend earlier models developed by de Pillis et examine how therapy would affect T cell activation
al. [19, 20] to include chemo- and immunotherapy status. Modification of networks identified in the
and use control theory to determine optimal therapy literature may require additional experimental in-
protocols, again addressing multiple big challenges put, depending on the current availability of data.
discussed herein. These models can give insights as Such models would allow a more granular systems-
to mechanism and efficacy of combination therapies level analysis of mechanisms of pharmaceutical ac-
in order to guide clinical development. tion and subsequent therapy optimization and com-
bination. Furthermore, since immunotherapy drugs
can act on multiple cell targets, for example CTLA-
4 is expressed not only on CD8+ T cells but also

10
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

Tregs, development of multi-scale models that in- duce adverse events related to Fc-FcR interactions
corporate both the interactions of different immune (reviewed in [57]). By developing models of Fc-
and tumor cell types and their respective intracellu- FcR action in concert with experimentalists, model-
lar dynamics would have the power to elucidate the ers can hasten this process and use their models to
multi-scale mechanisms of therapy action. Indeed, guide development of optimized therapy options.
multi-scale modeling has already been identified as a
important method for generating systems-level pre-
dictions of cancer progression and therapy effective-
ness [21, 65].
7 Conclusion

Immunotherapy is one of the most exciting recent


6.2 Addressing toxicity
developments in cancer treatment, with new drugs
Immune-mediated toxicities, including retinal dys- coming on the market at an increasing rate, for
function, liver toxicity, and pancreatitis can oc- an ever-larger number of cancers. But, as this
cur upon administration of immunotherapy. Tox- review highlights, many unanswered questions re-
icity, which is generally caused by targeting of main, and the field faces several challenges. Math-
self-antigens by the drug-strengthened immune re- ematical modelers have addressed these challenges
sponse, is often associated with clinical response as early as the 1980’s (e.g., [15]). The models we
[1]. Incorporation of immunotherapy-related tox- have reviewed here and their use in understanding
icity into models can result in estimates of ther- various aspects of immunotherapy crucial for effec-
apy dose that take into account toxicity-related ef- tive treatment, show that mathematical and com-
fects, as was already done in [7]. Development of putational models can play the role of a key en-
more mechanistic models can help to optimize ther- abling technology to improve the application of this
apies to maximize effectiveness and minimize tox- type of treatment. However, as this review also dis-
icity if mechanisms for each are not entirely over- cusses, there is much work to be done. Almost all
lapping. This may be especially important in mod- immunotherapy-specific modeling efforts we could
els of combination immunotherapy, where toxicity- identify focus on the population level, even though
related events tend to be more common than with it is clear that a multi-scale approach is needed that
monotherapies [45]. One route of interest towards also incorporates the molecular scale. To aid this
this goal can be a joint experimental-modeling fo- process and to stay at the forefront of immunother-
cus on Fc-FcR interactions. Immunomodulatory apy technology development, high-throughput data
mAbs such as ipilimumab, a mAb to CTLA-4, are derived from technologies such as microarrays and
most often of IgG isotope: in addition to contain- RNA-Seq for cell-level transcriptional information
ing two antigen-binding (Fab) domains, they con- and mass cytometry (CyTOF) for population-level
tain a fragment crystallizable (Fc) ’tail’ that can marker distribution can be incorporated into model
bind Fc receptors (FcRs) found on effector cells development [13].
such as macrophages and DCs. Fc-FcR interactions Collaborations between modelers, basic scien-
tists, and clinicians are crucial for the realization of
have been shown to contribute to cytokine-related
the translational potential of mathematical model-
adverse events during mAb treatment, and hence
ing in the service of a precision medicine approach to
modulation of these interactions during mAb de- this revolutionary new cancer treatment. As many
sign could help to decouple therapeutic from toxic of the models show, parameter values matter greatly
responses. A better mechanistic understanding of in predicting treatment outcomes and devising op-
Fc-FcR engagement and response during treatment timal treatment approaches. Personalized models
could help lead to rational design of mAbs that re- that can be calibrated with parameters characteris-

11
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

tic of an individual patient will turn mathematical [7] Thalya Burden, Jon Ernstberger, and K. Renee Fis-
models into powerful tools that can play an essential ter. Optimal control applied to immunotherapy.
role at the bedside, not just in the laboratory. Discrete and Continuous Dynamical Systems - Se-
ries B, 4(1):135–146, 2004.
[8] HM Byrne and CE Kelly. Macrophage-tumour in-
Acknowledgments teractions: In vivo dynamics. Discrete and Con-
tinuous Dynamical Systems - Series B, 4(1):81–98,
2004.
AK gratefully acknowledges support from the National [9] Antonio Cappuccio, Filippo Castiglione, and
Cancer Institute of the National Institutes of Health Benedetto Piccoli. Determination of the optimal
postdoctoral fellowship award F32CA214030. RL grate- therapeutic protocols in cancer immunotherapy.
fully acknowledges support from the National Cancer In- Math Biosci, 209(1):1–13, Sep 2007.
stitute of the National Institutes of Health under grant
[10] F Castiglione and B Piccoli. Cancer immunother-
1R01CA188025-01.
apy, mathematical modeling and optimal control. J
Theor Biol, 247(4):723–32, Aug 2007.
[11] Filippo Castiglione and Benedetto Piccoli. Optimal
References control in a model of dendritic cell transfection can-
cer immunotherapy. Bull Math Biol, 68(2):255–74,
[1] Sally M Amos, Connie P M Duong, Jennifer A Feb 2006.
Westwood, David S Ritchie, Richard P Junghans, [12] S. Chareyron and M. Alamir. Mixed immunother-
Phillip K Darcy, and Michael H Kershaw. Autoim- apy and chemotherapy of tumors: Feedback design
munity associated with immunotherapy of cancer. and model updating schemes. Journal of Theoreti-
Blood, 118(3):499–509, Jul 2011. cal Biology, 258(3):444–454, 2009.
[2] J Bachmann, A Raue, M Schilling, V Becker, [13] Pornpimol Charoentong, Mihaela Angelova, Mir-
J Timmer, and U Klingmüller. Predictive math- jana Efremova, Ralf Gallasch, Hubert Hackl,
ematical models of cancer signalling pathways. J Jerome Galon, and Zlatko Trajanoski. Bioinfor-
Intern Med, 271(2):155–65, Feb 2012. matics for cancer immunology and immunother-
apy. Cancer Immunol Immunother, 61(11):1885–
[3] Sandip Banerjee, Subhas Khajanchi, and Swapna 903, Nov 2012.
Chaudhuri. A mathematical model to elucidate
brain tumor abrogation by immunotherapy with [14] Daniel S Chen and Ira Mellman. Oncology meets
t11 target structure. PLoS One, 10(5):e0123611, immunology: the cancer-immunity cycle. Immu-
2015. nity, 39(1):1–10, Jul 2013.
[15] R J De Boer, P Hogeweg, H F Dullens, R A
[4] V Brusic, G Rudy, G Honeyman, J Hammer, and
De Weger, and W Den Otter. Macrophage T
L Harrison. Prediction of MHC class ii-binding
lymphocyte interactions in the anti-tumor immune
peptides using an evolutionary algorithm and artifi-
response: a mathematical model. J Immunol,
cial neural network. Bioinformatics, 14(2):121–30,
134(4):2748–58, Apr 1985.
1998.
[16] Carla De Giovanni, Giordano Nicoletti, Lorena
[5] Svetlana Bunimovich-Mendrazitsky, Helen Byrne, Landuzzi, Annalisa Astolfi, Stefania Croci, Al-
and Lewi Stone. Mathematical model of pulsed im- berto Comes, Silvano Ferrini, Raffaella Meazza,
munotherapy for superficial bladder cancer. Bull Manuela Iezzi, Emma Di Carlo, Piero Musiani,
Math Biol, 70(7):2055–76, Oct 2008. Federica Cavallo, Patrizia Nanni, and Pier-Luigi
[6] Svetlana Bunimovich-Mendrazitsky, Eliezer Lollini. Immunoprevention of HER-2/neu trans-
Shochat, and Lewi Stone. Mathematical model of genic mammary carcinoma through an interleukin
BCG immunotherapy in superficial bladder cancer. 12-engineered allogeneic cell vaccine. Cancer Res,
Bull Math Biol, 69(6):1847–70, Aug 2007. 64(11):4001–9, Jun 2004.

12
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

[17] Lisette G de Pillis, K. Renee Fister, Weiqing Gu, ematical models. Bull Math Biol, 73(1):2–32, Jan
Craig Collins, Michael Daub, David Gross, James 2011.
Moore, and Benjamin Preskill. Mathematical [27] Steffen Eikenberry, Craig Thalhauser, and Yang
model creation for cancer chemo-immunotherapy. Kuang. Tumor-immune interaction, surgical treat-
Computational and Mathematical Methods in ment, and cancer recurrence in a mathemati-
Medicine, 10(3):165–184, 2009. cal model of melanoma. PLoS Comput Biol,
[18] Lisette G de Pillis, Weiqing Gu, and Ami E Radun- 5(4):e1000362, Apr 2009.
skaya. Mixed immunotherapy and chemotherapy [28] Lawrence C. Evans. An introduction to math-
of tumors: modeling, applications and biological ematical optimal control theory, Version 0.2.
interpretations. J Theor Biol, 238(4):841–62, Feb https://math.berkeley.edu/ evans/control.course.pdf.
2006. Accessed: 2017-05-03.
[19] Lisette G de Pillis and Ami E Radunskaya. Im- [29] Olivera J Finn. Cancer immunology. N Engl J Med,
mune response to tumor invasion. In K. Bathe, 358(25):2704–15, Jun 2008.
editor, Computational Fluid and Solid Mechanics [30] Jérôme Galon, Franck Pagès, Francesco M Marin-
2003, volume 2, pages 1661–1668, Cambridge, MA, cola, Helen K Angell, Magdalena Thurin, Alessan-
2003. MIT Press. dro Lugli, Inti Zlobec, Anne Berger, Carlo Bi-
[20] Lisette G de Pillis, Ami E Radunskaya, and fulco, Gerardo Botti, Fabiana Tatangelo, Cedrik M
Charles L Wiseman. A validated mathematical Britten, Sebastian Kreiter, Lotfi Chouchane,
model of cell-mediated immune response to tumor Paolo Delrio, Hartmann Arndt, Martin Ass-
growth. Cancer Res, 65(17):7950–8, Sep 2005. laber, Michele Maio, Giuseppe V Masucci, Mar-
tin Mihm, Fernando Vidal-Vanaclocha, James P
[21] Thomas S Deisboeck, Zhihui Wang, Paul Macklin,
Allison, Sacha Gnjatic, Leif Hakansson, Christoph
and Vittorio Cristini. Multiscale cancer modeling.
Huber, Harpreet Singh-Jasuja, Christian Ottens-
Annu Rev Biomed Eng, 13:127–55, Aug 2011.
meier, Heinz Zwierzina, Luigi Laghi, Fabio Grizzi,
[22] Alberto d’Onofrio. Metamodeling tumor–immune Pamela S Ohashi, Patricia A Shaw, Blaise A
system interaction, tumor evasion and im- Clarke, Bradly G Wouters, Yutaka Kawakami,
munotherapy. Mathematical and Computer Mod- Shoichi Hazama, Kiyotaka Okuno, Ena Wang,
elling, 47:614–637, 2008. Jill O’Donnell-Tormey, Christine Lagorce, Graham
[23] Alberto d’Onofrio, Ledzewixa Urszula, and Heinz Pawelec, Michael I Nishimura, Robert Hawkins,
Schattler. New challenges for cancer sys- Réjean Lapointe, Andreas Lundqvist, Samir N
tems biomedicine, chapter On the Dynamics of Khleif, Shuji Ogino, Peter Gibbs, Paul War-
Tumor-Immune System Interactions and Combined ing, Noriyuki Sato, Toshihiko Torigoe, Kyogo
Chemo- and Immunotherapy. Springer Milan, 2012. Itoh, Prabhu S Patel, Shilin N Shukla, Richard
Palmqvist, Iris D Nagtegaal, Yili Wang, Corrado
[24] D. Dréau, D. Stanimirov, T. Carmichael, and D’Arrigo, Scott Kopetz, Frank A Sinicrope, Giorgio
M. Hadzikadic. An agent-based model of solid tu- Trinchieri, Thomas F Gajewski, Paolo A Ascierto,
mor progression. In Rajasekaran S., editor, Bioin- and Bernard A Fox. Cancer classification using the
formatics and Computational Biology, volume 5462 immunoscore: a worldwide task force. J Transl
of Lecture Notes in Computer Science, pages 187– Med, 10:205, Oct 2012.
198. Springer, 2009.
[31] A Ghaffari and N Naserifar. Optimal therapeutic
[25] Gavin P Dunn, Allen T Bruce, Hiroaki Ikeda, protocols in cancer immunotherapy. Comput Biol
Lloyd J Old, and Robert D Schreiber. Cancer im- Med, 40(3):261–70, Mar 2010.
munoediting: from immunosurveillance to tumor [32] F Stephen Hodi, Steven J O’Day, David F Mc-
escape. Nat Immunol, 3(11):991–8, Nov 2002. Dermott, Robert W Weber, Jeffrey A Sosman,
[26] Raluca Eftimie, Jonathan L Bramson, and David John B Haanen, Rene Gonzalez, Caroline Robert,
J D Earn. Interactions between the immune sys- Dirk Schadendorf, Jessica C Hassel, Wallace Ak-
tem and cancer: a brief review of non-spatial math- erley, Alfons J M van den Eertwegh, Jose Lutzky,

13
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

Paul Lorigan, Julia M Vaubel, Gerald P Linette, model can cause an allee effect. Bull Math Biol,
David Hogg, Christian H Ottensmeier, Celeste 78(4):754–85, Apr 2016.
Lebbé, Christian Peschel, Ian Quirt, Joseph I [42] Natalie Kronik, Yuri Kogan, Paul G Schlegel, and
Clark, Jedd D Wolchok, Jeffrey S Weber, Jason Matthias Wölfl. Improving T-cell immunotherapy
Tian, Michael J Yellin, Geoffrey M Nichol, Axel for melanoma through a mathematically motivated
Hoos, and Walter J Urba. Improved survival with strategy: efficacy in numbers? J Immunother,
ipilimumab in patients with metastatic melanoma. 35(2):116–24, 2012.
N Engl J Med, 363(8):711–23, Aug 2010.
[43] Natalie Kronik, Yuri Kogan, Vladimir Vainstein,
[33] John H Holland. Adaptation in natural and artifi-
and Zvia Agur. Improving alloreactive CTL im-
cial systems. University of Michigan Press. (Second
munotherapy for malignant gliomas using a simu-
edition: MIT Press, 1992), 1975.
lation model of their interactive dynamics. Cancer
[34] Jorrit J Hornberg, Frank J Bruggeman, Hans V Immunol Immunother, 57(3):425–39, Mar 2008.
Westerhoff, and Jan Lankelma. Cancer: a systems
[44] Pier-Luigi Lollini, Santo Motta, and Francesco
biology disease. Biosystems, 83(2-3):81–90, 2006.
Pappalardo. Discovery of cancer vaccination pro-
[35] Laura Jeanbart and Melody A Swartz. Engineering tocols with a genetic algorithm driving an agent
opportunities in cancer immunotherapy. Proc Natl based simulator. BMC Bioinformatics, 7:352, Jul
Acad Sci U S A, 112(47):14467–72, Nov 2015. 2006.
[36] Benjamin F Johnson, Timothy M Clay, Amy C [45] Ignacio Melero, David M Berman, M Angela Az-
Hobeika, H Kim Lyerly, and Michael A Morse. Vas- nar, Alan J Korman, José Luis Pérez Gracia, and
cular endothelial growth factor and immunosup- John Haanen. Evolving synergistic combinations of
pression in cancer: current knowledge and potential targeted immunotherapies to combat cancer. Nat
for new therapy. Expert Opin Biol Ther, 7(4):449– Rev Cancer, 15(8):457–72, Aug 2015.
60, Apr 2007.
[46] Ira Mellman, George Coukos, and Glenn Dranoff.
[37] Badal Joshi, Xueying Wang, Sayanti Banerjee, Cancer immunotherapy comes of age. Nature,
Haiyan Tian, Anastasios Matzavinos, and Mark 480(7378):480–9, Dec 2011.
A J Chaplain. On immunotherapies and cancer
vaccination protocols: a mathematical modelling [47] Andrea Minelli, Francesco Topputo, and Franco
approach. J Theor Biol, 259(4):820–7, Aug 2009. Bernelli-Zazzera. Controlled drug delivery in can-
cer immunotherapy: Stability, optimization, and
[38] Philip W Kantoff, Celestia S Higano, Neal D
monte carlo analysis. SIAM Journal on Applied
Shore, E Roy Berger, Eric J Small, David F
Mathematics, 71(6):2229–2245, 2011.
Penson, Charles H Redfern, Anna C Ferrari,
Robert Dreicer, Robert B Sims, Yi Xu, Mark W [48] Melanie Mitchell. An Introduction to Genetic Al-
Frohlich, Paul F Schellhammer, and IMPACT gorithms. MIT Press, Cambridge, MA, USA, 1998.
Study Investigators. Sipuleucel-T immunotherapy [49] Deepak Mittal, Matthew M Gubin, Robert D
for castration-resistant prostate cancer. N Engl J Schreiber, and Mark J Smyth. New insights into
Med, 363(5):411–22, Jul 2010. cancer immunoediting and its three component
[39] Kanchi Lakshmi Kiran and S. Lakshminarayanan. phases–elimination, equilibrium and escape. Curr
Optimization of chemotherapy and immunother- Opin Immunol, 27:16–25, Apr 2014.
apy: In silico analysis using pharmacokinetic– [50] F Nani and H I Freedman. A mathematical model
pharmacodynamic and tumor growth models. Jour- of cancer treatment by immunotherapy. Math
nal of Process Control, 23(3):396–403, 2013. Biosci, 163(2):159–99, Feb 2000.
[40] D Kirschner and J C Panetta. Modeling im- [51] Abigail E. Overacre, Sema Kurtulus, Mario Sznol,
munotherapy of the tumor-immune interaction. J Drew M. Pardoll, Ana Anderson, and Dario A. A.
Math Biol, 37(3):235–52, Sep 1998. Vignali. Combination immunotherapy: Where do
[41] Anna Konstorum, Thomas Hillen, and John Lowen- we go from here? J Immunother Cancer, 3(38),
grub. Feedback regulation in a cancer stem cell 2015.

14
bioRxiv preprint first posted online Jun. 9, 2017; doi: http://dx.doi.org/10.1101/146902. The copyright holder for this preprint (which was not
peer-reviewed) is the author/funder. It is made available under a CC-BY-NC-ND 4.0 International license.

[52] Arianna Palladini, Giordano Nicoletti, Francesco Joseph Ciccolini, Fabrice Barlesi, Xavier Muracci-
Pappalardo, Annalisa Murgo, Valentina Grosso, ole, and Dominique Barbolosi. Mathematical mod-
Valeria Stivani, Marianna L Ianzano, Agnese An- eling of cancer immunotherapy and its synergy with
tognoli, Stefania Croci, Lorena Landuzzi, Carla radiotherapy. Cancer Res, 76(17):4931–40, Sep
De Giovanni, Patrizia Nanni, Santo Motta, and 2016.
Pier-Luigi Lollini. In silico modeling and in vivo [61] Luis Soto-Ortiz and Stacey D Finley. A can-
efficacy of cancer-preventive vaccinations. Cancer cer treatment based on synergy between anti-
Res, 70(20):7755–63, Oct 2010. angiogenic and immune cell therapies. J Theor Biol,
[53] F Pappalardo, P-L Lollini, F Castiglione, and 394:197–211, Apr 2016.
S Motta. Modeling and simulation of can- [62] Pramod K Srivastava and Fei Duan. Harnessing the
cer immunoprevention vaccine. Bioinformatics, antigenic fingerprint of each individual cancer for
21(12):2891–7, Jun 2005. immunotherapy of human cancer: genomics shows
[54] Benedetto Piccoli and Filippo Castiglione. Opti- a new way and its challenges. Cancer Immunol Im-
mal vaccine scheduling in cancer immunotherapy. munother, 62(5):967–74, May 2013.
Physica A: Statistical Mechanis and its Applica- [63] Pam Tarapchak. Tecentriq approved for targeted
tions, 370(2):672–680, 2006. treatment of bladder cancer. Oncology Times, 2016.
[55] Mark Robertson-Tessi, Ardith El-Kareh, and Alain [64] Matthew Vanneman and Glenn Dranoff. Combin-
Goriely. A mathematical model of tumor-immune ing immunotherapy and targeted therapies in can-
interactions. J Theor Biol, 294:56–73, Feb 2012. cer treatment. Nat Rev Cancer, 12(4):237–51, Mar
[56] Steven A Rosenberg, Nicholas P Restifo, James C 2012.
Yang, Richard A Morgan, and Mark E Dudley. [65] Zhihui Wang, Joseph D Butner, Romica Kerketta,
Adoptive cell transfer: a clinical path to effective Vittorio Cristini, and Thomas S Deisboeck. Sim-
cancer immunotherapy. Nat Rev Cancer, 8(4):299– ulating cancer growth with multiscale agent-based
308, Apr 2008. modeling. Semin Cancer Biol, 30:70–8, Feb 2015.
[57] Joseph M Ryan, Jeffrey S Wasser, Adam J Adler, [66] Jeffrey Weber. Review: anti-ctla-4 antibody
and Anthony T Vella. Enhancing the safety of ipilimumab: case studies of clinical response
antibody-based immunomodulatory cancer therapy and immune-related adverse events. Oncologist,
without compromising therapeutic benefit: Can we 12(7):864–72, Jul 2007.
have our cake and eat it too? Expert Opin Biol [67] Daniel K Wells, Yishuan Chuang, Louis M. Knapp,
Ther, 16(5):655–74, 2016. Dirk Brockmann, William L. Kath, and Joshua N.
[58] Julio Saez-Rodriguez, Luca Simeoni, Jonathan A Leonard. Spatial and functional heterogeneities
Lindquist, Rebecca Hemenway, Ursula shape collective behavior of tumor-immune net-
Bommhardt, Boerge Arndt, Utz-Uwe Haus, works. PLoS Comput Biol, 11(4):e1004181, 2015.
Robert Weismantel, Ernst D Gilles, Steffen Klamt, [68] Darrell Whitley. A genetic algorithm tutorial.
and Burkhart Schraven. A logical model provides Statistics and Computing, 4(2):65–85, 1994.
insights into T cell receptor signaling. PLoS
[69] Shelby Wilson and Doron Levy. A mathematical
Comput Biol, 3(8):e163, Aug 2007.
model of the enhancement of tumor vaccine efficacy
[59] Robert D Schreiber, Lloyd J Old, and Mark J by immunotherapy. Bull Math Biol, 74(7):1485–
Smyth. Cancer immunoediting: integrating immu- 500, Jul 2012.
nity’s roles in cancer suppression and promotion.
[70] Meng Michelle Xu, Yang Pu, Yuan Zhang, and
Science, 331(6024):1565–70, Mar 2011.
Yang-Xin Fu. The role of adaptive immunity in
[60] Raphael Serre, Sebastien Benzekry, Laetitia the efficacy of targeted cancer therapies. Trends
Padovani, Christophe Meille, Nicolas André, Immunol, 37(2):141–53, Feb 2016.

15

You might also like