Вы находитесь на странице: 1из 9

Hindawi Publishing Corporation

BioMed Research International


Volume 2016, Article ID 5621313, 8 pages
http://dx.doi.org/10.1155/2016/5621313

Research Article
Optimal Control Model of Tumor Treatment with
Oncolytic Virus and MEK Inhibitor

Yongmei Su, Chen Jia, and Ying Chen


School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China

Correspondence should be addressed to Yongmei Su; suym71@ustb.edu.cn

Received 21 September 2016; Accepted 27 November 2016

Academic Editor: Tun-Wen Pai

Copyright © 2016 Yongmei Su et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Tumors are a serious threat to human health. The oncolytic virus is a kind of tumor killer virus which can infect and lyse cancer
cells and spread through the tumor, while leaving normal cells largely unharmed. Mathematical models can help us to understand
the tumor-virus dynamics and find better treatment strategies. This paper gives a new mathematical model of tumor therapy with
oncolytic virus and MEK inhibitor. Stable analysis was given. Because mitogen-activated protein kinase (MEK) can not only lead
to greater oncolytic virus infection into cancer cells, but also limit the replication of the virus, in order to provide the best dosage
of MEK inhibitors and balance the positive and negative effect of the inhibitors, we put forward an optimal control problem of the
inhibitor. The optimal strategies are given by theory and simulation.

1. Introduction a growing tumor and an oncolytic virus population as follows


[18]:
Tumors are a serious threat to human health, and chemother-
apy and radiotherapy may not only kill cancer cells, but also 𝑑𝑥 𝑥+𝑦
= 𝜆𝑥 (1 − ) − 𝛽𝑥V,
damage human body normal cells at the same time [1]. The 𝑑𝑡 𝐾
oncolytic virus is a kind of tumor killer virus which can infect 𝑑𝑦
and lyse cancer cells and spread through the tumor, while = 𝛽𝑥V − 𝛿𝑦, (1)
𝑑𝑡
leaving normal cells largely unharmed [2]. When oncolytic
viruses are inoculated into a cancer patient or directly 𝑑V
= 𝑏𝛿𝑦 − 𝛽𝑥V − 𝛾V,
injected into a tumor, these viruses will spread throughout the 𝑑𝑡
tumor and infect tumor cells. The viruses can be replicated in where variables 𝑥, 𝑦, and V stand for the population of
the infected tumor cells. When an infected tumor cell is lysed, uninfected cells, infected tumor cells, and oncolytic viruses,
it can burst out a mass of new oncolytic viruses. Then, these respectively. The coefficient 𝛽 represents the infection of the
new viruses can infect much more neighboring tumor cells virus. The tumor growth is modeled by logistic growth, and
[3]. 𝐾 is the maximal tumor size. 𝜆 is the per capita tumor growth
Experiments using oncolytic viruses such as adenovirus, rate. 𝛿 means the lysis rate of the infected tumor cells. 𝑏
CN706 [4], and ONYX-15 [5] in animal tumors show that represents the burst size of new viruses coming out from the
these viruses are nontoxic and infect tumor cells specifically. lysis of an infected tumor cell. 𝛾 represents the death rate of
Now, treatment of cancer with oncolytic virus has been the virus.
clinically tested [6–8]. This treatment of cancer with oncolytic It was shown that when the threshold 𝑏 < 1 + 𝛾/(𝐾𝛽), the
viruses has been explored by clinicians [9–11]. equilibrium solution (𝐾, 0, 0) is globally asymptotically stable
In recent years, in order to understand the cancer- [18], indicating that the oncolytic virus therapy finally has no
virus dynamics and find better treatment strategies, some effect. Obviously, the smaller the value of 𝐾, the more easily
mathematical models have been set up [12–19]. Tian pro- 𝑏 < 1 + 𝛾/(𝐾𝛽) holds. Since 𝐾 represents the total number
posed a mathematical model to describe the development of of tumor cells, smaller tumors may be more resistant to the
2 BioMed Research International

treatment by oncolytic virus than large ones, which should be Based on models (2) and (3), we establish the following
a contradiction. In [19], by replacing 𝛽𝑥V with 𝑥V/(𝑥 + 𝑦 + 𝜀), mathematical model:
we proposed the model 𝑑𝑥 𝑥+𝑦 𝛽𝑥V𝑧
= (1 − 𝑢) 𝑟𝑥 (1 − )− ,
𝑑𝑡 𝐾 𝑥+𝑦+𝜀

𝑑𝑥 𝑥+𝑦 𝑥V 𝑑𝑦 𝛽𝑥V𝑧
= 𝜆𝑥 (1 − )−𝛽 , = − (1 − 𝑢) 𝛿𝑦,
𝑑𝑡 𝐾 𝑥+𝑦+𝜀 𝑑𝑡 𝑥 + 𝑦 + 𝜀
(4)
𝑑𝑦 𝑥V 𝑑V 𝛽𝑥V𝑧
=𝛽 − 𝛿𝑦, (2) = 𝑏 (1 − 𝑢) 𝛿𝑦 − − 𝛼V,
𝑑𝑡 𝑥+𝑦+𝜀 𝑑𝑡 𝑥+𝑦+𝜀
𝑑𝑧
𝑑V 𝑥V = 𝑔𝑢 (𝑝 − 𝑧) − 𝑐𝑧.
= 𝑏𝛿𝑦 − 𝛽 − 𝛾V. 𝑑𝑡
𝑑𝑡 𝑥+𝑦+𝜀
The variables 𝑥, 𝑦, V, and 𝑧 have the same meanings as those
in model (3). The parameters 𝜆, 𝛽, 𝛿, 𝛾, 𝑏, and 𝐾 are the
same as those of (1). The parameter 𝑢 has the same meaning
The meanings of variables 𝑥, 𝑦, and V and parameters as that in model (3). All the parameters are strictly positive.
𝜆, 𝛽, 𝛿, 𝛾, 𝑏, and 𝐾 are the same as those in model (1), and Since the use of MEK inhibitors not only results in
𝜀 is positive and sufficiently small. The threshold obtained enhanced oncolytic virus entry into the tumor cells, but also
by our model is 𝑏 < 1 + (𝛾/𝛽)(1 + 𝜀/𝐾), which is almost renders infected cells temporarily unable to produce viruses,
independent of 𝐾 when 𝜀 is sufficiently small. the maximum dosage of MEK use may not result in the best
On the other hand, all the above papers did not consider treatment effect, so the optimal control-based schedules of
coxsackie-adenovirus receptor (CAR). In fact, CAR is a main MEK inhibitor application should be studied. The optimal
receptor when oncolytic viruses enter into tumor cells [20– MEK inhibitor application strategy can increase the efficacy
22]. The successful entry of viruses into cancer cells is related of this treatment in an economical fashion. So, first, in this
to the presence of CAR. When oncolytic viruses infect the paper, we let the control variable 𝑢 be a constant; a stability
tumor cells, firstly, they combine with the CAR and are analysis of our model is conducted, and then the optimal
absorbed into the cells. control strategy is discussed; we also compare the optimal
Mitogen-activated protein kinase (MEK) inhibitors have control with constant control by simulation.
been shown to promote CAR expression and could increase
oncolytic viruses infection into tumor cells. But MEK
inhibitors may also limit the replication of viruses [23–25], 2. Materials and Methods
which will affect the treatment by oncolytic virus. With the
2.1. Stability Analysis. System (4) always has two equilibrium
function of MEK, [25] gave a model:
points:
𝑔𝑢𝑝
𝐸0 = (0, 0, 0, ),
𝑑𝑥 𝛽𝑧𝑥V 𝑔𝑢 + 𝑐
= 𝜌𝑥 (1 − 𝑢) − 𝑑𝑥 − , (5)
𝑑𝑡 1 + 𝜀V 𝑔𝑢𝑝
𝐸1 = (𝐾, 0, 0, ).
𝑑𝑦 𝛽𝑧𝑥V 𝑔𝑢 + 𝑐
= − 𝑑𝑦 − 𝑎 (1 − 𝑢) 𝑦,
𝑑𝑡 1 + 𝜀V If 𝜀 is sufficiently small, when 𝑏 > 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾)((𝑔𝑢 +
(3) 𝑐)/𝑔𝑢𝑝), the third steady state 𝐸2 = (𝑥2 , 𝑦2 , V2 , 𝑧2 ) exists in
𝑑V
= 𝑘 (1 − 𝑢) 𝑦 − 𝑏V, which
𝑑𝑡
Δ + √Δ2 + 4 (𝑏 − 1) 𝐾𝛼𝛽𝑧2 𝑟 (1 − 𝑢)2 𝛿𝜀
𝑑𝑧 𝑥2 = ,
= 𝜂𝑢 (𝑝 − 𝑧) − 𝑐𝑧. 2 (𝑏 − 1) 𝛽𝑧2 𝑟 (1 − 𝑢)
𝑑𝑡
(𝑏 − 1) 𝛽𝑧2
𝑦2 = [ − 1] 𝑥2 − 𝜀,
𝛼 (6)
The variables 𝑥, 𝑦, and V have the same meanings as those in
model (2); 𝑧 represents the average expression level of CAR (𝑏 − 1) (1 − 𝑢) 𝛿
V2 = 𝑦2 ,
on the surface of the cells. The intensity of MEK inhibitor 𝛼
application is captured in the parameter 𝑢, 𝑢 ∈ [0, 1]. If 𝑔𝑢𝑝
𝑢 = 0, there is no MEK inhibitor application, and the 𝑧2 = .
𝑔𝑢 + 𝑐
CAR expression level will gradually decline. If 𝑢 = 1, the
MEK inhibitor has the maximum possible effect. The model Here,
assumes that exponential growth can be slowed down by the Δ = (1 − 𝑢) 𝑟𝛼 (𝐾 + 𝜀) − 𝐾 (𝑏 − 1) (1 − 𝑢) 𝛿𝛽𝑧2
inhibitor with expression 1 − 𝑢. CAR grow at the rate of (7)
𝑔(𝑝 − 𝑧) and become extinct at the rate of 𝑐. + 𝐾𝛼 (1 − 𝑢) 𝛿.
BioMed Research International 3

It should be noted that 𝑏 > 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾)((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝) 𝑐)/𝛽𝐾𝑔𝑢𝑝. But we need to show the boundness of system (4).
is equivalent to (𝑏 − 1)𝛽𝑧2 /𝛼 − 1 > 0 to ensure that 𝑦2 > 0 From the first two equations, we obtain
when 𝜀 is sufficiently small.
The Jacobi matrix at point 𝐸0 is 𝑑 (𝑥 (𝑡) + 𝑦 (𝑡))
𝑑𝑡
𝑟 (1 − 𝑢) 0 0 0 𝑥 (𝑡) + 𝑦 (𝑡)
= (1 − 𝑢) 𝑟𝑥 (𝑡) (1 − ) − (1 − 𝑢) 𝛿𝑦 (𝑡) (12)
0 − (1 − 𝑢) 𝛿 0 0 𝐾
𝐽|𝐸0 = ( ). (8) 𝑥 (𝑡) + 𝑦 (𝑡)
0 𝑏𝛿 −𝛼 0 ≤ 𝑟 (𝑥 (𝑡) + 𝑦 (𝑡)) (1 − ).
𝐾
0 0 0 −𝑔𝑢 − 𝑐
By the comparison principle, we can obtain lim𝑡→∞ sup(𝑥(𝑡) +
𝑦(𝑡)) ≤ 𝐾.
Obviously, 𝜇1 = 𝑟(1 − 𝑢) > 0 is one eigenvalue of 𝐽|𝐸0
From the third equation of (4), we can have
which means 𝐸0 is unstable. The unstable result of 𝐸0 seems
consistent with the biological meaning that, without viruses 𝑑V 𝑥V𝑧
and infected tumor cells, the tumor will grow from an initial ≤ (1 − 𝑢) 𝑏𝛿𝐾 − 𝛽 − 𝛼V
𝑑𝑡 𝑥+𝑦+𝜀 (13)
small value around 𝐸0 .
As for equilibrium point 𝐸1 , we have the following ≤ (1 − 𝑢) 𝑏𝛿𝐾 − 𝛼V.
theorem.
It is easily shown that V(𝑡) ≤ (1 − 𝑢)𝑏𝛿𝐾/𝛼.
Theorem 1. When 𝑏 < 1+(𝛼(𝐾+𝜀)/𝛽𝐾)⋅((𝑔𝑢+𝑐)/𝑔𝑢𝑝), 𝐸1 is Similarly, from
locally asymptotically stable. When 𝑏 > 1+(𝛼(𝐾+𝜀)/𝛽𝐾)((𝑔𝑢+
𝑐)/𝑔𝑢𝑝), 𝐸1 is unstable. 𝑑𝑧 𝑔𝑢 + 𝑐
= 𝑔𝑢 (𝑝 − 𝑧) − 𝑐𝑧 = 𝑔𝑢𝑝 (1 − 𝑧) , (14)
𝑑𝑡 𝑔𝑢𝑝
Proof. At the equilibrium point 𝐸1 , the Jacobi matrix is
we can get that 𝑧(𝑡) ≤ 𝑔𝑢𝑝/(𝑔𝑢 + 𝑐) holds.

𝐽|𝐸1 Theorem 2. When 𝑏 < 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾) ⋅ ((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝),


𝐸1 is globally asymptotically stable.
− (1 − 𝑢) 𝑟 − (1 − 𝑢) 𝑟 −𝐻 0
Proof. Consider the Lyapunov function 𝑉 = 𝑦 + (1/𝑏)V; the
0 − (1 − 𝑢) 𝛿 𝐻 0 (9)
=( ), derivative along a solution is given by
0 𝑏 (1 − 𝑢) 𝛿 −𝛼 − 𝐻 0
1
0 0 0 −𝑔𝑢 − 𝑐 𝑉̇ = 𝑦̇ + V̇
𝑏
𝑥V𝑧
where 𝐻 = 𝛽𝐾𝑔𝑢𝑝/(𝐾 + 𝜀)(𝑔𝑢 + 𝑐). The eigenvalues of 𝐽|𝐸1 =𝛽 − (1 − 𝑢) 𝛿𝑦
𝑥+𝑦+𝜀
are 𝜇1 = −(1 − 𝑢)𝑟, 𝜇2 = −𝑔𝑢 − 𝑐, (15)
1 𝑥V𝑧
+ (𝑏 (1 − 𝑢) 𝛿𝑦 − 𝛽 − 𝛼V)
𝑏 𝑥+𝑦+𝜀
− ((1 − 𝑢) 𝛿 + 𝛼 + Δ 2 ) ± 𝐺
𝜇3,4 = . (10)
2 1 𝛽𝑥V𝑧 𝛼
= (1 − ) − V.
𝑏 𝑥+𝑦+𝜀 𝑏
Here, 𝐺 = √(𝛿(1 − 𝑢) − 𝛼 − Δ 2 )2 + 4Δ 2 𝑏𝛿(1 − 𝑢) in which Since 0 < 𝑥 ≤ 𝐾, 0 < 𝑥 + 𝑦 ≤ 𝐾, we have 𝑥𝐾 + 𝑥𝜀 ≤
Δ 2 = 𝛽𝐾𝑢𝑔𝑝/(𝐾 + 𝜀)(𝑔𝑢 + 𝑐). 𝑥𝐾 + 𝐾𝜀 + 𝐾𝑦, which implies
When 𝑏 < 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾)((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝), we have
𝑥 𝐾
≤ , (16)
𝑥+𝑦+𝜀 𝐾+𝜀
2
𝐺 = ((1 − 𝑢) 𝛿 − 𝛼 − Δ 2 ) + 4Δ 2 𝑏 (1 − 𝑢) 𝛿
(11) and because 𝑧 ≤ 𝑔𝑢𝑝/(𝑔𝑢 + 𝑐), therefore,
2
< ((1 − 𝑢) 𝛿 + 𝛼 + Δ 2 ) , 1 𝛽𝑥V𝑧 𝛼
𝑉̇ = (1 − ) − V
𝑏 𝑥+𝑦+𝜀 𝑏
which ensure that 𝜇3 and 𝜇4 are negative, so 𝐸1 is locally
1 𝛽𝐾V 𝑔𝑢𝑝 𝛼
asymptotically stable. ≤ (1 − ) − V (17)
Similarly, when 𝑏 > 1 + 𝛼(𝐾 + 𝜀)(𝑔𝑢 + 𝑐)/𝛽𝐾𝑔𝑢𝑝, 𝜇3 is 𝑏 𝐾 + 𝜀 𝑔𝑢 + 𝑐 𝑏
positive, and 𝐸1 is unstable.
𝛽 (𝑏 − 1) 𝐾𝑔𝑝 − 𝛼 (𝐾 + 𝜀) (𝑔𝑢 + 𝑐)
Actually, we can prove that the equilibrium solution 𝐸1 is = V.
globally asymptotically stable when 𝑏 < 1 + 𝛼(𝐾 + 𝜀)(𝑔𝑢 + 𝑏 (𝐾 + 𝜀) (𝑢𝑔 + 𝑐)
4 BioMed Research International

When 𝑏 < 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾) ⋅ ((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝), we can have 109


𝑉̇ ≤ 0.
Let 𝐸 = {(𝑥, 𝑦, V, 𝑧) | 𝑉̇ = 0}; it is clear that 𝐸 ⊂
{(𝑥, 𝑦, V, 𝑧) | V = 0}. Let 𝑀 be the largest positively invariant x 108
subset of the set 𝐸; by the third equation of system (4), we
can know that 𝑦(𝑡) = 0, so 𝑀 = {(𝑥, 𝑦, V) | 𝑦 = 0, V = 0}. By
LaSalle invariance principal [26], we know 107
0 200 400 600 800 1000
t
lim 𝑦 (𝑡) = 0, ×108
𝑡→∞
(18) 6
lim V (𝑡) = 0.
𝑡→∞
4
So, the limit equation of system (4) is y
2
𝑑𝑥 𝑥
= (1 − 𝑢) 𝑟𝑥 (1 − ) , 0
𝑑𝑡 𝐾 0 200 400 600 800 1000
(19)
𝑑𝑧 t
= 𝑔𝑢 (𝑝 − 𝑧) − 𝑐𝑧.
𝑑𝑡 Figure 1: State dynamics for uninfected and infected tumor cells
when 𝑏 = 10.
Therefore, 𝑥(𝑡) → 𝐾, 𝑧 → 𝑔𝑢𝑝/(𝑔𝑢 + 𝑐) when 𝑡 → ∞. So,
𝐸1 is globally attractive; note that 𝑏 < 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾) ⋅
((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝) can also ensure the local asymptotical stability 109
of 𝐸1 , so we can know that 𝐸1 of system (4) is globally
asymptotically stable when 𝑏 < 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾) ⋅ ((𝑔𝑢 + 108
𝑐)/𝑔𝑢𝑝). x
Although we can prove the global asymptotical stability 107
of E1 , we would not want this to happen, because the global
asymptotical stability means the therapy does not have any 106
0 200 400 600 800 1000
effect. When 𝑏 > 1 + (𝛼(𝐾 + 𝜀)/𝛽𝐾)((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝) holds, t
the coexistent steady state 𝐸2 = (𝑥2 , 𝑦2 , V2 , 𝑧2 ) exists, but it is ×108
difficult to give the stable analysis of E2 , so we just give some 8
simulations about it. 6
We choose 𝛽 = 0.2 day−1 , 𝜀 = 0.009, 𝛿 = 0.5 day−1 ,
y 4
𝑟 = 6 cells day−1 , 𝛼 = 0.5 day−1 , 𝑝 = 10, 𝑔 = 0.1 day−1 ,
𝑐 = 0.5 day−1 , and 𝐾 = 9 × 108 cells. 2
The initial condition is 𝑥0 = (6×107 , 0, 5×104 , 4×102 ), 0
where the unit of each is cells. 0 200 400 600 800 1000
We choose 𝑏 = 10 cell−1 day−1 , 𝑏 = 14 cell−1 day−1 , t
respectively, and 𝑏 > 1+(𝛼(𝐾+𝜀)/𝛽𝐾)((𝑔𝑢+𝑐)/𝑔𝑢𝑝) = 2.6389 Figure 2: State dynamics for uninfected and infected tumor cells
all hold; the simulation results are shown in Figures 1 and 2. when 𝑏 = 14.
The simulation results show that oncolytic virus therapy
may keep the tumor stable at some level as shown in Figure 1
or keep oscillating at a certain range as shown in Figure 2.
cells temporarily unable to produce viruses, how to use the
Since the cured equilibrium is always unstable, just from our
MEK inhibitors optimally should be studied. In model (4),
model, we could not give the condition that ensures the tumor
the function of MEK inhibitors was embodied by parameter
can be cured by oncolytic virus therapy, but if we choose
𝑢; we use it as the control variable. The control goal is not
appropriate u which satisfies
only to formulate an objective functional which lowers the
𝛼 (𝐾 + 𝜀) 𝑔𝑢 + 𝑐 levels of tumor cells during and at the end of therapy, but
𝑏>1+ , (20) also to minimize the cost of MEK, so the objective function
𝛽𝐾 𝑔𝑢𝑝
is defined as follows:
the simulation shows that oncolytic virus therapy can prevent
1 1
the tumor from getting worse and worse. Some other therapy 𝐽 (𝑢) = 𝑎11 𝑥2 + 𝑎22 𝑦2
methods should be combined to cure the tumor. 2 2
(21)
1 𝑡𝑓
+ ∫ (𝑏11 𝑥2 + 𝑏22 𝑦2 + 𝑏33 V2 + 𝑐11 𝑢2 ) 𝑑𝑡,
2.2. The Optimal Control of MEK. In the simulation of 2 𝑡0
Figures 1 and 2, we choose the control 𝑢 as constant. Since
the use of MEK inhibitors not only results in enhanced where 𝑡0 represents the beginning time of the treatment
oncolytic virus entry into the cells, but also renders infected and 𝑡𝑓 represents the terminal time of the treatment.
BioMed Research International 5

𝑎11 , 𝑎22 , 𝑏11 , 𝑏22 , 𝑏33 , and 𝑐11 represent the cost coefficients (30 )
for the variables, respectively.
For convenience, we define the state vector 𝑋 = (𝑥, 𝑦, V,
𝜕𝐻
𝑇
𝑧) ; system (4) can be written as 𝜆̇ = −
𝜕𝑋

𝑋̇ = 𝑓 (𝑋 (𝑇) , 𝑢 (𝑡) , 𝑡) , 𝜆 1 ⋅ 𝑝𝑓11 + 𝜆 2 ⋅ 𝑝𝑓21 + 𝜆 3 ⋅ 𝑝𝑓31 + 𝜆 4 ⋅ 𝑝𝑓41 + 𝑏11 𝑥


(22) [ ] (28)
[𝜆 1 ⋅ 𝑝𝑓12 + 𝜆 2 ⋅ 𝑝𝑓22 + 𝜆 3 ⋅ 𝑝𝑓32 + 𝜆 4 ⋅ 𝑝𝑓42 + 𝑏22 𝑦]
𝑋 (𝑡0 ) = 𝑋0 . = −[
[ 𝜆 ⋅ 𝑝𝑓 + 𝜆 ⋅ 𝑝𝑓 + 𝜆 ⋅ 𝑝𝑓 + 𝜆 ⋅ 𝑝𝑓 + 𝑏 V ] ,
]
[ 1 13 2 23 3 33 4 43 33 ]

And the corresponding cost function is defined as follows: [ 𝜆 1 ⋅ 𝑝𝑓14 + 𝜆 2 ⋅ 𝑝𝑓24 + 𝜆 3 ⋅ 𝑝𝑓34 + 𝜆 4 ⋅ 𝑝𝑓44 ]

1 1 𝑡𝑓
𝐽 = 𝑋𝑇 𝐴𝑋 + ∫ (𝑋𝑇 𝐵𝑋 + 𝐶𝑢2 ) 𝑑𝑡. (23)
2 2 𝑡0
where
Here,
2𝑥 + 𝑦 𝛽 (𝑦 + 𝜀) V𝑧
𝑎11 0 0 0 𝑝𝑓11 = 𝑟 (1 − 𝑢) (1 − )− 2
,
𝐾 (𝑥 + 𝑦 + 𝜀)
0 𝑎22 0 0
𝐴=( ), 𝛽𝑥V𝑧 𝑟
0 0 0 0 𝑝𝑓12 = 2
− (1 − 𝑢) 𝑥,
(𝑥 + 𝑦 + 𝜀) 𝐾
0 0 0 0
𝛽𝑥𝑧
𝑏11 0 0 0 (24) 𝑝𝑓13 = − ,
𝑥+𝑦+𝜀
0 𝑏22 0 0 𝛽𝑥V
𝐴=( ), 𝑝𝑓14 = − ,
0 0 𝑏33 0 𝑥+𝑦+𝜀
0 0 0 0
𝛽 (𝑦 + 𝜀) V𝑧
𝑝𝑓21 = 2
,
𝐶 = 𝑐11 . (𝑥 + 𝑦 + 𝜀)

Based on the dynamic constraint 𝑓(𝑋(𝑇), 𝑢(𝑡), 𝑡) and the 𝛽𝑥V𝑧


𝑝𝑓22 = − (1 − 𝑢) 𝛿 − 2
,
Lagrangian 𝐿(𝑋(𝑡), 𝑢(𝑡), 𝑡), the Hamiltonian is as follows: (𝑥 + 𝑦 + 𝜀)

𝐻 (𝑋 (𝑡) , 𝑢 (𝑡) , 𝑡) = 𝐿 (𝑋 (𝑡) , 𝑢 (𝑡) , 𝑡) 𝛽𝑥𝑧


𝑝𝑓23 = ,
(25) 𝑥+𝑦+𝜀
+ 𝜆𝑇 𝑓 (𝑋 (𝑡) , 𝑢 (𝑡) , 𝑡)
𝛽𝑥V (29)
𝑝𝑓24 = ,
where 𝐿(𝑋(𝑡), 𝑢(𝑡), 𝑡) = (1/2)(𝑋𝑇 𝐵𝑋 + 𝑐11 𝑢2 ). 𝑥+𝑦+𝜀
Using Pontryagin’s minimum principle, the necessary
conditions are given as follows: 𝛽 (𝑦 + 𝜀) V𝑧
𝑝𝑓31 = − 2
,
(𝑥 + 𝑦 + 𝜀)
(10 )
𝛽𝑥V𝑧
𝑝𝑓32 = 𝑏 (1 − 𝑢) 𝛿 + ,
𝑋̇ = 𝑓 (𝑋 (𝑡) , 𝑢 (𝑡) , 𝑡) (𝑥 + 𝑦 + 𝜀)
2

{ 𝑑𝑥 𝑥+𝑦 𝛽𝑥V𝑧 𝛽𝑥𝑧


{
{ = (1 − 𝑢) 𝑟𝑥 (1 − )− 𝑝𝑓33 = − − 𝛼,
{
{ 𝑑𝑡
{
{ 𝛽𝑥V𝑧
𝑘 𝑥+𝑦+𝜀 𝑥+𝑦+𝜀
{
{ 𝑑𝑥
{
{ = − (1 − 𝑢) 𝛿𝑦 (26)
𝛽𝑥V
= { 𝑑𝑡 𝑥 + 𝑦 + 𝜀 𝑝𝑓34 = − ,
{
{ 𝑑V 𝛽𝑥V𝑧 𝑥+𝑦+𝜀
{
{ = 𝑏 (1 − 𝑢) 𝛿𝑦 − − 𝛼V
{
{ 𝑑𝑡 𝑥 +𝑦+𝜀
{
{
{
{ 𝑑𝑧 = 𝑔𝑢 (𝑝 − 𝑧) − 𝑐𝑧, 𝑝𝑓41 = 0,
{ 𝑑𝑡
𝑝𝑓42 = 0,
(20 ) 𝑝𝑓43 = 0,

𝑋 (𝑡0 ) = 𝑋0 , (27) 𝑝𝑓44 = −𝑔𝑢 − 𝑐,


6 BioMed Research International

(40 ) 1
0.9
𝑎11 0 0 0 𝑥 (𝑡𝑓 )
[ ][ ] 0.8
[ 0 𝑎22 0 0] [
[𝑦 (𝑡𝑓 )]
]
[
𝜆 (𝑡𝑓 ) = 𝐴𝑋 (𝑡𝑓 ) = [ ] [ ], (30) 0.7
]
[ 0 0 0 0] [ ]
[ V (𝑡𝑓 ) ] 0.6
[ 0 0 0 0] [ 𝑧 (𝑡𝑓 ) ] u 0.5
0.4
(50 )
0.3
𝜕𝐻 𝑥+𝑦 0.2
= 𝑐11 𝑢 − 𝜆 1 𝑟𝑥 (1 − ) + 𝜆 2 𝛿𝑦 − 𝜆 3 𝑏𝛿𝑦
𝜕𝑢 𝐾 (31) 0.1
+ 𝜆 4 𝑔 (𝑧 − 𝑝) . 0
0 20 40 60 80 100
t
3. Results and Discussion
Figure 3: The optimal control of MEK.
In this part, based on the minimum principle, we will give
the simulation of optimal strategy by using the Runge–Kutta
fourth-order scheme and the steepest gradient method [27]. ×108
10
We choose 100 days as the control time. The efficacy 𝑢
can theoretically lie between 0 and 1, where 0 corresponds 8
to no effectiveness of the MEK and 1 corresponds to full 6
x
effectiveness of the MEK. However, the perfect efficacy of 4
MEK is unlikely to be achieved totally, so we suppose that the 2
maximum effect is 0.98. 0
It is difficult to choose the parameters exactly based on 0 20 40 60 80 100
biological meaning without experiment data, since the global t
stability of E1 means the therapy has no effect when ×108
4
𝛼 (𝐾 + 𝜀) 𝑔𝑢 + 𝑐 3
𝑏<1+ ⋅ . (32)
𝛽𝐾 𝑔𝑢𝑝
y 2
−1 −1
We choose 𝛽 = 0.2 day , 𝜀 = 0.009, 𝛿 = 0.5 day , 𝑟 = 6 1
cells day−1 , 𝛼 = 0.5 day−1 , 𝑝 = 10, 𝑔 = 0.1 day−1 , 𝑐 = 0
0.5 day−1 , 𝐾 = 9 × 108 cells, 𝑏 = 4 cell−1 day−1 , 𝑎11 = 𝑎22 = 1, 0 20 40 60 80 100
𝑏11 = 𝑏22 = 𝑏33 = 10−5 , and 𝑐11 = 8000. t
Even if we use the maximum constant 𝑢 = 0.98, 𝑏 > 1 + Figure 4: State dynamics for uninfected and infected tumor cells
(𝛼(𝐾 + 𝜀)/𝛽𝐾)((𝑔𝑢 + 𝑐)/𝑔𝑢𝑝) = 2.5255 also holds. with different control strategies.
We give and compare two control strategies with the same
initial conditions:

𝑥0 = (6 × 107 , 0, 5 × 104 , 4 × 102 ) . (33) will begin to decrease and keep lower than that of constant
control. As for the infected tumor, it is apparent that more
The optimal control simulation results are shown in Fig- tumor cells are infected with the optimal strategy; this in turn
ure 3. The corresponding state dynamics for uninfected and can help tumor cells keep at a contrarily lower level.
infected tumor cells under the optimal control are shown in
Figure 4 with solid line. 4. Conclusion
We choose constant control 𝑢 = 0.98 to compare with the
optimal control effect; the state dynamics for uninfected and This paper introduces a new mathematical model of tumor
infected tumor cells under the constant control are shown in therapy with oncolytic virus and MEK inhibitor. The stability
Figure 4 with dotted line. of the equilibrium points is analyzed. Because inhibitors
From the simulation, we can see that even if we use the (MEK) can not only lead to greater oncolytic virus infection
maximum constant 𝑢 = 0.98, the tumor cells still increase at into cancer cells, but also cause cell cycle to stop, from
the former stage and then keep stable at some level. But if we theoretical analysis and numerical simulations, we compare
use the optimal control strategy as shown in Figure 3, that is optimal control strategy about the dosage of MEK inhibitor
to say, we need not use the maximum dosage of MEK all the and constant control strategy with the same initial conditions.
time, though the tumor cells increase quickly with the lower Simulations show that the optimal control has better control
dosage of MEK at the beginning stage, about 5 days later, it effect than constant control. But it should be pointed out that
BioMed Research International 7

our model has no cure equilibrium point, so, just from our [12] W. Si and W. Zhang, “Control exponential growth of tumor
model, we could not say that the tumor can be cured only cells with slow spread of oncolytic virus,” Journal of Theoretical
by oncolytic virus therapy. But the optimal control strategy Biology, vol. 367, pp. 111–129, 2015.
can help to prevent the tumor from getting worse and worse. [13] D. Wodarz, “Viruses as antitumor weapons: defining conditions
Some other therapy methods should be combined to cure the for tumor remission,” Cancer Research, vol. 61, no. 8, pp. 3501–
tumor. 3507, 2001.
As we cannot get the exact parameters based on biological [14] B. S. Choudhury and B. Nasipuri, “Efficient virotherapy of
meaning, more work should be done about the modeling and cancer in the presence of immune response,” International
simulations. Journal of Dynamics and Control, vol. 2, no. 3, pp. 314–325, 2014.
[15] Z. Bajzer, T. Carr, K. Josić, S. J. Russell, and D. Dingli, “Mod-
Competing Interests eling of cancer virotherapy with recombinant measles viruses,”
Journal of Theoretical Biology, vol. 252, no. 1, pp. 109–122, 2008.
The authors declare that there are no competing interests [16] A. S. Novozhilov, F. S. Berezovskaya, E. V. Koonin, and G.
regarding the publication of this paper. P. Karev, “Mathematical modeling of tumor therapy with
oncolytic viruses: regimes with complete tumor elimination
within the framework of deterministic models,” Biology Direct,
Acknowledgments vol. 1, article 6, pp. 1–18, 2006.
This paper is supported by 2015 National Traditional Chinese [17] Y. Wang, J. P. Tian, and J. Wei, “Lytic cycle: a defining process in
Medicine Clinical Research Base Business Construction Spe- oncolytic virotherapy,” Applied Mathematical Modelling, vol. 37,
no. 8, pp. 5962–5978, 2013.
cial Topics (JDZX2015299).
[18] J. P. Tian, “The replicability of oncolytic virus: defining con-
ditions in tumor virotherapy,” Mathematical Biosciences and
References Engineering. MBE, vol. 8, no. 3, pp. 841–860, 2011.

[1] L. Zhou, W. W. He, Z. N. Zhu et al., “The clinical research [19] Y. Chen and Y. M. Su, “An improved model of tumor therapy
progress for oncolytic adenovirus targeting cancer therapy,” with oncolytic virus,” Journal of Henan University of Science &
China Biotechnology, vol. 33, no. 12, pp. 105–113, 2013. Technology, vol. 37, no. 4, pp. 92–96, 2016.
[2] N. L. Komarova and D. Wodarz, “ODE models for oncolytic [20] K. A. Rauen, D. Sudilovsky, J. L. Le et al., “Expression of
virus dynamics,” Journal of Theoretical Biology, vol. 263, no. 4, the coxsackie adenovirus receptor in normal prostate and in
pp. 530–543, 2010. primary and metastatic prostate carcinoma: potential relevance
to gene therapy,” Cancer Research, vol. 62, no. 13, pp. 3812–3818,
[3] E. Kelly and S. J. Russell, “History of oncolytic viruses: genesis to 2002.
genetic engineering,” Molecular Therapy, vol. 15, no. 4, pp. 651–
659, 2007. [21] M. D. Lacher, M. I. Tiirikainen, E. F. Saunier et al., “Trans-
forming growth factor-𝛽 receptor inhibition enhances ade-
[4] R. Rodriguez, E. R. Schuur, H. Y. Lim, G. A. Henderson, J. W. noviral infectability of carcinoma cells via up-regulation of
Simons, and D. R. Henderson, “Prostate attenuated replication coxsackie and adenovirus receptor in conjunction with reversal
competent adenovirus (ARCA) CN706: a selective cytotoxic for of epithelial-mesenchymal transition,” Cancer Research, vol. 66,
prostate-specific antigen-positive prostate cancer cells,” Cancer no. 3, pp. 1648–1657, 2006.
Research, vol. 57, no. 13, pp. 2559–2563, 1997.
[22] M. Anders, C. Christian, M. McMahon, F. McCormick, and
[5] I. Ganly, D. Kirn, G. Eckhardt et al., “A phase I study of ONYX-
W. M. Korn, “Inhibition of the Raf/MEK/ERK pathway up-
015, an E1B attenuated adenovirus, administered intratumorally
regulates expression of the coxsackievirus and adenovirus
to patients with recurrent head and neck cancer,” Clinical
receptor in cancer cells,” Cancer Research, vol. 63, no. 9, pp.
Cancer Research, vol. 6, no. 3, pp. 798–806, 2000.
2088–2095, 2003.
[6] R. M. Eager and J. Nemunaitis, “Clinical development direc-
[23] G. Cherubini, T. Petouchoff, M. Grossi, S. Piersanti, E. Cun-
tions in oncolytic viral therapy,” Cancer Gene Therapy, vol. 18,
dari, and I. Saggio, “E1B55K-deleted Adenovirus (ONYX-015)
no. 5, pp. 305–317, 2011.
overrides G1/S and G 2/M checkpoints and causes mitotic
[7] O. G. Donnelly, F. Errington-Mais, R. Prestwich et al., “Recent catastrophe and endoreduplication in p53-proficient normal
clinical experience with oncolytic viruses,” Current Pharmaceu- cells,” Cell Cycle, vol. 5, no. 19, pp. 2244–2252, 2006.
tical Biotechnology, vol. 13, no. 9, pp. 1834–1841, 2012.
[24] N. Bagheri, M. Shiina, D. A. Lauffenburger, and W. M. Korn,
[8] S. J. Russell, K.-W. Peng, and J. C. Bell, “Oncolytic virotherapy,” “A dynamical systems model for combinatorial cancer therapy
Nature Biotechnology, vol. 30, no. 7, pp. 658–670, 2012. enhances oncolytic adenovirus efficacy by MEK-inhibition,”
[9] T. S. Miest and R. Cattaneo, “New viruses for cancer therapy: PLoS Computational Biology, vol. 7, no. 2, Article ID e1001085,
meeting clinical needs,” Nature Reviews Microbiology, vol. 12, 2011.
no. 1, pp. 23–34, 2014. [25] R. Zurakowski and D. Wodarz, “Model-driven approaches
[10] M. R. Patel and R. A. Kratzke, “Oncolytic virus therapy for can- for in vitro combination therapy using ONYX-015 replicating
cer: the first wave of translational clinical trials,” Translational oncolytic adenovirus,” Journal of Theoretical Biology, vol. 245,
Research, vol. 161, no. 4, pp. 355–364, 2013. no. 1, pp. 1–8, 2007.
[11] M. Aghi and R. L. Martuza, “Oncolytic viral therapies—the [26] J. P. LaSalle, The Stability of Dynamical Systems, Regional Con-
clinical experience,” Oncogene, vol. 24, no. 52, pp. 7802–7816, ference Series in Applied Mathematics, SIAM, Philadelphia, Pa,
2005. USA, 1976.
8 BioMed Research International

[27] G. Pachpute and S. P. Chakrabarty, “Dynamics of hepatitis C


under optimal therapy and sampling based analysis,” Commu-
nications in Nonlinear Science and Numerical Simulation, vol. 18,
no. 8, pp. 2202–2212, 2013.
International Journal of

Peptides

Advances in
BioMed
Research International
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Stem Cells
International
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
Virolog y
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
International Journal of
Genomics
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014

Journal of
Nucleic Acids

Zoology
 International Journal of

Hindawi Publishing Corporation Hindawi Publishing Corporation


http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Submit your manuscripts at


http://www.hindawi.com

Journal of The Scientific


Signal Transduction
Hindawi Publishing Corporation
World Journal
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Genetics Anatomy International Journal of Biochemistry Advances in


Research International
Hindawi Publishing Corporation
Research International
Hindawi Publishing Corporation
Microbiology
Hindawi Publishing Corporation
Research International
Hindawi Publishing Corporation
Bioinformatics
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Enzyme International Journal of Molecular Biology Journal of


Archaea
Hindawi Publishing Corporation
Research
Hindawi Publishing Corporation
Evolutionary Biology
Hindawi Publishing Corporation
International
Hindawi Publishing Corporation
Marine Biology
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Вам также может понравиться