Informatics in Medicine Unlocked: Joseph Malinzi, Kevin Bosire Basita, Sara Padidar, Henry Ademola Adeola

You might also like

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

Informatics in Medicine Unlocked 23 (2021) 100534

Contents lists available at ScienceDirect

Informatics in Medicine Unlocked


journal homepage: http://www.elsevier.com/locate/imu

Prospect for application of mathematical models in combination


cancer treatments
Joseph Malinzi a, b, Kevin Bosire Basita c, Sara Padidar d, Henry Ademola Adeola e, f, *
a
Department of Mathematics, Faculty of Science and Engineering, University of Eswatini, Private Bage 4, Kwaluseni, Eswatini
b
Institute of Systems Science, Durban University of Technology, Durban, 4000, South Africa
c
Department of Mathematics, Faculty of Applied Science and Technology, Technical University of Kenya, Haile Selassie Avenue, Nairobi, Kenya
d
Department of Biological Sciences, Faculty of Science and Engineering, University of Eswatini, Private Bage 4, Kwaluseni, Eswatini
e
Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
f
Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, University of the Western Cape and Tygerberg Hospital, Cape Town, South Africa

A R T I C L E I N F O A B S T R A C T

Keywords: The long-term efficacy of targeted therapeutics for cancer treatment can be significantly limited by the type of
Combination cancer therapy therapy and development of drug resistance, inter alia. Experimental studies indicate that the factors enhancing
Mathematical models of combination cancer acquisition of drug resistance in cancer cells include cell heterogeneity, drug target alteration, drug inactivation,
therapy
DNA damage repair, drug efflux, cell death inhibition, as well as microenvironmental adaptations to targeted
Chemoimmunotherapy
therapy, among others. Combination cancer therapies (CCTs) are employed to overcome these molecular and
Chemovirotherapy
Chemoradiotherapy pathophysiological bottlenecks and improve the overall survival of cancer patients. CCTs often utilize multiple
Radioimmunotherapy combinatorial modes of action and thus potentially constitute a promising approach to overcome drug resistance.
Immunovirotherapy Considering the colossal cost, human effort, time and ethical issues involved in clinical drug trials and basic
Radiovirotherapy medical research, mathematical modeling and analysis can potentially contribute immensely to the discovery of
better cancer treatment regimens. In this article, we review mathematical models on CCTs developed thus far for
cancer management. Open questions are highlighted, and plausible combinations are discussed based on the
level of toxicity, drug resistance, survival benefits, preclinical trials and other side effects.

1. Introduction Computational biology and mathematical modeling have enabled re­


searchers to de-velop new treatments, from screening potential candi­
Cancer is the second leading cause of death globally, with 70% of date compounds [8] and building and identifying chemical structures
cancer mortality from low- and middle-income countries [1]. To date, that will best fit target sites [6,9] to predicting the outcome of novel
cancer treatment has heavily relied on surgery, often followed by combinations of drug regi-mens [10–17]. This paper reviews a novel use
chemotherapy and/or radiotherapy [2–4]. Recent advances with the use of mathematical modeling to enable researchers to predict the treatment
of virological and immunological agents including nanotechnology de­ outcomes of combined therapeutic strategies in the treatment of cancers.
livery methods have increased our arsenal in treating this complex dis­ Benign cancers and locally confined malignant cells can often be
ease [5]. Given the high costs involved in treating cancer, and the treated by a single treatment strategy such as surgery or radiation
continued development of better and less toxic treatment approaches, therapy alone. In some cases, a combination approach is employed such
mathematical modeling provides a valuable and low-cost tool for re­ as the use of (neo) adjuvant radiotherapy or chemotherapy before or
searchers by pre-dicting treatment outcomes and identifying potential after surgery to reduce the size of a tumour, thereby increasing chances
best combinations to therapeutic approaches with minimal adverse of complete surgical resection and/or destroying of any post-surgical
effects. residual cancer cells [18,19]. Employing combined therapeutic strate­
The use of mathematical modeling in understanding a variety of gies is essential to many malignant tumours where a single approach (e.
disease states, including cancer, has been well documented [6,7]. g. surgery) is not always suitable, for instance in bladder cancer or

* Corresponding author. Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town,
Observatory 7925, Cape Town, South Africa.
E-mail address: henry.adeola@uct.ac.za (H.A. Adeola).

https://doi.org/10.1016/j.imu.2021.100534
Received 4 September 2020; Received in revised form 29 January 2021; Accepted 5 February 2021
Available online 13 February 2021
2352-9148/© 2021 Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

non-small cell lung cancer that may not be operable.


Although mono-therapeutic strategies claim a large share in the
medical world for the treatment of several cancer types, they are
considered less effective in comparison with the combination therapy
approach [20]. Most standard mono-therapeutic strategies (e.g.
chemotherapy alone) destroy all the actively proliferating cells
non-selectively, ultimately causing the annihilation of both healthy and
cancerous cells [20]. In addition, chemother-apy is known to produce
toxic effects on the patients with a myriad of side effects, threat to life,
suppressing bone marrow cells and increasing vulnerability to infection
[20,21]. Hence, there is an overarching demand for tumour-targeted
therapies, that are minimally toxic especially in the era of precision
and personalized medicine. Combination cancer treatments hold great
appeals in boosting the efficacy of anticancer drugs, enhancing
apoptosis, suppressing tumour growth and decreasing cancer stem cell
population [22]. Although chemotherapy-based combination therapy
could be toxic, the toxicity can be greatly reduced when different
pathways are tar-geted [22,23]. Moreover, dosing for these agents works
in an additive manner, thereby lowering the cumulative amount of
therapeutic dosage needed in each individual drug [23].
Several advances have been made in tumour profiling, and deep
sequencing has revealed driver mutations as well as novel targets for the
development of new cancer drugs [24,25]. There have been remarkable
achieve-ments on combination therapy in recent years with much
attention focusing on most effective combinations that are likely to
produce significant effects on the tumour. However, this journey has not
been smooth with the process of designing plausible combinations fac­
ing numerous challenges. Current combination cancer therapy presumes
that better results are obtained by amalgamating therapeutic drugs at
their maximal tolerated doses [26]. Nevertheless, differences in the
pharmacokinetics of single drugs bring about inconsistency in the de­
livery of synergistic drug ratios to tumour cells. Compounded toxicity
observed in combination treatment protocols calls for application of
suboptimal dosages to counter the effects. Additionally, there are chal­
lenges regarding the identification of the best combination strategies
and the complications involved in the combination therapy [26].
Upon the selection of a reasonably designed combination, the pro­
cess of its early clinical development is complicated; thus, requiring
attention to detail [26]. Implementation of combination strategies is Fig. 1. A schematic diagram showing the evolution of cancer treatments. It
depicts better therapeutic results with the use of combination cancer strategies
influenced by several issues including toxicity, pharmacokinetic and
and that mathematical models play an integral role.
pharmacodynamic interactions, proper timing of resistance develop­
ment and the identification of robust biomarker in predicting the
response. Clearly, the questions of how to implement the combination, propose plausible combinations of treatment strategies to treat a variety
when to implement the combination and to whom it is implemented for of cancers.
are the major problems that face the clinical community during devel­
opment of treatment regimens [26]. 2. Mathematical models for combination cancer therapy
Production of new anticancer drugs takes a long time, is risky, and
requires costly medical experimentation; and thus, mathematical and 2.1. Models of chemoimmunotherapy
computational approaches are needed to supplement clinical trials in
making predictions. For this reason, efficient mathematical approaches Cancer chemotherapeutic drugs normally disrupt pathways neces­
are needed to try and understand different mechanisms of cancer ac­ sary for the growth and survival of tumours. However, they are often
tivities and effective combination therapy for cancer at a relatively limited due to the development of drug resis-tance and the damages
affordable cost. Many Mathematical models have been formulated to exerted to normal cells of the host tissue [27]. Recent years have focused
determine effective combination cancer therapies. Nonetheless, the cure on host-tumour interactions and it has been established that
for cancer is still far from reach, with complexities associated with host-tumour interactions play a pivotal role in determining clinical
therapeutic combinations remaining a major impediment. course and the outcomes of the treatments of human malignancies [27].
Here, we carry out a critical review of published mathematical Several mathematical models have been constructed in an attempt to
models that have been used to predict the outcome of combination understand tumour-immune interactions in depth and determine the
therapy strategies. The therapeutic strategies reviewed are chemo­ elements in the immune system that play a critical role in responding to
immunotherapy, chemovirothrapy, chemoradiotherapy, radio­ immunotherapy [28]. Several studies [29–35], have attempted to un­
immunotherapy, immunovirotherapy, radiovirotherapy and targeted derstand the effects of immune modulation by using mathematical
therapies (see Fig. 1: which shows the evolution of cancer treatments. approaches.
The figure further depicts that better therapeutic outcomes are achieved Modern cancer treatment methods rely on the ability of certain
when combination cancer strategies are used. It as well depicts that cancers to trigger the immune response. In- corporation of the immune
mathe-matical models shall play an integral role in deciding on strate­ component in the formulation of mathematical models has played a key
gies that are viable). Existing open problems are discussed, and we role in the clinically observed phenomena, for instance, tumour

2
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

dormancy, unchecked growth of tumours and oscillations in tumour size such viruses include vaccinia virus, measles virus, reovirus, adenovirus
[36]. The first attempt to illustrate the immunotherapy related effects in and vesicular stomatitis virus [45,46]. During carcinogenesis, tumour
a suitable ordinary differential equations (ODEs) model was given by cells are able to gain genetic and cellular changes that interferes with the
Kirschner and Panetta [37]. The study considered inter-leukin (IL-2) host immunosurveillance capabilities [23]. Such targeted,
along with adaptive cellular immunotherapy (ACI) by developing tumour-specific properties make oncolytic virotherapy, either solo or in
dynamical equations explaining external inflow of both IL-2 and im­ combination (e.g. chemovirotherapy), an attractive therapy for cancer.
mune cells. In recent years, chemovirotherapy has been shown to enhance
Chappell et al. [10] presented a high-level abstraction mathematical therapeutic effects that are far from reach when each therapy is used
model that explored the interaction of immune cells and tumour cells, independently [11,20,22,47–49]. Ottolio-Perry et al. [20] reviewed the
where they further explored the merits of combining immunothera­ combination of oncolytic viruses with the existing treatment modalities
peutic agents with chemotherapy and radiotherapy. The model was with a view of helping investigators make informed decisions regarding
tested, and numerical simulations were carried out using data similar to the clinical development of these products. In another study, Nguyen
that shown by Deng et al. [38]. Numerical results showed that tumour et al. [47] highlighted the mechanisms that guarantee the successful
mass reduced significantly when radiotherapy and immunotherapy are combination of oncolytic viruses with other therapeutic drugs where,
used in combination whereas single therapy recorded no significant however, he concluded that the success of these combinations is subject
decrease in tumour mass. Further, a combination of radiotherapy and to several factors including the type of oncolytic virus used, type of
immunotherapy leads to an increase in the number of T-cells that are cancer targeted, dosage and the timing.
activated compared to single therapies. There is a wealth of mathematical models that have been developed
Another mathematical model, comprising of ODEs, focussed on the in an attempt to comprehend as well as characterize viral dynamics
interplay between NK and CD8+ T cells with different varieties of [50–56]. Not many mathematical models have, nonetheless-thus far,
tumour cell lines [39]. The results found that ligand transduced cells been de-veloped and studied to investigate the combination of chemo­
trigger enough immune response to put tumour growth under control, therapeutic drugs and viruses. Malinzi et al. [11] constructed and
whereas control-transduced tumour cells evade immunity. In a more analyzed a model of parabolic non-linear partial differential equations
recent study, de Pillis et al. [28] extended their earlier study [39] by (PDEs) to examine the spatiotemporal dynamics of tumour cells under
incorporating the latest research on baseline NK and activated CD8+ the treatment of chemovirotherapy. The proposed model con-sisted
T-cells in both healthy donors and cancer patients. The model was compartments of uninfected and infected tumour cells densities, a free
quali-tatively analyzed and supplemented with numerical simulations of virus, and a chemotherapeutic drug. It was shown that the success of
chemoimmunotherapy and vaccine therapy. virotherapy is highly determined by the virus infection rate and burst
The results of the study intimated that, depending on the efficacy of size. Further, travelling wave solutions revealed that tumour diffusivity
the CD8+ T-cells, chemoimmunotherapy is likely to successfully control and growth rate are critical parame-ters during chemovirotherapy
tumour cells in body tissue. treatment and numerical simulations confirmed that combining
Rodrigues et al. [40] developed a simple ODE model to study the chemotherapeutic drugs with oncolytic viruses is more effective
dynamics of chemoimmunotherapy of chronic lymphocytic leukemia. compared to either monotherapies.
Their model analysis showed that the application of chemotherapy In an effort to investigate the enhancement of chemotherapy by
paves way for immunotherapy in a way that it decreases the number of oncolytic viruses, Malinzi et al. [12] pro-posed an ODE mathematical
cancer cells thus reducing the tumour density on which immune cells model and an optimal control problem for chemovirotherapy. The
acts upon. It was established that there is a minimum number of immune model described the interplays between tumour cells, immune response
cells needed to be transplanted in adoptive treatment so as to ensure a and a treatment combination by combining virotherapy and chemo­
complete cure or remission. Nonetheless, more studies are needed to therapy. Stability analysis showed that tumours could grow to their
model adoptive cellular immunotherapy. maximum capacity given that there is no form of treatment. It was
Pang et al. [41] discussed single chemotherapy, immunotherapy and shown that chemotherapy alone is likely to clear tumour cells on the
mixed treatments as well as conditions triggering tumour eradication. con-dition that the efficacy of the drug is more than the intrinsic growth
They analyzed the effects of least effective concentration and the rate of the tumour. The combined effects of oncolytic virotherapy and
half-life of the therapeutic drugs where they found that better results can chemotherapy were evaluated using sensitivity analysis of the model
be achieved if the half-life of the drug is ex-tended. Further, the impact parameters. Optimal control simulations revealed that the half of the
of drug resistance on therapeutic results was considered and a mathe­ maximum tolerated dozes (MTDs) for chemotherapy and virotherapy
matical model explaining the cause of chemotherapeutic failure that optimize treatment outcomes. Sensitivity analysis and numerical simu­
uses a single drug was proposed. In the end, numerical simulations lations affirmed that the success of chemovirotherapy depends on virus
showed that chemoimmunotherapy is likely to achieve a better treat­ burst size, the rate of viral infection and the dose. Moreover, the right
ment effect. However, de-spite the reported achievement, tumour cells dose of chemotherapy required to produce effective results in unison
are continuously becoming resistant to numerous structurally and with virotherapy has been a concern for both clinicians and
mechanistically unrelated drugs, limiting the effectiveness of chemo­ mathematicians.
immunotherapy. Thus, determining how to effectively amalgamate Malinzi [22] proposed a mathematical model for chemovirotherapy
those treatment modes and design optimal dosage combination regi­ where he considered three-drug infusion methods and compared their
mens deserves further research. efficacies, carried out mathematical analysis to forecast the outcomes of
the OVs in combination with chemotherapy and also compared the ef­
2.2. Models of chemovirotherapy ficacy of each single treatment modality. The model was formulated
based on two aspects, that is, model without delay and with delay.
The use of viruses as targeted therapy in cancer originated in the Numerical simulations for both models were carried out, where it was
early 20th century with numerous viruses already tested in humans and shown that if the tumour burst is big, then regardless of the drug infusion
experimental animals [42]. Oncolytic viruses (OVs) are a heterogeneous method, chemovirotherapy is more effective than any of the single
group of low or non-pathogenic viruses employed in the treatment of treatments. Simulations further indicated that the success of chemo­
cancers, taking advantage of their ability to selectively infect and lyse virotherapy depends on virus burst size, the rate of viral infection and
cancer cells without damaging normal cells [43,44]. Many viruses, both the dose. Further, simulations showed that incorporating delays in the
genetically engineered and naturally occurring, are currently being system increased the time within which tumour clearance occurred in
researched as oncolytic cancer virotherapeutic agents [43]. Examples of body tissue. Despite this study, there still exists open questions which

3
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

need further investigation, for example, the model does not address the result if a drug exhibits both radiosensitization and independent cyto­
quantity of dose required to completely eradicate the tumour and at toxic properties [61].
what specific point in the tumour should the drug be infused to have a Ghaffari et al. [62] proposed an ODE model that considered
greater effect. Interestingly, ascertaining the optimal dosing schedule is chemotherapy and radiotherapy for metastatic cancer. They examined
still a challenge. the interplay between immune and cancer cells, with chemotherapy
regarded as a predator on both normal and cancer cells. The model
2.3. Models of chemoradiotherapy aimed at understanding the specific system dynamics and as well guide
the creation of combination therapies. It was shown that a decaying
The paradigm of the interplay between radiation and chemotherapy dosage protocol is weaker compared to a constant dosage protocol in
was first conceptualized by Steel and Peckham in 1979 [57] and was eliminating small metastases at a given time.
later summarized by Seiwert et al. [58]. Two of the main advantages for Marcu et al. [63] simulated ciplatin-radiotherapy treatment with
the use of chemoradiotherapy, assuming there is no interaction between emphasis on time sequencing and combined treatment scheduling of the
the two, is to use cytotoxic drugs that will be used to tackle the disease, drug and radiation. They examined different time courses of cisplatin
not within the radiation field and to irradiate seclusion sites [57]. and radio-therapy for advanced head and neck cancer. It was agreed that
In a study by Goldie and Coldman [13], a stochastic model for to discover the true synergistic effect between cisplatin and radio­
alternating radiation and chemotherapy was proposed. Their model was therapy reported in the literature, they would have to incorporate a
based on the earliest approaches for combination therapy where they proper radiosensitizing term. They further indicated that by adminis­
used three compartments, that is, stem, differentiation and end cells in tering cisplatin on a weekly basis would result to enhanced ther-apeutic
modeling tumour growth. Their model incorporated chemo-resistant, effects compared to daily doses and that giving cisplatin in cross prox­
radio-resistant and extra parameters to measure cells with joint resis­ imity in time to the radiation fractions would definitely result to
tance. They found that a regimen treatment with three doses of maximal combined effect.
chemotherapy and radiotherapy acting in an alternating manner is Barazzuol et al. [64] formulated a mathematical model of radio­
pow-erful compared to sequential treatment under the same conditions therapy in the treatment of glioblastoma where they used a linear
because it suppresses subpopulation resistant responsible for treatment quadratic model and investigated the additional effect of temozolomide
failure. in two simpli-fied scenarios. Their main aim was to address the question
In another study by Beil and Wein [3], a mathematical model was of whether the benefit of temozolomide is drawn from its radio­
formulated with the aim of determin-ing the best way to sequence the sensitizing properties in the concurrent phase or from the additional cell
three standard therapies for cancer, that is, radiotherapy, chemotherapy kill of the adjuvant phase. They concluded that the model presuming
and surgery. Differential equations were used to model the growth of only radiosensitization fits the observed survival curves more closely
tumours and its metastases with an underlying assumption that primary than assuming independent cytotoxicity.
and metastatic tumours behave identically. The model aimed to show Powathil et al. [65] presented a multiscale mathematical model
which combina-tion achieved higher curative probability: surgery fol­ which relied on cellular automata to examine the spatial distribution
lowed by chemotherapy and radiotherapy (SCR), Surgery followed by and microenvironment of a cell population during treatment. A log
radiotherapy and chemotherapy (SRC) and surgery followed by radio­ cell-kill model was used to estimate chemotherapeutic effects whereas
therapy then chemotherapy and radiotherapy (SRCR) given that the the linear-quadratic model was used in the case of radiation effects. They
primary tumour is big enough or that the metastatic cancer population applied their model to show a clinical application to two known and one
outnumbers the primary tumour. Their model included metastatic can­ hypothetical treatment regimen for esophageal cancer, where they
cer originating from dormant cells and angiogenesis, which eventually concluded that their proposed alternative regimen was more effective.
led to the interactions between primary tumour compartment and Several mathematical models allow broad disruptions regarding the
metastatic compartment and thus the emergence of optimality concepts of pure radio-sensitization and independent chemotherapy cell
sequences. kill [66–74].
Ergun et al. [59] used a similar approach to find optimal scheduling
and doses of angiogenic inhibitors and radiotherapy that maximize the 2.4. Models of radioimmunotherapy
elimination of a primary tumour. The model was analyzed using two
compartments of tumour cells and vascular endothelial cells and the Successful implementation of radioimmunotherapy for cancer treat-
damage by radiation was modelled in accordance with the ment has proven to be significantly more difficult than was expected
linear-quadratic which widely determines fractionation schedules in initially [75–79]. This is, in part, as a result of highly diverse, complex
radiobiology society. It was shown that optimal amalgamation treat­ and interrelated biological and physical factors that are considered
ment regimen happens when antiangiogenetic therapy is constantly when designing a successful protocol [75]. By utilizing mathematical
increased in order to maintain an optimal tumour endothelial cells ratio, models incorporating plasma antibody pharmacokinetics, extravasation
and the fraction sizes of radiation keep changing with treatment so as to and interstitial transport and the antibody-antigen interaction, Fujimori
maximize the probability of tumour control. However, the model was et al. and van Osdol et al. [80–83] investigated the connection between a
solely concerned with the primary tumour and did not include the variety of tumour and antibody specific parameters and the microscopic
antimetastatic effect of antiangiogenic treatment, which might inhibit distribution of antibody and absorbed dose within a tumour [75].
metastatic growth during radiotherapy if it is applied methodically. O’Donoghue et al. [14] proposed a Uniform tumour Dosimetry
Further, the model did not take into consideration the fact that radiation mathematical model that compared single-dose and fractionated radi­
may cause vascular endothelial growth factor (VEGF) expression, which oimmunotherapy. The model compared large single administrations
attenuates the destruction of endothelial cells by radiotherapy. (LSAs) with rapid fractionation (RF) administered in small quantities
In a study by Salari et al. [60] it was shown that the addition of within a short period of the time interval. It was indicated that for ho­
chemotherapy drug and its mode of action makes optimal fractionation mogeneous absorbed dose distributions throughout tumours, LSAs are
regimen change for concurrent chemoradiotherapy. They contend that predicted to give a greater possibility of tumour cure than rapidly
the introduc-tion of chemotherapeutic drugs containing cytotoxic effects fractionated treatments of the same marrow toxicity. If dose distribu­
only does not hamper optimal fractionation regimen when there is no tions are heterogeneous, RF may have a therapeutic advantage,
concurrent chemotherapy whereas a radiosensitization agent has the depending on how tumour uptake varies from one fraction to another.
capacity to tamper with the optimality choice of fraction size. They also Kumar [84] proposed a mathematical model for radio­
show that the emergence of optimal non-uniform fractionation may immunotherapy where they formulated radiotherapy dose distributions

4
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

with regard to optimizing tumour cure probability (TCP) where the rate maximum tumour volume to be reduced under the restriction of a
of dose distribution was assumed to be high enough to enable the de­ maximum tolerated dose. Results showed that combining GVAX and
livery of dose distribution instantaneously. Numerical results showed anti-PD-1 in suitable quantities could consequentially reduce the growth
that the tumour cells density and dose distribution are not sensitive to of a tumour. More studies need to be carried out to explore the effects of
other various functional forms of tumour parameters. The results of this treatment with GM–CFC–secreting vaccine (GVAX) and anti-PD- 1 drug.
study revealed that the immune response plays a critical role during To date, oncolytic virotherapy still possess an exercise in population
cancer treatment. dynamics where the interplays between the viruses, tumour cells and the
Flux et al. [85] described a dosimetry 3-D mathematical model that immune system components play a major role in determining treatment
quantified the absorption of dose dis-tribution as a result of adminis­ out-comes [17,50,94,95,100–105]. Several mathematical models have
tration of an intralesional radiolabeled monoclonal antibody which been formulated to describe the outcome of such interactions [17,95,
allowed the distribution of spatial and temporal heterogeneity of 100–105]. A study that addresses the combined effects of viral oncolysis
radionuclide without a calibration scan. The model was tested by using a and T-cell- mediated oncolysis was carried out in Ref. [15] wherein a
set of registered patient data where dose profiles and histograms of the mathematical model of virotherapy that induces release of cytokine
dose volume were produced. It was established that 3-D dose distribu­ IL-12 and co-stimulatory molecule 4-1BB ligand was developed. It was
tion was significantly non-uniform. Initially, intralesional infusion re­ found that whereas viral oncolysis is important in reducing the tumour
sults suggested that the model offered a means of determining how the burden, increased stimulation of cytotoxic T cells leads to a short-term
absorbed dose was distributed within a tumour. reduction in tumour size, but a faster relapse. In addition, it was
Serre et al. [86] suggested a discrete-time pharmacodynamic math­ found that amalgamations of specialist viruses expressing either IL-12 or
ematical model of the amalgamation of radiotherapy and immune 4-1BBL might initially act more potently against tumours than a general
checkpoint inhibitors: PD1-PDL1 axis and the CTLA4. The model shows virus that simultaneously expresses both, but the advantage is likely not
how a growing tumour triggers and inhibits tumour-immune responses large enough to replace treatment using the generalist virus.
and describes the effects of irradiation. The model’s ability to predict A mathematical model incorporating cytokine and co-stimulatory
pharmacodynamic endpoints was justified in retrospective by exam­ molecule expressing OVs in combination with DC injections was stud­
ining that it could explain clearly data obtained from experimental ied by Wares et al. [91]. The aim of this study was to investigate the
studies, which took into consideration the combination of radiotherapy effect of varying doses of OV and DC injections during tumour treat­
and immune checkpoint inhibitors. They concluded that different de­ ment. It was shown using simulations that treatment of a tumour with
signs in silico could be com-pared by simulating Kaplan-Meier curves immunostimulatory OVs first followed by a sequence of DC injections is
and mathematical tools could be used to partially automate optimized more effective than alternating OV and DC injections. It was concluded
protocols. that the efficacy of a dosing strategy depends on the ordering of onco­
More work is needed to focus on integrating the developed mathe­ lytic adenoviruses and dendritic cells, temporal tumour spacing and
matical methods into clinical practice that will heavily deal with man­ dosages chosen. This model was later extended by Gevertz and Wares
aging drug and radiation-induced toxicities during the process of [106] in which used different techniques such as information criteria
optimization. Fur-ther combining radioimmunotherapy with other analyses, sensitivity analyses and a parameter sloppiness analysis and
treatment regimens including targeted agents and metronomic chemo­ discovered that it is possible to reduce their model to one variable and
therapy is a subject to be explored in future research. More still, dose three parameters and still be fitted to the data. They argued that
effect relationships should be exam-ined and a pharmacokinetic model reduction of the model to minimal form allows for tractability of the
integrated to clearly shade light on the relationship between the system in the face of parametric uncertainty.
experimental results and the actual dosing protocols. Timalsina et al. [107] proposed a PDE model with the aim of
studying tumour virotherapy and mediated immunity. The model
2.5. Models of immunovirotherapy incorporated adaptive and innate immunity responses with interactions
in tumour cells, OVs and immune system in a territory containing a
Viruses have been engineered genetically to promote the expression boundary in motion. They assumed that all tumour cells exhibit homo­
of pro-inflammatory cytokines and co-stimulatory molecules which, geneity and viruses follow a diffusion process. They used the results
upon release, instigates a targeted immune response on the tumour obtained to examine tumour development and provide insight into
[87–91]. Several treatment approaches combining viral oncolysis with crucial aspects of virotherapy such as dependence of the efficacy on vital
production of immunostimulatory molecules and dendritic cell (DC) parameters and the delay in the adaptive immunity.
injections are currently under construction with the aim of bettering Jenner et al. [108] suggested a mathematical model with a view of
treatment outcomes [91–93]. Several studies have been carried out to reproducing the results for tumour growth synonymous to results pro­
investigate the outcome of immunovirotherapy on myeloma [94], to duced experimentally during treatment with an adenovirus virus. The
establish how initial conditions hamper the efficacy of OV therapy on model was fitted to data using parameters from the literature in order to
melanoma [95] and determine factors inhibiting and enhancing the reduce the degrees of freedom of the model. Optimization of the model
transmission of oncolytic viruses via a tumour site [96]. Other recent was done using a least-squares non-linear fitting algorithm ‘lsqnonlin’ in
studies focusing on the interaction of immune cells with DC vaccines MATLAB. The model indicates that decreasing APC stimulation and
include [97–99]. raising helper T cell stimulation is likely to improve treatment. None­
Lai and Friedman [16] considered a combination therapy where one theless, it is still imperative to establish the range of these parameters
drug was used as a vaccine to activate dendritic cells so as to instigate when the absolute elimination of tumour cells occur.
more T cells for the purpose of invading the tumour while the other drug Mahasa et al. [109] presented a mathematical model that elucidated
is a checkpoint inhibitor which suppresses cancer cells. They developed the interplays between oncolytic viruses, tumour cells, normal cells and
a mathematical model in the form of PDEs to address the question of the antitumoural and antiviral immune responses. The model consisted
whether combining treatment of two drugs administered at certain of delay differential equations (DDEs) with one discrete delay indicating
levels is better than using a treatment of one drug with almost twice the the time required to trigger a tumour-specific immune response. The
dosage level. The model describes the interaction of dendritic cells, model aimed to predict effects of antitumoural and antiviral immune
cancer cells, cytokines IL-I2 and IL-2, CD8+ cells (T8), GM–CFC–secret­ responses in the presence of oncolytic viruses, the response of the
ing cancer cells (GVAX) and anti-PD-1. The concept of synergy between tumour to the oncolytic viral infections and the oncolytic virus
drugs was introduced and a synergy map was developed suggesting the tumour-specificity responsible for maximizing the reduction of the
proportion in which drugs should be administered so as to realize the tumour while concurrently ensuring that the toxicity on the normal

5
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

tissue encircling tumour cells are minimized. targeted therapies together with other treatment methods. For example,
Cassidy and Humphries [110] formulated and analyzed a mathe­ with immunotherapy; a great number of preclinical and clinical trials
matical model of tumour growth under on-colytic virotherapy and have revealed that there is a synergistic antitumor effect with the use of
immunotherapy. The model explicitly included heterogeneity in tumour targeted therapy with immunotherapy [116].
propagation speed by considering the cell cycle duration as a random The development or improvement of these targeted therapies re­
variable. The interrelationship between the estimated number of cells quires a molecular understanding of cancer pathogenesis and a char­
surviving in the cell cycle and tumour eradication was established by the acterization of the interplay between tumour cells and therapeutic
help of linear stability analysis. The results of their analysis showed that agents. Nonetheless, not many attempts have been made to mathemat­
an increase in immune recruitment acts synergistically to lyse tumour ically investigate the dynamic relationships between cancerous cells and
cells during oncolytic virotherapy. Nonetheless, they noted that their targeted therapeutic agents; moreover only a few mathematical studies
model was limited by the fact that it oversimplified tumour-immune exist, thus far, for the prediction of the outcome of combination treat­
interplays. ment with targeted therapies. A comprehensive review of mathematical
models for targeted therapy can be found in Abbott and Michor [114].
2.6. Models of radiovirotherapy We complement these with a review of some mathematical studies on
targeted therapies and combination treatments involving targeted
The interaction dynamics of oncolytic viruses with tumour cells and therapies.
im-mune responses are highly intricate [17,52,53,102,111]. Deter­ Green et al. [117] proposed a compartmental mathematical model
mining the outcome of radiovirotherapy is de-pendent on understanding for antibody-targeted therapy of colorectal cancer, using clinical trial
the complex interactions between the various components involved data from bio-distribution of antibodies against carcinoembryonic an­
during treatment with radiovirotherapy [17]. Thus, modeling the ki­ tigen. The study indicated the most significant parameters that deter­
netics of virotherapy could act as an impetus for the devel-opment of mine antibody localization are the affinity for the antibody, the flow of
improved therapeutic protocols as well as gaining further insight on the the antibody through the tumour and the rate of elimination of the
outcome of such therapies [17,52,53,102,111]. antibody from the tumour.
A study by Dingli et al. [17] focused on attenuated strains of measles Shen et al. [118] developed a mathematical biphasic model that
virus that highly infect tumour cells because of their high expression of accurately described the cell-drug response based on an analysis of
CD46, a property that gives viruses an advantage to attach and enter into targeted inhibition of colorectal cancer cell lines. Their model used three
the target cells. They proposed and analyzed a mathematical model that kinetic param-eters: ratio of target-specific inhibition, F1, potency of
took into account population dynamics that encapsulated crucial com­ target-specific inhibition, Kd1, and potency of off-target toxicity, Kd2 to
ponents of radiovirotherapy. Analysis of the model included deter­ describe the drug response where the determination of these kinetic
mining and establishing existence of corresponding equilibria related to parameters are valuable in predicting effective combination targeted
complete cure, partial cure, and treatment failure. Through numerical therapy for multi-driver cancer cells. The model and the mechanistic
simulations, they analyzed the influence of factors, in the form of pa­ insights give a new mechanistic perspective and mathematical tool for
rameters, that determine the outcome of radiovirotherapy treatment. predicting effective combination targeted blockades against
Morestill, they evaluated relevant therapeutic scenarios for effective multi-driver cancer cells.
radiovirotherapy. The study showed that radiotherapy is more effective Stochastic differential equations (SDEs) have been employed to
when used synergistically with oncolytic virotherapy. model the stochastic evolution of resistance of therapeutic drugs. Sun
Tao and Guo [112] developed a PDE model describing cancer radi­ et al. [119] proposed a stochastic model by using a set of SDEs to
ovirotherapy, which is a generalization of the existing ODE models given examine the dynamics of drug sensitive cells, drug-resistant cells and
in Refs. [17,51]. The model was developed as a moving boundary new metastatic cells. They used clinical data to vali-date their model and
problem. Global existence and uniqueness of solutions to the model was predicted distinct patterns of drug dose-dependent synergy for two
proved, and a new explicit parameter condition corre-sponding to different sets of drug combinations in the treatment of melanoma. This
treatment success was determined. By modeling combined action of approach was expected to enhance the study of effective and robust
virotherapy and radiotherapy, they aimed to design the possible optimal cancer drugs.
therapy strategies. Numerical experiments verified that Kozlowska et al. [120] presented a comprehensive stochastic math­
radiovirother-apy is more effective compared to monotherapy. To ematical model and simulator approach that describes platinum resis­
explore possible optimal therapy strategies, a number of numerical tance and standard of care therapy in high-grade serous ovarian cancer
simulations were further carried out. The results suggested that there is (HGSOC). Their study used post treatment clinical data, including
an optimal timing of radio-iodine administration and an optimal dose of 18F-FDG-PET/CT images, to accurately predict the model parameters
the radioactive iodide, which needs to be tested by experimental data. and simulate ‘virtual patients with HGSOC’. The model revealed that the
treatment responses exhibited by these virtual patients was in line with
2.7. Mathematical models of targeted therapies those of real-life patients with HGSOC. They utilized their approach to
evaluate survival benefits of combination therapies containing up to six
Targeted therapies involve the use of novel cancer drugs that interact drugs targeting platinum resistance mechanisms where they indicated
with specific molecular targets that are pertinent to cancer progression that combining standard of care with a drug specifically targeting the
[113,114]. Examples of targeted therapies include; protein-kinase in­ most dominant resistance sub-population resulted in a significant sur­
hibitors: Imatinib (Glivec®)-used for the treatment of leukemia, Dasa­ vival benefit.
tanib (Sprycel®)-used for treating prostate cancer, and Temsirolimus Jarrett et al. [121] constructed a mathematical model that evaluated
(Torisel®)-used for treating several cancer types, monoclonal anti­ the combination of trastuzumab-paclitaxel therapies for drug interaction
bodies: Bevacizumab (Avastin ®)-for treating colon cancer, and pro­ relations on the basis of order, timing and quantity of the drug dosage.
teasome in-hibitors: Bortezomib (Valcade®)-for treating myeloma and Their model was based on time-resolved microscopy data that captured
leukemia [114]. Dozens of other targeted therapies for treating different changes in vitro cell confluence in response to the combination of
types of cancer do exist and have already been approved for use whereas paclitaxel and trastazumub treatment. The model showed increased
others are under-going clinical trials [115]. Single targeted therapy synergy for treatment reg-imens where trastuzumab was administered
drugs with short-lived treatment effects often fail [116]. The alternative before paclitaxel and indicated that trastuzumab accelerates the cyto­
strategy previously proposed is to synergistically use a combination of toxicity of paclitaxel.
targeted therapy drugs which employ different pathways, or to use Bozic et al. [115] proposed a mathematical model to predict the

6
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

effect of combining targeted therapies. Their model was based on data study examined dynamic effects of the drug STI-571 (Glivec) on the
obtained from 20 melanoma patients. Their model simulation results autophosphorylation of the BCR-ABL oncoprotein and subsequent
revealed that combination treatment with two targeted therapy drugs is signaling through the Crkl pathway, and they used simulations to pre­
far more effective than using a single drug; and those drugs should dict a minimal concentration for drug effectiveness. The model was
simultaneously be administered for optimal results. fitted to mouse Bcr-Abl autophosphorylation data where it is revealed
Owen et al. [122] constructed a spatiotemporal mathematical model that cellular drug clearance mechanisms such as drug efflux significantly
to determine the outcome of combining macrophage-based hypo­ reduces the effectiveness of Glivec in blast crisis cells. Additionally, it is
xia-targeted gene therapy with chemotherapy. Model simulation results stipulated that these resistance mechanisms might be present from the
showed that combining conventional drugs and macrophage based tar­ onset of disease.
geted therapies would work synergistically and result to better Other mathematical models of combination cancer therapy include
anti-cancer effects than the individual effects from each of the therapies. [75,104,128–160]. Bajzer et al. [104] modelled cancer virotherapy with
Arciero et al. [33] presented a mathematical model where they recombinant measles viruses where the interactions of the tumour and
considered a novel treatment strategy known as small interfering RNA virus were based on the already established biology. Hadjiandreou and
(siRNA) therapy in which the treatment suppresses TGF-β production by Mitsis [158] developed a mathematical model that used Gompertz
targeting the mRNA codes for TGF-β, and thus reducing the presence and growth law and pharmacokinetic-pharmacodynamic approach to model
effect of TGF-β in tumour cells. The model predicts conditions within the effects of drugs on tumour progression and designed optimal ther­
which siRNA treatment can be successful in transformation of TGF-β apeutic patterns. Su et al. [159] formulated a mathematical model of
producing tumours to either non-producing or producing a small value tumour therapy with oncolytic viruses and MEK inhibitor.
of TGF-β tumours.
Tang et al. [123] introduced a novel model, called TIMMA (Target 3. Discussion and conclusion
Inhibition inference using Maximization and Minimization Averaging)
with the aim of demonstrating its feasibility in systematic investigation Combination cancer therapy strategies have fast become the estab­
of the model predictions using kinome-wide single and pairwise siRNA lished method for oncologists to treat cancer and has the potential to
knock-down experiments. The model used functional data on drugs for counter treatment side effects and resistance to drugs. There is much
its construction and target combination predictions. The validation of concern over the toxicity and effectiveness of combining therapeutic
TIMMA re-sults using systematic siRNA-mediated silencing of the strategies. Ideally, there are several factors that necessitate the selection
selected targets and their pairwise combinations showed an increased of a certain cancer combination therapy strategy such as im-provement
ability to identify both druggable kinase targets and synergistic in­ in the quality of life of the patient, nonoverlapping safety profiles,
teractions. The model was ap-plied to case studies in MCF-7 and reduction of drug resistance and preclinical evidence of synergy be­
MDA-MB-231 breast cancer and BxPC-3 pancreatic cancer cells where it tween the interacting therapeutic agent. However, developing novel
was confirmed that TIMMA-predicted kinase targets are essential for combi-nations of therapeutic strategies is time-consuming and expen­
tumour survival, either individually or in combination. Further, it was sive and depends on a myriad of factors unique to each cancer. More­
shown that the construction of model algorithm resulted in significantly over, there are still several questions that need to be answered
better predictive accuracy and computational efficiency in comparison pertaining to combination cancer therapy. These include determining
with an existing algorithmic solution. the optimal dosage combinations and investigating the side effects of
In another study, Tang et al. [124] applied network pharmacology using the different combination treatments. Mathematics can help to
model to predict synergistic drug combi-nations. Kinome-wide answer such questions by allowing researchers to model therapeutic
drug-target profiles and gene expression data were used to pinpoint a options (including new, repurposed and old drugs) and predict their
synergistic target interaction between Aurora B and ZAK kinase inhibi­ treatment and toxicity outcomes before embarking on lengthy and
tion that resulted to an enhanced growth inhibition and cytotoxicity. expensive trials. Scientists use a range of methods, tools and software to
They employed stochastic simulation algorithm (SSA) to model analyze mathematical models; the choice depends on the problem being
signaling pathways, implement and understand the mechanisms of ac­ solved and the model type. Mathematical models in the form of differ­
tion of the identified target interactions. An MDA-MB-231 signaling ential equations are normally analyzed using qualita-tive methods; to
network was constructed using dynamic modeling approach to simulate investigate the long-term characteristics of the model solutions
the effect of perturbing the genes of interests on the cell viability. The [161–163]. Model analysis is usually complemented with numerical and
observed similarity of the model predictions to the experimental ob­ computational simulations using several methods implemented by a
servations indicates that the constructed signaling network consists of number of programing languages and software [164,165]. Some ex­
key protein-protein interactions that may mediate the synergistic and amples of tools and software that are used in analyzing mathematical
antagonistic relationship of Aurora B with ZAK and CSF1R, respectively. models reviewed in this paper and tools that can be used for users to
Araujo et al. [125] presented a mathematical model to investigate upload and analyze input data are summarized in Table 2. We as well
combination therapy in which multiple nodes in a signaling cascade provide some existing databases for combinations of cancer therapy in
pathway are simultaneously targeted with specific inhibitors. The model Table 3.
sug-gested that the attenuation of biochemical signals is significantly Complete drug development takes more than 10 years with huge
enhanced when several upstream processes are inhibited, and that this costs ranging from USD 12 million to USD 12 billion. Moreover, only a
weakening is most pronounced in signals downstream of serially con­ small fraction of drugs that enter clinical trials are eventually approved
nected targets. [166]. This makes it imperative to devise methods of reducing the cost of
Komarova and Dominik [126] developed a mathematical framework drug development and one such way is through mathematical modeling.
to examine the principles underlying the emergence and prevention of Drug developers, researchers, clinicians and other bio-scientists can use
resistance in the treatment of cancers with targeted drugs. They model simulations and results from mathematical analyses to assimilate
considered a stochastic dynamical system which took into account complex mechanisms of cancer therapy dynamics thus leading the
turnover rate of tumour cells and the rate of generating resistant mu­ development of more effective treatments.
tants.The model was applied chronic myeloid leukemia where it was In Section 2, we reviewed mathematical models for a number of
suggested that combining three targeted drugs with different specific­ combination cancer therapy strategies de-veloped thus far. In doing so,
ities is likely to overcome the problem of resistance. we highlighted the important discoveries that have been made via
Charusanti et al. [127] presented a mathematical model describing mathematical modeling and identified the gaps that still need to be fil­
several signaling events in chronic myeloid leukemia (CML) cells. Their led. All the mathematical studies, reviewed here, indi-cate that

7
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

Table 1
A summary of merits and demerits associated with the use of certain forms of combination therapies.
Therapies Merits Demerits References

Chemoimmunotherapy Application of low-dose chemotherapy with immune- stimulating Although chemoim- munotherapy in animal models is well [10,38,61,
vaccination significantly lengthens sur- vival both in a prophy­ established, it is extremely hard to transfer this knowledge into 172–177]
lactic and therapeutic setting. clinical practice as there are sev- eral hurdles that must be
overcome.
Improved progression-free survival and the overall survival of Side effects include sec- ondary malignancies, myelo- and
physically fit patients with previously un- treated symptomatic immunosuppression which are likely to be the onset of myeloid
chronic lymphocytic leukemia. malig- nancies (acute myeloid leukemia and myelodysplas- tic
syndrome), hematological non-hematological toxicities.

Chemovirotherapy Several studies have shown that combining chemother- apy with Although this combination has proved promising, it is however [11,12,20,22,
oncolytic viruses re- sults to enhanced therapeutic effects. limited by the side effects such as viremia and cardiac toxicity. 167,178–181]
Benefits include significant improvement of overall sur- vival,
enhanced efficacy and decreased tumour volume.

Chemoradiotherapy Cisplatin chemoradiotherapy (CRT) delays disease recur- rence Moderate to acute gastroin- testinal tract effects in the form of [3,13,20,57,
and boosts survival in contrast to radiotherapy (RT) alone in bleeding proctitis due to telangiectasia and ulcera- tion, which 182–185]
women with stage IIIB squamous cell carci- noma of the cervix. has been poorly reported in the literature.
CCRT has a survival advan- tage compared with RT alone in Increased incidences of tu- mour recurrence, gastroin- testinal
patients between 65 and 72 years. and severe hemato- logic complications.
Enhances local tumour con- trol in the treatment of cervi- cal Has low overall survival rate and late toxicity
cancer.
Concurrent chemoradiother- apy has been shown to reduce the
risk of cancer recurring by 50% in patients having ad- vanced stage
disease regional spread.

Radioimmunotherapy Radioimmunotherapy treat- ment regimen has been used in the Limited by hematologic toxicity, secondary cancers and of [14,168,186,
treatment of haemato- logical malignancies with a fractionated myelodysplastic syndrome. 187]
dosing regimen achieving remarkable regres- sion of bulky masses
with non-Hodgkin lymphoma.
Studies on other aggressive non-Hodgkin lymphoma have indi­ There is considerable ev- idence indicating that the overall risk
cated promising efficacy of anti-CD20 ra- dioimmunotherapy and for Radioim- munotherapy is no higher than chemotherapy.
anti-CD22 radioimmunother- apy reduced hematologic toxicity,
improved survival benefits and improved efficacy.
Other demerits include the development of drug resis- tance as
well as being costly.
Convincing of the oncohe- matologist community to in- corporate
radioimmunother- apy would require designing of randomized
clinical tri- als and stratification of pa- tients for response to ra-
dioimmunotherapy. Clinical efficacy of radioimmunother- apy
applications in solid tu- mours remain limited.

Immunovirotherapy Selective virus replication in rapidly proliferating cells. Genetic mutations allow for the development of resis- tance of [87–91,106,
tumour cells to OVs. Moreover tumour cells can learn to avoid 108]
adaptive immu- nity via immunoediting.
Directs tumour microenvi- ronment towards immune re- sponse. Viruses can trigger anti-viral immune responses.
Enhances survival.
OVs produces pathogens that are responsible for instigating as well
as enhanc- ing anti-tumour immunity. Combining OVs with im-
munostimulatory cytokines initiates an additional anti- tumour­
immune response with apoptosis of tumour cells occurring due to
suppressive properties of immune cells.

Radiovirotherapy In the treatment of prostate cancer, it is reported that MV- NIS Side effects including gas- trointestinal toxicity and weight loss. [17,20,
results to significant tu- mour regression as well as highly 179–181,
significant prolonga- tion of survival in animals. 188–194]
In addition, administration of iodine-131 enhances the anti- The efficacy of radiovirother- apy can be limited by sev- eral
tumour effect of MV-NIS vi- rotherapy. factors in thyroid tumour treatment.
Several HSV1 mutants act synergistically with radiation therapy The biodistribution of the virus, its propagation in the tumour
83–85, although in some experiments the impact of radiation is through numerous replication cycles and the strength of the
essentially ad- ditive sodium/iodide symporter expression can be inadequate or
hetero- geneous and thus it may affect an effective therapeutic
response.

Targeted therapies Foundation of precision medicine. Side effects include gastroin- testinal toxicity and weight loss. [113,114,116,
125]
Drug combinations targeting different pathways can be used. Cancer cells can eventually become resistant to targeted
therapies.
Attenuation of biochemical signals is highly enhanced. Imatinib Drugs for some targets are hard to develop.
(Glivec®), the first targeted therapy to be ap- proved by the FDA,
has proven to be successful in the treatment of chronic myeloid
leukemia.

combining cancer treatments strategies leads to enhanced therapeutic reducing the tumour volume. This ap-proach leads to an increase in the
outcomes. Mathematical models of chemoimmunotherapy showed that number of T-cells that are activated when chemotherapy and immuno­
with the use of this strategy for the treatment of chronic lymphocytic therapy are combined compared to a single therapy such as chemo­
leukemia, the patient’s immune system would be boosted in addition to therapy [10]. Further, ligand transduced cells can trigger enough

8
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

Table 2 Table 2 (continued )


Examples of existing software for implementing mathematical models reviewed Software Packages Functionality References
in this paper.
to automatically quan-
Software Packages Functionality References tify phenomena such as
Matlab Contains several Popular system for data [195] synergism and
differential equa- tion visual- ization and inhibition.
solvers, for example, carrying out numeri- TIMMA-R R packages. Predicts the effects of [208]
ode 23 and pdepe and cal simulations of drug combi- nations
tool boxes, for ex- differential equa- tions. based on their binary
ample, jLab; used for drug- target
data analysis interactions and single-
Maple Differential equation Powerful software for [196] drug sensitivity profiles
packages like dsolve and symbolic computation in a given cancer
PDEtools. and qualitative analy- sample.
sis. Synergyfinder Web application. Allows for pre- [209]
Mathematica Contains functions such Powerful symbolic [197] processing, analyz- ing
as Dsolve and NDsolve computation; offers and visualising pairwise
numerical evaluation, drug combinations in
opti- mization and an interactive man- ner.
visualization of a very Combenefit Enables for the analysis, [210]
wide range of quantifica- tion and
numerical functions. visualization of drug
Python Contains packages like Open source software [198] com- bination effects in
Scilab; a numerical and a high level, terms of synergy and/or
computational package numerically oriented antagonism.
that can be used for pro- graming language. URDME Simulates stochastic [211]
handling ODEs and reaction- diffusion
PDEs. processes on arbitrary
SageMath Rich in several packages Open source software [199] meshes.
such as de- solve for covering nu- merous COMSOL Solves highly nonlinear [212]
solving differential areas including differential equations.
equa- tions. differential calculus,
algebra, numerical
analy- sis, calculus and immune response to put tumour growth under control, whereas
statistics. control-transduced tumour cells evade immunity [28]. However, none
Maxima desolve function for Used for symbolic [200] of these models can yet provide an optimal dosage for the combination
solving ordi- nary computation and
differential equations. solving differential
of chemotherapy and immunotherapy.
equations. Chemovirotherapy has experienced notable successes both clinically
GNU Octave ODE solver; LSODE. Powerful open source [201] and experimentally, however, much is still unknown about it [12]. The
software with built-in principal mechanisms underlying the anti-tumoural effects of many or­
2D/3D plotting and
dinary virotherapies in conjunction with chemotherapeutics still re­
visual- ization tools.
Mathcad Contains functions like Contains equation- [202] mains a mystery. Fundamental questions such as prospective
odesolve and rkfixed. solving pack- ages ramifications of chemotherapeutic agents on viral replication and the
which can be used to immunotherapeutic effects caused by virotherapy are yet to be fully
plot func- tions, solve answered. Another troubling problem is that of fully distinguishing
blocks of equations,
perform curve-fitting
chemotherapy-related toxicity from that of virotherapy induced toxicity
operations as well as [167]. Designing optimal scheduling of chemovirotherapy treatment
solving differential attracts questions in an attempt to decipher the right dose combination,
equa- tions using deciding the most effectual method of drug infusion and the critical
iterative methods.
treatment characteristics [12]. Furthermore, fig-uring out the potential
Julia Contains solvers like Open source software [203]
OrdinaryDif-fEq.jl and designed for high implications of chemotherapeutics on viral reproduction and/or the
LSODA.jl. performance immunotherapeutic effects possibly caused by virotherapeutics is never
computing; can be used easy [167]. Combinations related to chemotherapy produce com­
for determining pounded toxicity and bone-marrow suppression and therefore their
numerical simulations
stochastic ODEs and
potential remains to be seen. Despite a great deal of clinical research in
PDEs among others. virotherapy, not many mathematical models of chemovirotherapy yet
R Contains ODE solvers Open source software [204] exist. Mathematical modeling has shown enhancement of chemothera­
like deSolve. designed mainly for peutic drugs with oncolytic viruses leads to the depletion of all tumour
statistical computing
cells from a patient’s body tissue in less than a month [11,12,22]. This
and graphics; can be
used for carrying out time frame, nonetheless, leads to questions relating to toxicity. It is
numerical simulations therefore important for future mathematical studies to investigate the
of ODEs and PDEs. effect of oncolytic viruses on the tumour microenvironment. Mathe­
COPASI Contains solvers like An open source [205] matical models of immunovirotherapy showed that the influx of im­
Hybrid LSODA/SSA and software that can be
LSODAR. used for solving
mune cells around the tumour coupled with oncolytic viruses acts
mathematical mod-els synergistically to lyse cancer cells. Nonetheless, one important aspect of
of biological processes. oncolytic virotherapy that has not thoroughly been investigated, using
CompuSyn Analysis of [206] mathematical models, is the effect of the viruses on normal body cells.
combination data.
Moreover, viruses can trigger anti-viral immune responses, and this as
CalcuSyn Analyzes combined [207]
drug effects and is able well needs further investigation. Other models have shown radiotherapy
to be more effective when used synergistically with oncolytic virother­
apy [17,112], including estimating optimal dosage combinations [112].

9
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

Table 3 are still open [169].


Existing database for combinations of cancer therapy. Several mathematical models reviewed in this article, for simplicity,
Database Potential applications Website References ignore pertinent factors in cancer dy-namics for example, the pharma­
cogenomics of cancer treatment and tumour-immune interactions yet
DrugComb A data portal for https://drugcomb. [213]
integrative cancer fimm.fi/ these would ultimately influence the experimental outcomes and
drug combinations. mathematical results. It is therefore essential for future studies to
NCI Provides data showing [214] employ more complex mathematical models, considering detailed in­
ALMANAC how well pairs of FDA- formation for example the tumour microenvironment. Furthermore,
approved cancer drugs
kill tumour cells from
future mathematical models should focus on using different tools other
the NCI-60 Human than differential equations to consider scenarios that cannot be captured
Tumour Cell Lines. by ODEs and PDEs. Generally, there is also a dire need for more research
DrugCombDB Comprehensive http://drugcombdb. [215] to focus on consolidating the discovered mathematical insights into
database dedicated to denglab.org/main
clinical practice.
collecting drug
amalgamations from In contrast to the combinations summarized in Table 1, chemo­
various data sources. virotherapy represents a rationally designed combination due to the
CMTTdb Focuses on the key https://www.biosino.or [216] improved overall survival benefits, nonoverlapping safety profiles,
information for g/CMTTdb/ reduction of drug resistance, the impact on the tumour and of note is the
randomized clinical
trials (RCTs), such as
preclinical evidence of synergy between the components. In support of
patient selection this are the mathematical models on chemovirotherapy which have been
criteria, tumour presented in the previous sections. For example, a study by Malinzi [22]
conditions, therapy on mathematical analysis of chemovirotherapy indicated that the
modes (single or
tumour is likely to be cleared from the body in less than a month if both
combined therapy).
CANCROX Provides important http://cancrox.gmb.bio. [217] chemotherapy and virotherapy are used. Further, the study contended
information about br/view/index.php that in the treatment of cancer, oncolytic viruses enhance chemothera­
cancer treatment, drug peutic drugs which is also in agreement with [12]. Nonetheless,
combinations, genes designing an effective combo of viruses and chemothera-peutic drugs
and types of cancer.
will need to have a perfect balance between immune anti-viral and
OncoRx Detecting drug https://onco-informa [218]
interactions with tics.com/ anti-tumour responses.
chemotherapy Whilst some forms of cancer are incurable and treatment is only
regimens. administered to improve the quality of life and/or prolong life, a com­
SynToxProfiler Promotes the https://syntoxprofiler.fi [219]
bination therapy strategy that proves clinically to be of benefit to the
prioritization of drug mm.fi/stp/eerr
combinations based on nKuW9DeMSZ3r patient taking into account the cost incurred will prove highly valuable.
integrated analysis of clWtuLyrBpHbUvN% Today there are many treatment options for patients diagnosed with
synergy, efficacy and 2BWDUAXDxZLUg% cancer than there was a decade or two ago. Some of these treatments can
toxicity profiles. 3D/ produce significant responses that could completely eradicate metasta­
ReDO-DB Focuses exclusively on http://www.redo-pr [220]
static tumours [170]. Nonetheless, these treatments all experience the
the potential use of oject.org/db/
licensed non-cancer same problem, that is, drug resistance. This resistance of cancer cells to
medica- tions as drugs could ultimately lead to cancer relapse or recurrence. Molecular
sources of new cancer alterations may play a pivotal role in either acquired or intrinsic drug
ther- apeutics.
resistance and some of these alterations contributing to these resistance
CancerDR Identification of https://webs.iiitd.edu. [221]
mutations in can-cer in/ragh
include mutation of the drug’s molecular target, changes in the way the
genes (targets) as well ava/cancerdr/#thumb drug interacts with the tumour, broad cellular changes, and changes in
as natural variations in the tumour microenvironment among others [170]. Efforts are being
targets. made in order to understand how cells develop this resistance and more
importantly, how it can be overcome. While everyone knows that
Nonetheless, just like with other combina-tions, it is important that finding a cure to cancer is a hard task, it is evident today that combining
future studies focus on investigating the effect of using both treatments different strategies and minimizing the chances that it recurs, is defi­
on body tissue. Mathematical models of chemoradiotherapy and radio­ nitely the correct path to seek.
immunotherapy have shown that such combinations may lead to better We anticipate our understanding of the mechanisms of cancer to
treatment outcomes compared to a single approach. Whilst some studies ameliorate its burden in the near future, but the success will depend on a
[13,14] have attempted to identify optimal dosage combinations, no multidisciplinary approach across multiple sections of science, including
mathematical study has investigated the toxicity of the combina-tion mathematics, ecology, epidemiology, genetics and immunology. The
despite the adverse effects of radiotherapy alone being well docu­ early euphoria following breakthroughs exploiting combination thera­
mented. Although radioimmunotherapy is a promising treatment strat­ peutic strategies has been tainted by the existence of numerous chal­
egy, questions regarding its long-term safety issues still exist. There still lenges. To this end, we highlight the outstanding open problems that still
exist difficulties in estimating responses based on dosimetry and radio­ exist despite the reported success and numerous efforts in the research
biological models [168]. An understanding of how these treatments can community to combat cancer using combination anti-cancer therapy.
be best administered, how frequent the treatments can be administered Despite the growing expectations, in practice, combination cancer
and how to best integrate them with other agents to increase the overall therapy strategies have demonstrated in-consistent results that can be
response is yet to be developed [168]. In spite of the fact that it is still attributed to the problems incurred when conjoining these molecularly
possible to administer higher doses of radioactivity, it is still not known targeted agents [171]. It is however hard to establish whether the het­
if the ratio of the tumour to that of non-tumour is higher. In addition, erogeneity of response can be ascribed to the complications of tumour
practical questions such as ascertaining the efficacy of the amalgamation biology, poor drug penetration into the tumour or sub­
specifically the abscopal effect, how to sequence the combination to give optimal/uncoordinated exposure to the conjoined agents [171].
the best result and determining the optimal timing of the amalgamation Furthermore, the problem of determining the ratio at which therapeutic
agents should be mixed to produce maximal synergistic and minimal

10
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

anti-cancer effects is unanswered. The problems of how to find the best that is not included in the article is available on request from the cor­
drug combinations, as well as how to effectively combine therapy ex responding author.
vivo in human cells to avoid doing thousands of clinical trials is still
disturbing. Funding
It will be interesting to know how to effectively combine therapeutic
agents to produce effective results with appropriate sequencing of these No author received funding to specifically carry out this study.
agents. It is likely that combining two or more therapies will probably
increase toxicity if not ideally designed and thus the challenge of opti­ Authors’ contributions
mally constructing these agents to circumvent the adverse side effects to
a tolerable level is of great concern. This is likely to lead to suboptimal J.M conceptualized the idea and designed the study. K.B.B and J.M
administration of drugs and thus encouraging drug resistance in the wrote the initial draft of the article. H.A.A. and S.P reviewed and revised
course of the treatment. However, in clinical trials, it has always been the manuscript and made critical intellectual inputs. H.A.A designed the
essential to de-escalate the dose until the drugs become least toxic, a figures and finalized the manuscript. All authors proofread and revised
process which is time-consuming and laborious. the article before submission.
In conclusion, while there has been some remarkable achievement
and promise in the utilization of mathe-matical models in the combi­ Declaration of competing interest
nation treatment strategies of cancer, there is still a long way left to go.
Since cancer is a complex group of diseases, their mechanisms of The authors declare that they have no known competing financial
development are poorly understood and thus mathematical models interests or personal relationships that could have appeared to influence
developed, thus far, are relatively simple. Thus, an ideal and novel the work reported in this paper.
combination therapy selected for cancer treatment should significantly
lyse only cancer cells, increase the overall survival of a patient, reduce Acknowledgement
drug resistance, increase the efficacy and show preclinical evidence of
synergy. H.A.A thanks the South African Medical Research Council (SAMRC)
Mathematical modeling is making a significant contribution to the for a mid-career scientist and Self-initiated research grant; and the South
treatment of cancer. Thus given the pressure to find a value-based African National Research Foundation (NRF) for Research Development
healthcare cancer therapy, there is absolutely no qualm that a mathe­ Grant (RDG) for rated researchers.
matical approach will be an integral tool in determining the best com­
bination strategy at a reduced cost while improving cancer treatment References
outcomes in the years to come. However, despite the promising results
reported in various studies towards cancer treatment, most of them have [1] World Health Organisation. https://www.who.int/news-room/fact-sheets/deta
not been integrated for routine clinical use. il/cancer. Accessed: April 2020.
[2] Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail chemotherapy by
It is widely acknowledged that defeating cancer is unlikely to happen combination drug delivery systems. Adv Drug Deliv Rev 2016;98:19–34.
from a single therapeutic agent but rather a reasonable amalgamation of [3] Beil DR, Wein LM. Sequencing surgery, radiotherapy and chemotherapy: insights
harmonious compatible strategies chosen from diverse therapeutics from a mathematical analysis. Breast Canc Res Treat 2002;74(3):279–86.
[4] Benzekry S. Contributions in mathematical oncology: when theory meets reality.
includ-ing traditional and modern cancer therapies. Ideally, there is a PhD thesis. Université de Bordeaux; 2017.
dire need to optimize dosing, duration as well as relative scheduling of [5] Heath JR, Davis ME. Nanotechnology and cancer. Annu Rev Med 2008;59:
the diverse components by considering the potential synergies and the 251–65.
[6] Byrne HM. Dissecting cancer through mathematics: from the cell to the animal
probable antag-onisms, as well as toxicities. With the numerous possi­
model. Nat Rev Canc 2010;10(3):221–30.
bilities of combinatorial therapies and the outstanding challenge of [7] Araujo RP, McElwain DLS. A history of the study of solid tumour growth: the
figuring out the interplays between complex and intricated pharmaco­ contribution of mathematical modelling. Bull Math Biol 2004;66(5):1039–91.
[8] Barbolosi D, Ciccolini J, Lacarelle F, B, Barlési, André N. Computational
logic processes, we trust that optimization of these combinations in
oncology-mathematical modelling of drug regimens for precision medicine. Nat
silico before testing them in patients could be made possible by devel- Rev Clin Oncol 2016;13(4):242.
oping and analyzing integrated mathematical models. [9] Kuang Y, Nagy JD, Eikenberry SE. Introduction to mathematical oncology, vol.
Finally, we would like to note that this article does not provide an 59. CRC Press; 2016.
[10] Chappell M, Chelliah V, Cherkaoui M, Derks G, Dumortier T, Evans N,
intricate review of the mathematical equations which constitute the Ferrarini M, Fornari C, Ghazal P, Guerriero ML, et al. Mathematical modelling for
models that have been discussed. Rather, a comprehensive review of combinations of immuno-oncology and anti-cancer therapies. In: Quantitative
mathematical models for combination cancer therapy has been carried systems pharmacology UK meeting. Macclesfeld; 2015.
[11] Malinzi J, Eladdadi A, Sibanda P. Modelling the spatiotemporal dynamics of
out. We envisage that these models shall potentially play an essential chemovirotherapy cancer treatment. J Biol Dynam 2017;11(1):244–74.
role in deciding on strategies that are viable both medically and [12] Malinzi J, Ouifki R, Eladdadi A, Torres DFM, White KA. Enhancement of
economically with a high likelihood of yielding clinical benefits. chemotherapy using oncolytic virotherapy: mathematical and optimal control
analysis. Math Biosci Eng 2018;15(6):1435.
[13] Goldie JH, Coldman AJ, Ng V, Hopkins HA, Looney WB. A mathematical and
Ethical approval and consent to participate computer-based model of alternating chemotherapy and radiation therapy in
experimental neoplasms 1. In: Treatment modalities in lung cancer, vol. 41.
Karger Publishers; 1988. p. 11–20.
This study is exempt from ethical approval in all the authors’ [14] O’Donoghue JA, Sgouros G, Divgi CR, Humm JL. Single-dose versus fractionated
affiliations. radioimmunotherapy: model compar- isons for uniform tumor dosimetry. J Nucl
Med 2000;41(3):538–47.
[15] Kim PS, Crivelli JJ, Choi I, Yun C, Wares JR. Quantitative impact of
Consent to publish
immunomodulation versus oncolysis with cytokine- expressing virus
therapeutics. Math Biosci Eng 2015;12(4):841–58.
The authors guarantee that this work has not been previously pub­ [16] Friedman A, Lai X. Combination therapy for cancer with oncolytic virus and
checkpoint inhibitor: a mathematical model. PloS One 2018;13(2):e0192449.
lished elsewhere.
[17] Dingli D, Cascino MD, Josić K, Russell SJ, Bajzer Ž. Mathematical modeling of
cancer radiovirotherapy. Mathematical Biosciences; 2006.
Data availability [18] Hsieh MC, Yu HH, Chang WW, Lu CY, Chang CL, Chow JM, Chen SU, Cheng Y,
Wu SY. Adjuvant radiotherapy and chemotherapy improve survival in patients
with pancreatic adenocarcinoma receiving surgery: adjuvant chemotherapy alone
The authors declare that all data supporting the findings of this study is insufficient in the era of intensity modulation radiation therapy. Cancer
are available within the article. Any other supplementary information Medicine 2018;7(6):2328–38.

11
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

[19] Trimble EL, Ungerleider RS, Abrams JA, Kaplan RS, Feigal EG, Smith MA, [52] Wein LM, Wu JT, Kirn DH. Validation and analysis of a mathematical model of a
Carter CL, Friedman MA. Neoadjuvant therapy in cancer treatment. Cancer 1993; replication-competent oncolytic virus for cancer treatment: implications for virus
72(S11):3515–24. design and delivery. Canc Res 2003;63(6):1317–24.
[20] Ottolino-Perry K, Diallo J, Lichty BD, Bell JC, McCart JA. Intelligent design: [53] Wu JT, Byrne HM, Kirn DH, Wein LM. Modeling and analysis of a virus that
combination therapy with oncolytic viruses. Mol Ther 2010;18(2):251–63. replicates selectively in tumor cells. Bull Math Biol 2001;63(4):731.
[21] Chan W, Lam K, Siu W, Yuen K. Chemotherapy at end-of-life: an integration of [54] Tao Y, Guo Q. The competitive dynamics between tumor cells, a replication-
oncology and palliative team. Support Care Canc 2016;24(3):1421–7. competent virus and an immune response. J Math Biol 2005;51(1):37–74.
[22] Malinzi J. Mathematical analysis of a mathematical model of chemovirotherapy: [55] Eftimie R, Eftimie G. Tumour-associated macrophages and oncolytic
effect of drug infusion method. Computational and Mathematical Methods in virotherapies: a mathematical investigation into a complex dynamics. Letters in
Medicine 2019;2019. Biomathematics 2018;5(sup1). S6–S35.
[23] B Mokhtari R, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, Yeger H. [56] Malinzi J, Sibanda P, Mambili-Mamoboundou H. Analysis of virotherapy in solid
Combination therapy in combating cancer. Oncotarget 2017;8(23):38022. tumor invasion. Journal of Mathematical Biosciences 2015;263:102–10.
[24] Dry JR, Yang M, Saez-Rodriguez J. Looking beyond the cancer cell for effective [57] Steel GG, Peckham Michael J. Exploitable mechanisms in combined radiotherapy-
drug combinations. Genome Med 2016;8(1):125. chemotherapy: the concept of additivity. Int J Radiat Oncol Biol Phys 1979;5(1):
[25] Doroshow JH, Simon RM. On the design of combination cancer therapy. Cell 85–91.
2017;171(7):1476–8. [58] Grassberger C, Paganetti H. Methodologies in the modeling of combined chemo-
[26] Lopez JS, Banerji U. Combine and conquer: challenges for targeted therapy radiation treatments. Phys Med Biol 2016;61(21):R344.
combinations in early phase trials. Nat Rev Clin Oncol 2017;14(1):57. [59] Ergun A, Camphausen K, Wein LM. Optimal scheduling of radiotherapy and
[27] Emens LA. Chemoimmunotherapy. Cancer Journal (Sudbury, Mass.) 2010;16(4): angiogenic inhibitors. Bull Math Biol 2003;65(3):407–24.
295. [60] Salari E, Unkelbach J, Bortfeld T. A mathematical programming approach to the
[28] de Pillis L, Fister KR, Gu W, Collins C, Daub M, Gross D, Moore J, Preskill B. fractionation problem in chemoradiotherapy. IIE Trans Healthc Syst Eng 2015;5
Mathematical model creation for cancer chemo-immunotherapy. Computational (2):55–73.
and Mathematical Methods in Medicine 2009;10(3):165–84. [61] Medina-Echeverz J, Berraondo P. How can chemoimmunotherapy best be used
[29] De Lillo S, Salvatori MC, Bellomo N. Mathematical tools of the kinetic theory of for the treatment of colon cancer? Immunotherapy 2012;4(12):1787–90.
active particles with some reasoning on the modelling progression and [62] Ghaffari A, Bahmaie B, Nazari M. A mixed radiotherapy and chemotherapy model
heterogeneity. Math Comput Model 2007;45(5–6):564–78. for treatment of cancer with metastasis. Math Methods Appl Sci 2016;39(15):
[30] Adam JA. General aspects of modeling tumor growth and immune response. In: 4603–17.
A survey of models for tumor-immune system dynamics. Springer; 1997. [63] Marcu L, Bezak E, Olver I. Scheduling cisplatin and radiotherapy in the treatment
p. 15–87. of squamous cell carcinomas of the head and neck: a modelling approach. Phys
[31] Chaplain M, Matzavinos A. Mathematical modelling of spatio-temporal Med Biol 2006;51(15):3625.
phenomena in tumour immunology. In: Tutorials in mathematical biosciences III. [64] Barazzuol L, Burnet NG, Jena R, Jones B, Jefferies SJ, Kirkby NF. A mathematical
Springer; 2006. p. 131–83. model of brain tumour response to radiotherapy and chemotherapy considering
[32] De Boer RJ, Hogeweg P, Dullens HF, De Weger RA, Den Otter W. Macrophage T radiobiological aspects. J Theor Biol 2010;262(3):553–65.
lymphocyte interactions in the anti-tumor immune response: a mathematical [65] Powathil GG, Gordon KE, Hill LA, Chaplain MAJ. Modelling the effects of cell-
model. J Immunol 1985;134(4):2748–58. cycle heterogeneity on the response of a solid tumour to chemotherapy: biological
[33] Arciero JC, Jackson TL, Kirschner DE. A mathematical model of tumor-immune insights from a hybrid multiscale cellular automaton model. J Theor Biol 2012;
evasion and sirna treatment. Discrete Continuous Dyn Syst - Ser B (DCDS-B) 2004; 308(1–19).
4(1):39–58. [66] Jones B, Dale RG. Inclusion of molecular biotherapies with radical radiotherapy:
[34] de Pillis LG, Gu W, Radunskaya AE. Mixed immunotherapy and chemotherapy of modeling of combined modality treatment schedules. Int J Radiat Oncol Biol Phys
tumors: modeling, applications and biological interpretations. J Theor Biol 2006; 1999;45(4):1025–34.
238(4):841–62. [67] Jones B, Dale RG. The potential for mathematical modelling in the assessment of
[35] de Pillis LG, Radunskaya AE, Wiseman CL. A validated mathematical model of the radiation dose equivalent of cytotoxic chemotherapy given concomitantly
cell-mediated immune response to tumor growth. Canc Res 2005;65(17):7950–8. with radiotherapy. Br J Radiol 2005;78(934):939–44.
[36] Isaeva OG, Osipov VA. Different strategies for cancer treatment: mathematical [68] Plataniotis GA, Dale RG. Use of concept of chemotherapy-equivalent biologically
modelling. Computational and Mathematical Methods in Medicine 2009;10(4): effective dose to provide quantitative eval- uation of contribution of
253–72. chemotherapy to local tumor control in chemoradiotherapy cervical cancer trials.
[37] Kirschner D, Panetta JC. Modeling immunotherapy of the tumor–immune Int J Radiat Oncol Biol Phys 2008;72(5):1538–43.
interaction. J Math Biol 1998;37(3):235–52. [69] Plataniotis GA, Dale RG. Assessment of the radiation-equivalent of chemotherapy
[38] Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu Y. contributions in 1-phase radio-chemotherapy treatment of muscle-invasive
Irradiation and anti–pd-l1 treatment synergis- tically promote antitumor bladder cancer. Int J Radiat Oncol Biol Phys 2014;88(4):927–32.
immunity in mice. J Clin Invest 2014;124(2):687–95. [70] Moraru IC, Tai A, Erickson B, Li XA. Radiation dose responses for chemoradiation
[39] de Pillis LG, Radunskaya A. A mathematical model of immune response to tumor therapy of pancreatic cancer: an analysis of compiled clinical data using
invasion. In: Computational fluid and solid mechanics 2003. Elsevier; 2003. biophysical models. Practical Radiation Oncology 2014;4(1):13–9.
p. 1661–8. [71] Durante M, Tommasino F, Yamada S. Modeling combined chemotherapy and
[40] Rodrigues DS, Mancera PFA, Carvalho T, Gonçalves LF. A mathematical model for particle therapy for locally advanced pancreatic cancer. Frontiers in Oncology
chemoimmunotherapy of chronic lymphocytic leukemia. Appl Math Comput 2015;5:145.
2019;349:118–33. [72] Hartley A, Sanghera P, Glaholm J, Mehanna H, McConkey C, Fowler J.
[41] Pang L, Shen L, Zhao Z. Mathematical modelling and analysis of the tumor Radiobiological modelling of the therapeutic ratio for the addition of
treatment regimens with pulsed immunotherapy and chemotherapy. synchronous chemotherapy to radiotherapy in locally advanced squamous cell
Computational and Mathematical Methods in Medicine 2016;2016. carcinoma of the head and neck. Clin Oncol 2010;22(2):125–30.
[42] Vähä-Koskela JV, Heikkilä JE, Hinkkanen AE. Oncolytic viruses in cancer [73] Meade S, McConkey C, Sanghera P, Mehanna H, Hartley A. Revised
therapy. Canc Lett 2007;254(2):178–216. radiobiological modelling of the contribution of syn- chronous chemotherapy to
[43] Guo ZS, Thorne SH, Bartlett DL. Oncolytic virotherapy: molecular targets in the rate of grades 3–4 mucositis in head and neck cancer. Journal of Medical
tumor-selective replication and carrier cell- mediated delivery of oncolytic Imaging and Radiation Oncology 2013;57(6):733–8.
viruses. Biochim Biophys Acta Rev Canc 2008;1785(2):217–31. [74] Meade S, Sanghera P, McConkey C, Fowler J, Fountzilas G, Glaholm J, Hartley A.
[44] Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: a new era of cancer Revising the radiobiological model of synchronous chemotherapy in head-and-
treatment at dawn. Canc Sci 2016;107(10):1373–9. neck cancer: a new analysis examining reduced weighting of accelerated
[45] Li S, Tong J, Rahman MM, Shepherd TG, McFadden G. Oncolytic virotherapy for repopulation. Int J Radiat Oncol Biol Phys 2013;86(1):157–63.
ovarian cancer. Oncolytic Virotherapy 2012;1(1). [75] Sgouros G. Plasmapheresis in radioimmunotherapy of micrometastases: a
[46] Yu Z, Li S, Brader P, Chen N, Yong AY, Zhang Q, Szalay AA, Fong Y, Wong RJ. mathematical modeling and dosimetrical analysis. J Nucl Med: official
Oncolytic vaccinia therapy of squamous cell carcinoma. Mol Canc 2009;8(1):45. publication, Society of Nuclear Medicine 1992;33(12):2167–79.
[47] Nguyen A, Ho L, Wan Y. Chemotherapy and oncolytic virotherapy: advanced [76] Epenetos AA, Snook D, Durbin H, Johnson PM, Taylor-Papadimitriou J.
tactics in the war against cancer. Frontiers in Oncology 2014;4:145. Limitations of radiolabeled monoclonal antibodies for localization of human
[48] Ungerechts G, Frenzke ME, Yaiw K, Miest T, Johnston PB, Cattaneo R. Mantle cell neoplasms. Canc Res 1986;46(6):3183–91.
lymphoma salvage regimen: synergy between a reprogrammed oncolytic virus [77] Order SE, Sleeper AM, Stillwagon GB, Klein JL, Leichner PK. Radiolabeled
and two chemotherapeutics. Gene Ther 2010;17(12):1506. antibodies: results and potential in cancer therapy. Canc Res 1990;50(3
[49] Ulasov IV, Sonabend AM, Nandi S, Khramtsov A, Han Y, Lesniak MS. Combination Supplement):1011s–3s.
of adenoviral virotherapy and temo- zolomide chemotherapy eradicates [78] Begent RHJ, Pedley RB. Antibody targeted therapy in cancer: comparison of
malignant glioma through autophagic and apoptotic cell death in vivo. Br J Canc murine and clinical studies. Canc Treat Rev 1990;17(2–3):373–8.
2009;100(7):1154. [79] Sharkey RM, Blumenthal RD, Hansen HJ, Goldenberg DM. Biological
[50] Friedman A, Tian JP, Fulci G, Chiocca EA, Wang J. Glioma virotherapy: effects of considerations for radioimmunotherapy. Canc Res 1990;50(3 Supplement):
innate immune suppression and increased viral replication capacity. Canc Res 964s–9s.
2006;66(4):2314–9. [80] Fujimori K, Covell DG, Fletcher JE, Weinstein JN. Modeling analysis of the global
[51] Friedman A, Tao Y. Analysis of a model of a virus that replicates selectively in and microscopic distribution of im- munoglobulin G, F (ab’)2, and fab in tumors.
tumor cells. J Math Biol 2003;47(5):391–423. Canc Res 1989;49(20):5656–63.

12
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

[81] Fujimori K, Covell DG, Fletcher JE, Weinstein JN. A modeling analysis of [113] National Cancer Institute. https://www.cancer.gov/about-cancer/treatment/t
monoclonal antibody percolation through tumors: a binding-site barrier. J Nucl ypes/targeted-therapies/targeted-therapies-fact-sheet. Accessed: October 2020.
Med: Official Publication, Society of Nuclear Medicine 1990;31(7):1191–8. [114] Abbott LH, Michor F. Mathematical models of targeted cancer therapy. Br J Canc
[82] Fujimori K, Fisher DR, Weinstein JN. Integrated microscopic-macroscopic 2006;95(9):1136–41.
pharmacology of monoclonal antibody radiocon- jugates: the radiation dose [115] Bozic I, Reiter JG, Allen B, Antal T, Chatterjee K, Shah P, Moon Y, Yaqubie A,
distribution. Canc Res 1991;51(18):4821–7. Kelly N, Le DT, et al. Evolutionary dynamics of cancer in response to targeted
[83] van Osdol W, Fujimori K, Weinstein JN. An analysis of monoclonal antibody combination therapy. eLife 2013;2:e00747.
distribution in microscopic tumor nodules: consequences of a “binding site [116] Yu C, Liu X, Yang J, Zhang M, Jin H, Ma X, Shi H. Combination of immunotherapy
barrier. Canc Res 1991;51(18):4776–84. with targeted therapy: theory and practice in metastatic melanoma. Front
[84] Kumar D, Kumar S. A mathematical model of radioimmunotherapy for tumor Immunol 2019;10:990.
treatment. Afr J Math Comput Sci Res 2010;3(6):101–6. [117] Green AJ, Johnson CJ, Adamson KL, Begent RHJ. Mathematical model of
[85] Flux GD, Webb S, Ott RJ, Chittenden SJ, Thomas R. Three-dimensional dosimetry antibody targeting: important parameters defined using clinical data. Phys Med
for intralesional radionuclide therapy using mathematical modeling and Biol 2001;46(6):1679.
multimodality imaging. J Nucl Med 1997;38(7):1059–66. [118] Shen J, Li L, Yang T, Cohen PS, Sun G. Biphasic mathematical model of cell–drug
[86] Serre R, Benzekry S, Padovani L, Meille C, André N, Ciccolini J, Barlesi F, interaction that separates target-specific and off-target inhibition and suggests
Muracciole X, Barbolosi D. Mathematical modeling of cancer immunotherapy and potent targeted drug combinations for multi-driver colorectal cancer cells.
its synergy with radiotherapy. Canc Res 2016;76(17):4931–40. Cancers 2020;12(2):436.
[87] Melcher A, Parato K, Rooney CM, Bell JC. Thunder and lightning: immunotherapy [119] Sun X, guang Bao J, Shao Y. Mathematical modeling of therapy-induced cancer
and oncolytic viruses collide. Mol Ther 2011;19(6):1008–16. drug resistance: connecting cancer mecha- nisms to population survival rates. Sci
[88] Cheema TA, Fecci PE, Ning J, Rabkin SD. Immunovirotherapy for the treatment of Rep 2016;6:22498.
glioblastoma. OncoImmunology 2014;3(1):e27218. [120] Kozłowska E, Färkkilä A, Vallius T, Carpén O, Kemppainen J, Grénman S,
[89] Hardcastle J, Mills L, Malo CS, Jin F, Kurokawa C, Geekiyanage H, Schroeder M, Lehtonen R, Hynninen J, Hietanen S, Hau- taniemi S. Mathematical modeling
Sarkaria J, Johnson AJ, Galanis E. Immunovirotherapy with measles virus strains predicts response to chemotherapy and drug combinations in ovarian cancer.
in combination with anti–pd-1 antibody blockade enhances antitumor activity in Canc Res 2018;78(14):4036–44.
glioblastoma treatment. Neuro Oncol 2017;19(4):493–502. [121] Jarrett AM, Shah A, Bloom MJ, T McKenna M, Hormuth DA, Yankeelov TE,
[90] Cerullo V, Vähä-Koskela M, Hemminki A. Oncolytic adenoviruses: a potent form Sorace Anna G. Experimentally-driven mathematical modeling to improve
of tumor immunovirotherapy. OncoImmunology 2012;1(6):979–81. combination targeted and cytotoxic therapy for HER2+ breast cancer. Sci Rep
[91] Wares JR, Crivelli JJ, Yun C, Choi I, Gevertz JL, Kim PS. Treatment strategies for 2019;9(1):1–12.
combining immunostimulatory oncolytic virus therapeutics with dendritic cell [122] Owen MR, Stamper IJ, Muthana M, Richardson GW, Dobson J, Lewis CE,
injections. Math Biosci Eng 2015;12(6):1237–56. Byrne HM. Mathematical modeling predicts synergistic antitumor effects of
[92] Bagheri N, Shiina M, Lauffenburger DA, Korn WM. A dynamical systems model combining a macrophage-based, hypoxia-targeted gene therapy with
for combinatorial cancer therapy enhances oncolytic adenovirus efficacy by mek- chemotherapy. Canc Res 2011;71(8):2826–37.
inhibition. PLoS Comput Biol 2011;7(2):e1001085. [123] Tang Jing, Karhinen Leena, Xu Tao, Szwajda Agnieszka, Yadav Bhagwan,
[93] Huang J, Zhang S, Choi K, Choi I, Kim J, Lee M, Kim H, Yun C. Therapeutic and Wennerberg Krister, Aittokallio Tero. Target in- hibition networks: predicting
tumor-specific immunity induced by combination of dendritic cells and oncolytic selective combinations of druggable targets to block cancer survival pathways.
adenovirus expressing il-12 and 4-1bbl. Mol Ther 2010;18(2):264–74. PLoS Comput Biol 2013;9(9):e1003226.
[94] Dingli D, Offord C, Myers R, Peng K, Carr TW, Josic K, Russell SJ, Bajzer Z. [124] Tang Jing, Gautam Prson, Gupta Abhishekh, He Liye, Timonen Sanna,
Dynamics of multiple myeloma tumor therapy with a recombinant measles virus. Akimov Yevhen, Wang Wenyu, Szwajda Agnieszka, Jaiswal Alok, Turei Denes,
Canc Gene Ther 2009;16(12):873. et al. Network pharmacology modeling identifies synergistic aurora b and zak
[95] Rommelfanger DM, Offord CP, Dev J, Bajzer Z, Vile RG, Dingli D. Dynamics of interaction in triple-negative breast cancer. NPJ systems biology and applications
melanoma tumor therapy with vesicular stomatitis virus: explaining the 2019;5(1):1–11.
variability in outcomes using mathematical modeling. Gene Ther 2012;19(5):543. [125] Araujo RP, Petricoin EF, Liotta LA. A mathematical model of combination therapy
[96] Mok W, Stylianopoulos T, Boucher Y, Jain RK. Mathematical modeling of herpes using the egfr signaling network. Biosystems 2005;80(1):57–69.
simplex virus distribution in solid tumors: implications for cancer gene therapy. [126] Komarova Natalia L, Wodarz Dominik. Drug resistance in cancer: principles of
Clin Canc Res 2009;15(7):2352–60. emergence and prevention. Proc Natl Acad Sci Unit States Am 2005;102(27):
[97] DePillis L, Gallegos A, Radunskaya A. A model of dendritic cell therapy for 9714–9.
melanoma. Frontiers in Oncology 2013;3. [127] Charusanti Pep, Hu Xiao, Chen Luonan, Neuhauser Daniel, DiStefano III Joseph J.
[98] Pappalardo F, Pennisi M, Ricupito A, Topputo F, Bellone M. Induction of t-cell A mathematical model of bcr-abl au- tophosphorylation, signaling through the
memory by a dendritic cell vaccine: a computational model. Bioinformatics 2014; crkl pathway, and gleevec dynamics in chronic myeloid leukemia. Discrete &
30(13):1884–91. Continuous Dynamical Systems-B 2004;4(1):99.
[99] Dritschel H, Waters SL, Roller A, Byrne HM. A mathematical model of cytotoxic [128] Shen S, Duan J, Meredith RF, Buchsbaum DJ, Brezovich IA, Pareek PN,
and helper T cell interactions in a tumour microenvironment. Letters in Bonner JA. Model prediction of treatment plan- ning for dose-fractionated
Biomathematics 2018;5(sup1):S36–68. radioimmunotherapy. Cancer: Interdisciplinary International Journal of the
[100] Wu JT, Kirn DH, Wein LM. Analysis of a three-way race between tumor growth, a American Cancer Society 2002;94(S4):1264–9.
replication-competent virus and an immune response. Bull Math Biol 2004;66(4): [129] Callahan MK. Two drugs are better than one-modeling drug combinations in
605. cancer therapy. Sci Transl Med 2013;5(194). 194ec116–194ec116.
[101] Berg DR, Offord CP, Kemler I, Ennis MK, Chang L, Paulik G, Bajzer Z, [130] Chakwizira A, Ahlstedt J, Nittby Redebrandt H, Ceberg C. Mathematical
Neuhauser C, Dingli D. In vitro and in silico multidimensional modeling of modelling of the synergistic combination of radio- therapy and indoleamine-2, 3-
oncolytic tumor virotherapy dynamics. PLoS Comput Biol 2019;15(3):e1006773. dioxygenase (Ido) inhibitory immunotherapy against glioblastoma. Br J Radiol
[102] Wodarz D. Gene therapy for killing p53-negative cancer cells: use of replicating 2018;91(1087):20170857.
versus nonreplicating agents. Hum Gene Ther 2003;14(2):153–9. [131] Monjazeb AM, Grossenbacher SK, Sckisel GD, Canter R, Sparger EE, Culp W,
[103] Wodarz D, Hofacre A, Lau JW, Sun Z, Fan H, Komarova NL. Complex spatial Kent MS, Murphy WJ. Combined radiotherapy and immunotherapy using CPG
dynamics of oncolytic viruses in vitro: mathematical and experimental oligodeoxynucleotides and indolamine 2, 3 dioxygenase (Ido) blockade. Journal
approaches. PLoS Comput Biol 2012;8(6):e1002547. for Immunotherapy of Cancer 2013;1(1):P256.
[104] Bajzer Ž, Carr T, Josić K, Russell SJ, Dingli D. Modeling of cancer virotherapy [132] Radunskaya A, Kim R, Woods II, et al. Mathematical modeling of tumor immune
with recombinant measles viruses. J Theor Biol 2008;252(1):109–22. interactions: a closer look at the role of a PD-L1 inhibitor in cancer
[105] Biesecker M, J K, Lu H, Dingli D, Bajzer Ž. Optimization of virotherapy for cancer. immunotherapy. Spora: A Journal of Biomathematics 2018;4(1):25–41.
Bull Math Biol 2010;72(2):469–89. [133] Nazari F, Pearson AT, Nör JE, Jackson TL. A mathematical model for il-6-
[106] Gevertz JL, Wares JR. Developing a minimally structured mathematical model of mediated, stem cell driven tumor growth and targeted treatment. PLoS Comput
cancer treatment with oncolytic viruses and dendritic cell injections. Biol 2018;14(1):e1005920.
Computational and Mathematical Methods in Medicine 2018;2018. [134] Wang S, Schättler H. Optimal control of a mathematical model for cancer
[107] Timalsina A, Tian JP, Wang J. Mathematical and computational modeling for chemotherapy under tumor heterogeneity. Math. Biosci. and Engr.-MBE 2016;13:
tumor virotherapy with mediated immunity. Bull Math Biol 2017;79(8):1736–58. 1223–40.
[108] Jenner AL, Yun C, Yoon A, Coster ACF, Kim PS. Modelling combined virotherapy [135] Malinzi J. Mathematical modeling of cancer treatments and the role of the
and immunotherapy: strengthening the antitumour immune response mediated immune system response to tumor invasion. PhD thesis. 2015.
by il-12 and gm-csf expression. Letters in Biomathematics 2018;5(sup1):S99–116. [136] Spring BQ, Lang RT, Kercher EM, Rizvi I, Wenham RM, Conejo-Garcia JR,
[109] Mahasa KJ, Eladdadi A, De Pillis L, Ouifki R. Oncolytic potency and reduced virus Hasan T, Gatenby RA, Enderling H. Illu- minating the numbers: integrating
tumor-specificity in oncolytic virotherapy. a mathematical modelling approach. mathematical models to optimize photomedicine dosimetry and combination
PloS One 2017;12(9):e0184347. therapies. Frontiers in Physics 2019;7:46.
[110] Cassidy T, Humphries AR. A mathematical model of viral oncology as an immuno- [137] Rihan FA, Abdelrahman DH, Al-Maskari F, Ibrahim F, Abdeen MA. Delay
oncology instigator. Math Med Biol 2020;37(1):117–51. 04. differential model for tumour-immune response with chemoimmunotherapy and
[111] Wodarz D. Viruses as antitumor weapons: defining conditions for tumor optimal control. Computational and Mathematical Methods in Medicine 2014;
remission. Canc Res 2001;61(8):3501–7. 2014.
[112] Tao Y, Guo Q. A free boundary problem modelling cancer radiovirotherapy. Math [138] Oke S, Matadi M, Xulu S. Optimal control analysis of a mathematical model for
Model Methods Appl Sci 2007;17:1241–59. 08. breast cancer. Math Comput Appl 2018;23(2):21.

13
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

[139] Mamat M, Subiyanto KA, Kartono A. Mathematical model of cancer treatments [171] Tolcher AW, Mayer LD. Improving combination cancer therapy: the combiplex®
using immunotherapy, chemotherapy and biochemotherapy. Appl Math Sci 2013; development platform. Future Oncol 2018;14(13):1317–32.
7(5):247–61. [172] Shrivastava S, Mahantshetty U, Engineer R, Chopra S, Hawaldar R, Hande V,
[140] Lai X, Stiff A, Duggan M, Wesolowski R, Carson WE, Friedman A. Modeling Kerkar RA, Maheshwari A, Shylasree TS, Ghosh J, et al. Cisplatin
combination therapy for breast cancer with bet and immune checkpoint chemoradiotherapy vs radiotherapy in figo stage iiib squamous cell carcinoma of
inhibitors. Proc Natl Acad Sci Unit States Am 2018;115(21):5534–9. the uterine cervix: a randomized clinical trial. JAMA Oncology 2018;4(4):506–13.
[141] Joshi B, Wang X, Banerjee S, Tian H, Matzavinos A, Chaplain MAJ. On [173] Maletzki C, Wiegele L, Nassar I, Stenzel J, Junghanss C. Chemo-immunotherapy
immunotherapies and cancer vaccination protocols: a mathematical modelling improves long-term survival in a preclinical model of mmr-d-related cancer.
approach. J Theor Biol 2009;259(4):820–7. Journal for Immunotherapy of Cancer 2019;7(1):8.
[142] Kosinsky Y, Dovedi SJ, Peskov K, Voronova V, Chu L, Tomkinson H, Al-Huniti N, [174] Brown JR, Hallek MJ, Pagel JM. Chemoimmunotherapy versus targeted treatment
Stanski DR, Helmlinger G. Radi- ation and pd-(l) 1 treatment combinations: in chronic lymphocytic leukemia: when, how long, how much, and in which
immune response and dose optimization via a predictive systems model. Journal combination? American Society of Clinical Oncology Educational Book 2016;36:
for Immunotherapy of Cancer 2018;6(1):17. e387–98.
[143] Rihan FA, Lakshmanan S, Maurer H. Optimal control of tumour-immune model [175] Hallek M, Fischer K, Fingerle-Rowson G, Fink AM, Busch R, Mayer J, Hensel M,
with time-delay and immuno-chemotherapy. Appl Math Comput 2019;353: Hopfinger G, Hess G, Von Grünhagen U, et al. Addition of rituximab to
147–65. fludarabine and cyclophosphamide in patients with chronic lymphocytic
[144] Akman Yıldız T, Arshad S, Baleanu D. Optimal chemotherapy and leukaemia: a randomised, open-label, phase 3 trial. Lancet 2010;376(9747):
immunotherapy schedules for a cancer-obesity model with caputo time fractional 1164–74.
derivative. Math Methods Appl Sci 2018;41(18):9390–407. [176] Galluzzi L, Vacchelli E, Bravo-San Pedro J, Buqué A, Senovilla L, Baracco EE,
[145] Ratajczyk E, Ledzewicz U, Schättler H. Optimal control for a mathematical model Bloy N, Castoldi F, Abastado J, Agostinis P, et al. Classification of current
of glioma treatment with oncolytic therapy and TNF-α inhibitors. J Optim Theor anticancer immunotherapies. Oncotarget 2014;5(24):12472.
Appl 2018;176(2):456–77. [177] Sasse AD, Sasse EC, Go Clark L, Ulloa L, Clark OAC. Chemoimmunotherapy versus
[146] Sharma S, Samanta GP. Analysis of the dynamics of a tumor–immune system with chemotherapy for metastatic malig- nant melanoma. Cochrane Database Syst Rev
chemotherapy and immunotherapy and quadratic optimal control. Differential 2007;(1).
Equations and Dynamical Systems 2016;24(2):149–71. [178] Zhang P, Xi M, Zhao L, Shen J, Li Q, He L, Liu S, Liu M. Is there a benefit in
[147] Ledzewicz U, Schättler H. An optimal control approach to cancer chemotherapy receiving concurrent chemoradiotherapy for elderly patients with inoperable
with tumor–immune system interactions. In: Mathematical models of tumor- thoracic esophageal squamous cell carcinoma? PloS One 2014;9(8):e105270.
immune system dynamics. Springer; 2014. p. 157–96. [179] Advani SJ, Sibley GS, Song PY, Hallahan DE, Kataoka Y, Roizman B, R
[148] Nazari M, Ghaffari A, Arab F. Finite duration treatment of cancer by using vaccine Weichselbaum R. Enhancement of replication of genetically engineered herpes
therapy and optimal chemotherapy: state- dependent riccati equation control and simplex viruses by ionizing radiation: a new paradigm for destruction of
extended kalman filter. J Biol Syst 2015;23:1–29. 01. therapeutically intractable tumors. Gene Ther 1998;5(2):160.
[149] Parra-Guillen ZP, Berraondo P, Grenier E, Ribba B, Troconiz IF. Mathematical [180] Chung SM, Advani SJ, Bradley JD, Kataoka Y, Vashistha K, Yan SY, Markert JM,
model approach to describe tumour re- sponse in mice after vaccine Gillespie GY, Whitley RJ, Roizman B, et al. The use of a genetically engineered
administration and its applicability to immune-stimulatory cytokine-based herpes simplex virus (r7020) with ionizing radiation for experimental hepatoma.
strategies. AAPS J 2013;15(3):797–807. Gene Ther 2002;9(1):75.
[150] Rodrigues DS, de Arruda Mancera PF, et al. Mathematical analysis and [181] Blank SV, Rubin SC, Coukos G, Amin KM, Albelda SM, Molnar-Kimber KL.
simulations involving chemotherapy and surgery on large human tumours under Replication-selective herpes simplex virus type 1 mutant therapy of cervical
a suitable cell-kill functional response. Math Biosci Eng 2013;10(1):221–34. cancer is enhanced by low-dose radiation. Hum Gene Ther 2002;13(5):627–39.
[151] Villasana M, Ochoa G, Aguilar S. Modeling and optimization of combined [182] Eifel PJ. Chemoradiotherapy in the treatment of cervical cancer. In: Seminars in
cytostatic and cytotoxic cancer chemotherapy. Artif Intell Med 2010;50(3): radiation oncology, vol. 16. Elsevier; 2006. p. 177–85.
163–73. [183] Neuner G, Patel A, Suntharalingam M. Chemoradiotherapy for esophageal cancer.
[152] Chareyron S, Alamir M. Mixed immunotherapy and chemotherapy of tumors: Gastrointestinal Cancer Research: GCR 2009;3(2):57.
feedback design and model updating schemes. J Theor Biol 2009;258(3):444–54. [184] Shitara K, Muro K. Chemoradiotherapy for treatment of esophageal cancer in
[153] Ledzewicz U, Schattler H, d’Onofrio A. Optimal control for combination therapy Japan: current status and perspectives. Gastroin- testinal Cancer Research: GCR
in cancer. In: 2008 47th IEEE conference on decision and control. IEEE; 2008. 2009;3(2):66.
p. 1537–42. [185] Baxi SS, O’Neill C, Sherman EJ, L Atoria C, Lee NY, Pfister Dd G, Elkin EB. Trends
[154] d’Onofrio A, Ledzewicz U, Maurer H, Schättler H. On optimal delivery of in chemoradiation use in elderly patients with head and neck cancer: changing
combination therapy for tumors. Math Biosci 2009;222(1):13–26. treatment patterns with cetuximab. Head Neck 2016;38(S1):E165–71.
[155] Powathil G, Kohandel M, Sivaloganathan S, Oza A, Milosevic M. Mathematical [186] Kraeber-Bodéré F, Barbet J, Chatal J. Radioimmunotherapy: from current clinical
modeling of brain tumors: effects of radio- therapy and chemotherapy. Phys Med success to future industrial breakthrough? J Nucl Med 2016;57(3):329–31.
Biol 2007;52(11):3291. [187] Gopal AK, Gooley TA, Maloney DG, Petersdorf SH, Eary JF, Rajendran JG,
[156] Imbs D, El Cheikh R, Boyer A, Ciccolini J, Mascaux C, Lacarelle B, Barlesi F, Bush SA, Durack LD, Golden J, Martin PJ, et al. High-dose radioimmunotherapy
Barbolosi D, Benzekry S. Revisiting bevacizumab+ cytotoxics scheduling using versus conventional high-dose therapy and autologous hematopoietic stem cell
mathematical modeling: Proof of concept study in experimental non-small cell transplantation for relapsed follicular non-hodgkin lymphoma: a multivariable
lung carcinoma. CPT Pharmacometrics Syst Pharmacol 2018;7(1):42–50. cohort analysis. Blood 2003;102(7):2351–7.
[157] Tao Y, Guo Q. A mathematical model of combined therapies against cancer using [188] Msaouel P, Iankov ID, Allen C, Aderca I, Federspiel MJ, Tindall DJ, Morris JC,
viruses and inhibitors. Sci China, Ser A: Mathematics 2008;51(12):2315–29. Koutsilieris M, Russell SJ, Gala- nis E. Noninvasive imaging and radiovirotherapy
[158] Hadjiandreou MM, Mitsis GD. Mathematical modeling of tumor growth, drug- of prostate cancer using an oncolytic measles virus expressing the sodium iodide
resistance, toxicity, and optimal therapy design. IEEE (Inst Electr Electron Eng) symporter. Mol Ther 2009;17(12):2041–8.
Trans Biomed Eng 2014;61(2):415–25. [189] Li H, Peng K, Russell SJ. Oncolytic measles virus encoding thyroidal sodium
[159] Su Y, Jia C, Chen Y. Optimal control model of tumor treatment with oncolytic iodide symporter for squamous cell cancer of the head and neck radiovirotherapy.
virus and mek inhibitor. BioMed Res Int 2016;2016. Hum Gene Ther 2011;23(3):295–301.
[160] Kim PS, Lee PP. Modeling protective anti-tumor immunity via preventative cancer [190] Dingli D, Peng K, Harvey ME, Greipp PR, O’Connor MK, Cattaneo R, Morris JC,
vaccines using a hybrid agent-based and delay differential equation approach. Russell SJ. Image-guided ra- diovirotherapy for multiple myeloma using a
PLoS Comput Biol 2012;8(10):e1002742. recombinant measles virus expressing the thyroidal sodium iodide symporter.
[161] Preziosi L. Cancer modelling and simulation. CRC Press; 2003. Blood 2004;103(5):1641–6.
[162] Kelley WG, Peterson AC. The theory of differential equations: classical and [191] Touchefeu Y, Franken P, Harrington KJ. Radiovirotherapy: principles and
qualitative. Springer Science & Business Media; 2010. prospects in oncology. Curr Pharmaceut Des 2012;18(22):3313–20.
[163] Brauer F, Castillo-Chavez C. Mathematical models in population biology and [192] Chiocca EA. Oncolytic viruses. Nat Rev Canc 2002;2(12):938.
epidemiology, vol. 2. Springer; 2012. [193] Spear MA, Sun F, Eling DJ, Gilpin E, Kipps TJ, Chiocca EA, Bouvet M.
[164] Eqworld: The world of mathematical equations. http://eqworld.ipmnet. Cytotoxicity, apoptosis, and viral replication in tumor cells treated with oncolytic
ru/en/software.htm. Accessed: October 2020. ribonucleotide reductase-defective herpes simplex type 1 virus (hrr3) combined
[165] Mathematical software. https://www.mat.univie.ac.at/~neum/software.html. with ionizing radiation. Canc Gene Ther 2000;7(7):1051.
Accessed: October 2020. [194] Jorgensen TJ, Katz S, Wittmack EK, Varghese S, Todo T, Rabkin SD, Martuza RL.
[166] DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of Ionizing radiation does not alter the antitumor activity of herpes simplex virus
drug development costs. J Health Econ 2003;22(2):151–85. vector g207 in subcutaneous tumor models of human and murine prostate cancer.
[167] Binz E, Lauer UM. Chemovirotherapy: combining chemotherapeutic treatment Neoplasia 2001;3(5):451–6.
with oncolytic virotherapy. Oncolytic Virotherapy 2015;4:39. [195] Mathworks. Accessed: October 2020, http://www.mathworks.com/help/matlab.
[168] Sharkey RM, Goldenberg DM. Cancer radioimmunotherapy. Immunotherapy [196] Differential equations in maple. Accessed: October 2020, https://www.maplesoft.
2011;3(3):349–70. com/applications/view.aspx?sid=103786&view=html.
[169] Shun L, Meng X, Feng R, Yang L, Xing L, Yu J. The role of radiation oncology in [197] Wolfram Language & System Documentation Centre. https://reference.wolfram.
immuno-oncology. Oncol 2019;24(Suppl 1):S42–52. com/language/guide/EquationSolving.html. Accessed: October 2020.
[170] Identifying novel drug combinations to overcome treatment resistance. National [198] Python. https://www.python.org/. Accessed: October 2020.
Cancer Institute, https://www.cancer.gov/about-cancer/treatment/research/dru [199] Sage. https://doc.sagemath.org/html/en/reference/calculus/sage/calculus/desol
g-combo-resistance, Accessed May 2019. vers.html. Ac- cessed: October 2020.

14
J. Malinzi et al. Informatics in Medicine Unlocked 23 (2021) 100534

[200] Maxima, a computer algebra system. http://maxima.sourceforge.net/. Accessed: [214] Nci Almanac. A new tool for research on cancer drug combination. https://www.
October 2020. cancer.gov/news-events/cancer-currents-blog/2017/nci-almanac-drug-combina
[201] GNU Octave. https://www.gnu.org/software/octave/index. Accessed: October tions", Accessed January 2021.
2020. [215] Liu Hui, Zhang Wenhao, Zou Bo, Wang Jinxian, Deng Yuanyuan, Deng Lei.
[202] Mathcad. https://www.wikiwand.com/en/Mathcad. Accessed: October 2020. Drugcombdb: a comprehensive database of drug combinations toward the
[203] Julia. https://julialang.org. Accessed: October 2020. discovery of combinatorial therapy. Nucleic Acids Res 2020;48(D1):D871–81.
[204] The R project for statistical computing. https://www.r-project.org. Accessed: [216] Xue Bai, Yang Xiaobo, Wu Liangcai, Zuo Bangyou, Lin Jianzhen, Wang Shanshan,
October 2020. Jin Bian, Sang Xinting, He Yungang, Yang Zhen, et al. Cmttdb: the cancer
[205] Copasi. http://copasi.org/. Accessed: November 2020. molecular targeted therapy database. Ann Transl Med 2019;7(22).
[206] Compusyn. http://www.combosyn.com/. Accessed: November 2020. [217] Muniz de Ávila Paulo, Cesar de Melo Bernardo Paulo, Teodoro Marques da
[207] Calusyn. http://www.biosoft.com/w/calcusyn.htm. Accessed: November 2020. Silva Ramon Gustavo. Ana Lúcia Fachin, Mozart Marins, Edilson Carlos Caritá,
[208] He Liye, Wennerberg Krister, Aittokallio Tero, Tang Jing. Timma-r: an r package et al. Cancrox: a cross-species cancer therapy database. Database 2019:2019.
for predicting synergistic multi-targeted drug combinations in cancer cell lines or [218] Yi-Lwern Yap Kevin, Chan Alexandre, Keung Chui Wai, Chen Yu Zong. Cancer
patient-derived samples. Bioinformatics 2015;31(11):1866–8. informatics for the clinician: an interaction database for chemotherapy regimens
[209] Ianevski Aleksandr, Giri Anil K, Aittokallio Tero. Synergyfinder 2.0: visual and antiepileptic drugs. Seizure 2010;19(1):59–67.
analytics of multi-drug combination synergies. Nucleic Acids Res 2020. [219] Ianevski Aleksandr, Timonen Sanna, Alexander Kononov, Aittokallio Tero,
[210] Veroli Giovanni Y Di, Fornari Chiara, Wang Dennis, Mollard Severine, Giri Anil K. Syntoxprofiler: an interactive analysis of drug combination synergy,
Bramhall Jo L, Richards Frances M, Jodrell Duncan I. Combenefit: an interactive toxicity and efficacy. PLoS Comput Biol 2020;16(2):e1007604.
platform for the analysis and visualization of drug combinations. Bioinformatics [220] Pan Pantziarka, Verbaanderd Ciska, Sukhatme Vidula, Capistrano I Rica,
2016;32(18):2866–8. Crispino Sergio, Gyawali Bishal, Rooman Ilse, Van Nuffel An MT, Meheus Lydie,
[211] URDME. http://urdme.github.io/urdme/. Accessed January 2021. Sukhatme Vikas P, et al. Redo_db: the repurposing drugs in oncology database.
[212] Understand, predict and optimize physics-based designs and processes with ecancermedicalscience 2018;12.
COMSOL Multiphysics. https://www.comsol.com/comsol-multiphysics. [221] Kumar Rahul, Chaudhary Kumardeep, Gupta Sudheer, Singh Harinder,
Accessed: November 2020. Kumar Shailesh, Gautam Ankur, Kapoor Pallavi, Raghava Gajen- dra PS.
[213] Zagidullin B, Aldahdooh J, Zheng S, Wang W, Wang Y, Saad J, Malyutina A, Cancerdr: cancer drug resistance database. Sci Rep 2013;3:1445.
Jafari M, Tanoli Z, Pessia A, et al. Drugcomb: an integrative cancer drug
combination data portal. Nucleic Acids Res 2019;47(W1):W43–51.

15

You might also like