ArticlePDF Available

Mathematical modelling of COVID-19 disease dynamics: Interaction between immune system and SARS-CoV-2 within host

Authors:

Abstract

SARS-COV-2 (Coronavirus) viral growth kinetics within-host become a key fact to understand the COVID-19 disease progression and disease severity since the year 2020. Quantitative analysis of the viral dynamics has not yet been able to provide sufficient information on the disease severity in the host. The SARS-CoV-2 dynamics are therefore important to study in the context of immune surveillance by developing a mathematical model. This paper aims to develop such a mathematical model to analyse the interaction between the immune system and SARS-CoV-2 within the host. The model is developed to explore the viral load dynamics within the host by considering the role of natural killer cells and T-cell. Through analytical simplifications, the model is found well-posed and asymptotically stable at disease-free equilibrium. The numerical results demonstrate that the influx of external natural killer (NK) cells alone or integrating with anti-viral therapy plays a vital role in suppressing the SARS-CoV-2 growth within-host. Also, within the host, the virus can not grow if the virus replication rate is below a threshold limit. The developed model will contribute to understanding the disease dynamics and help to establish various potential treatment strategies against COVID-19.
http://www.aimspress.com/journal/Math
AIMS Mathematics, 7(2): 2618–2633.
DOI: 10.3934/math.2022147
Received: 23 September 2021
Accepted: 05 November 2021
Published: 17 November 2021
Research article
Mathematical modelling of COVID-19 disease dynamics: Interaction
between immune system and SARS-CoV-2 within host
S. M. E. K. Chowdhury1, J. T. Chowdhury1, Shams Forruque Ahmed2,, Praveen Agarwal3,4,5,
Irfan Anjum Badruddin6and Sarfaraz Kamangar6
1Department of Mathematics, University of Chittagong, Chattogram 4331, Bangladesh
2Science and Math Program, Asian University for Women, Chattogram 4000, Bangladesh
3Department of Mathematics, Anand International College of Engineering, Jaipur 303012, India
4Nonlinear Dynamics Research Center (NDRC), Ajman University, Ajman AE 346, United Arab
Emirates
5International Center for Basic and Applied Sciences, Jaipur 302029, India
6Mechanical Engineering Department, College of Engineering, King Khalid University, Abha
61421, Saudi Arabia
*Correspondence: Email: shams.ahmed@auw.edu.bd, shams.f.ahmed@gmail.com.
Abstract: SARS-COV-2 (Coronavirus) viral growth kinetics within-host become a key fact to
understand the COVID-19 disease progression and disease severity since the year 2020. Quantitative
analysis of the viral dynamics has not yet been able to provide sucient information on the disease
severity in the host. The SARS-CoV-2 dynamics are therefore important to study in the context
of immune surveillance by developing a mathematical model. This paper aims to develop such a
mathematical model to analyse the interaction between the immune system and SARS-CoV-2 within
the host. The model is developed to explore the viral load dynamics within the host by considering
the role of natural killer cells and T-cell. Through analytical simplifications, the model is found well-
posed and asymptotically stable at disease-free equilibrium. The numerical results demonstrate that the
influx of external natural killer (NK) cells alone or integrating with anti-viral therapy plays a vital role
in suppressing the SARS-CoV-2 growth within-host. Also, within the host, the virus can not grow if the
virus replication rate is below a threshold limit. The developed model will contribute to understanding
the disease dynamics and help to establish various potential treatment strategies against COVID-19.
Keywords: MERS-CoV; SARS-CoV-2; COVID-19; mathematical model; immune system; basic
reproduction number
Mathematics Subject Classification: 34A12, 34A34, 37C75
2619
1. Introduction
Human civilization has been facing dierent infectious pathogen-caused epidemics for a long time.
Severe Acute Respiratory Syndrome (SARS-CoV), a coronavirus class, was first identified in 2003 in
Asia, with 8422 cases and an 11% fatality rate [1]. Another class of coronavirus, Middle East
Respiratory Syndrome (MERS-CoV) was reported with approximately 1572 human cases in 2012 in
Arabian Peninsula [2]. SARS-CoV-2, the new class of novel coronavirus has now become a
serious global threat due to its first advent in Wuhan, Hubei province, China in the year 2019 [3]. In
spite of taking dierent social interventions and pharmaceutical measures by dierent countries,
SARS-CoV-2 spreads over the globe within a very short time and tossed the biggest challenge ever to
global health [4]. Every morning the world gets introduced with record-breaking cases in both
infection and death caused by SARS-CoV-2. To pull down the race of the SARS-CoV-2 pandemic,
researchers are attempting their best to discover strategies to explore the virus-protein structure of the
SARS-CoV-2 [5], phylodynamics of the SARS-CoV-2 [6], and build up the antibody against
SARS-CoV-2 infection by developing vaccines [7].
Mathematical models are often useful in characterizing the infection dynamics and forecast disease
severity. Many pathogens were studied using a simple model including target cell, infected cell, and a
virus. The target cell limited model was used by many researchers to study the in-host dynamics of
HIV [8–10], Hepatitis [11], Ebola [12, 13], and Influenza viruses [14, 15]. Many mathematical
models [16–19] focused on the transmission dynamics of the SARS-CoV-2 virus and involved
dierent pharmacological, non-pharmacological interventions to reduce the spread of the COVID-19
disease. However, only a few models explained the replication cycle of SARS-CoV-2 within the host
and interactions between this virus and the host immune system [20–22]. A brief study [22] on
SARS-CoV-2 virus dynamics within-host discussed the target cell model with an eclipse phase, and
secondary infection in presence of lymphocytes. The study found a rapid rise in viral load to reach the
peak at the initial stage pointing to a plateau phase caused by lymphocytes. Also, an adaptive immune
response results in decreasing the viral load at the last stage. Hernandez-Vargas et al. [21] investigated
the interaction between the SARS-CoV-2 virus and the immune system within-host. Though the
authors discussed a couple of models explaining the SARS-CoV-2 dynamic within the host, the
immune cell (T-cell) response model performed better than others to fit the data indicating a slow
immuno-response reaching the peak in 5–10 days after symptom onset.
Some researchers [23–27] studied the COVID-19 disease severity and mortality based on the data
collected from the categorized patients. The viral load of SARS-CoV-2 in a group of patients with
divergent SARS-CoV-2 serotypes was measured in the study of Fajnzylber et al. [23]. The authors
stated that the spectrum of SARS-CoV-2 viremia is linked to inflammation markers and disease
severity, such as elevated C-reactive protein (CRP), IL-6, and a low lymphocyte count. Through a
virological study, Wolfel et al. [26] found sequence-distinct virus replication in the throat, lung tissue,
and upper respiratory. The pulmonary cell anity with the COVID-19 disease intensity was identified
by computer tomography (CT-scan) and radio-graph of the patients’ chest [25]. Later, Li et al. [24]
used chest radiograph scores to determine the lung cell destruction due to the SARS-CoV-2 virus
infection. They proposed a model describing the kinetics of the interactions between the virus
particles and the lung epithelial cells within-host. Using the chest radiograph data, the authors
determined the model parameters and found the basic reproduction number around 3.79 for the
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2620
SARS-CoV-2 growth within the host.
Against the SARS-CoV-2 virus infection, no therapeutic and prophylactic medications are yet
approved. Patients with acute symptoms are administered oxygen therapy, ventilation, antibiotics, and
corticosteroids to support the respiratory functions and to control inflammatory reactions. These
instructions allow time for activation of the adaptive immune response [28, 29]. Dierent possible
medical schemes include the use of antiviral drugs inhibiting the replications of the virus cycle.
Repurposing existing anti-viral drugs can be an eective drug strategy against the SARS-CoV-2 that
could reduce the time and limit the cost compared to the de novo drug discovery. In a study of drug
re-purpose designed for SARS-CoV-2, Zhou et al. [30] proposed sixteen repurposable drugs based on
network proximity analyses (NPA) of drug targets within the human host. The authors also proposed
three prospective drug combinations such as mercaptopurine and melatonin, sirolimus and
dactinomycin, toremifene and emodin, against the SARS-CoV-2 virus infection.
In any infections within the host, natural killer (NK) cells serve as first-line protection. NK cell
and T-cell are necessary for a strong confrontation and clearance of the virus from the host. Among
various anti-viral activities, these cells are accountable for directly killing the target cells and the
release of immunomodulatory cytokines [31]. After conducting a thorough literature review, it is
found that only a few research [21, 22, 24] have been carried out reporting the SARS-CoV-2 viral
growth in the host under the impact of T-cells. However, no studies have been found which analyses
the interaction between the immune system and SARS-CoV-2 within the host considering T -cells and
NK-cells together.
A deterministic mathematical model based on physiologically plausible assumptions performs
better in forecasting disease severity over time in the absence of relevant experimental data. A
deterministic model reflecting the short-term expected antiviral eect during SARS-CoV-2
development within the host is required in addition to forecasting illness severity. As a result,
developing a mathematical model under the eect of both T -cells and NK-cells becomes essential for
qualitative analysis of the viral load within the host. To comprehend the SARS-CoV-2 growth
dynamics within the host under immune surveillance, a mathematical model is therefore developed in
the present study by analysing the interaction between the immune system and SARS-CoV-2 within
the host integrating the function of NK cells and T-cells. In essence, the model looks into ways to
control the pace of SARS-CoV-2 viral replication within a host, because a quicker viral replication
rate inside the host is directly related to community transmission. This research will help physicians,
scientists, and policymakers to understand how patients’ immunity interact against COVID-19
disease, as well as to devise disease treatment strategies.
2. Model formulation
The mathematical framework of the proposed model is based on the interaction between virus
particles and dierent lymphocytes within-host including the most common lymphocytes, NK cells,
and T-cells. T-cells are in control of both adaptive and innate immune responses while NK cells,
which are part of the innate immune system, are the first immune eectors against virus infection
before any specific immunity arrives. Furthermore, in the absence of specific antibodies, the NK cell
recognizes and kills the distressed cell [32].
The target cell of the SARS-CoV-2 virus in the respiratory system is not completely known. Though
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2621
there is a dispute in current literature about the primary compartments of SARS-CoV-2 infection, it is
usually believed that the primary infection succeeds in respiratory epithelial cells [33]. Based on the
results came out from the studies [33–35], epithelial cell in the respiratory system has been considered
in the present model as a primary target of the SARS-CoV-2 virus. The infected epithelial cells are
assumed to be capable of producing new virus particles at a constant rate. Taking the interactions of
the virus-immune system and virus-epithelial cells, the proposed model is developed as follows:
dE
dt =a1a2Ea3EV,
dEi
dt =a3EV b1Ei,
dV
dt =c1Eic2Vc3V N c4V L,
dN
dt =d1d2N,
dL
dt =e1LV
(n+V)e2L+e3,
(2.1)
where E,Ei,V,N,Lrepresent the number of susceptible epithelial cells, infected epithelial cells, viral
load, natural killer cells, and T-lymphocytes respectively; a3is the infection rate of the virus; c1is
the production rate; and the terms a2,b1,c2,d2, and e2, are the natural death rates corresponding to
E,Ei,V,N,and Lrespectively.
The regeneration of the epithelial cells is considered as a1=a2×E(0) [21], where a2is the natural
death rate of the epithelial cells and E(0) represents the number of initial epithelial cells. The term
a3EV in the first equation denotes virus particles infecting a healthy epithelial cell at an infection rate
of a3. The growth of infected epithelial cells is denoted by a3EV in the second equation. The first
term c1Eiin the third equation implies the reproduction of new SARS-CoV-2 virus from an infected
cell at a rate c1. Also, c3VN and c4V L represent the local interaction dynamics of the virus (V) with
natural killer cells (N) and T- lymphocytes (L). Due to these interactions, virus particles are reduced
at the rates c3and c4respectively. Similarly, d1implies the constant external source of the natural killer
cells as expressed in the fourth equation of the model. The term e3is the natural recruitment rate of
T-lymphocytes and e1LV/(n+V) is the proliferation in response to the presence of virus particles. This
functional proliferation form is employed here as the model assumes the recruitment of T-lymphocytes
due to the signals to release dierent cytokines (prompting molecule that regulates and mediates the
immune system). The half-saturation constant nis theT-lymphocytes proliferation by a virus which is
estimated as 1.26 ×105for a sigmoidal function to the best data fit [21].
The model is considered under the initial conditions E(t)0,Ei(t)0,V(t)0,N(t)0 and
L(t)0 with non-negative value of the model parameters. It is obvious that the right-hand parts of
the system of Eq (2.1) are continuous and satisfy the Lipschitz condition [36]. The uniqueness and
existence criterion of the ordinary dierential equation confirms that model (2.1) has a unique solution
with the non-negative initial conditions [36]. The first equation of the model (2.1), is considered to
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2622
explore the non-negativity property of the solution curves as follows:
dE
dt +a2E0.(2.2)
The solution trajectory of inequality (2.2) is obtained as
E(t)E(0)ea2t.(2.3)
The inequality (2.3) gives E(t)0 for t→ ∞ which indicates that for every t[0,) the solution
trajectory E(t) will be entirely in the positive orthant for the given non-negative initial condition and
parameter values. In a similar approach, it is easy to show Ei(t)0,V(t)0,N(t)0,L(t)0 for
t→ ∞.
The first equation of the model (2.1) also reveals that
E(t)a1
a2
+ E(0)a1
a2!ea2t,(2.4)
i.e., for long term behaviour t→ ∞,E(t) is bounded by a1
a2= Π1(say)R.
Moreover, the solution trajectories of Ei(t),V(t), and L(t) of the system (2.1) give
Ei(t)=p
b1
+ Ei(0) p(0)
b1!eb1t;p=a3EV,
V(t)=q1
q2
+ Ei(0)q1(0)
q2(0) !eq2t;q1=c1Ei,q2=c2+c3N+c4L,
L(t)=r1
r2
+ L(0)r1(0)
r2!er2t;r1=e1V
(n+V)+e2;r2=e3,
(2.5)
where p(0),q1(0),q2(0),and r1(0) represent the initial values of the corresponding functions in terms of
state variables E,V,Nand L. Since Eand Vremain constant in the second equation of the model (2.1),
the supremum of Ei(t) is given by
lim
t→ ∞ sup Ei(t)=p
b1
Π2(say)R.
Defining a function F(t)=E(t)+Ei(t)+V(t)+N(t)+L(t) and its derivative along the trajectories
yields,
dF
dt =dE
dt +dEi
dt +dV
dt +dN
dt +dL
dt
=βa2Eb1Ei+c1Eic2Vd2Ne2Lc3VN c4V L +µ
(a2Eb1Eic2Vd2Ne2L)+β+µ+c1Π2
≤ −hF +H,
(2.6)
where H=β+µ+c1Π2,h=min {a2,b1,c2,d2,e2},β=(a1+d1+e3), and µ=e1LV/(n+V).
Inequality (2.6) reveals that F(t) is bounded by H/h, whereas E(t) and Ei(t) have bounds Π1and
Π2respectively. Therefore, N(t),V(t) and L(t) are eventually bounded with an other bound ΠR.
Denoting M=max {Π,Π1,Π2}, for suciently large time scale t, it reveals that E(t)M,Ei(t)M,
V(t)M,N(t)Mand L(t)M.
Therefore, under the non-negative initial conditions with the positive parameter values, the solution
space of the system (2.1) gives
Ω = (E,Ei,V,N,L)R5; 0 E(t),Ei(t),V(t),N(t),L(t)M.
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2623
3. Model analysis
Evaluation of the basic reproduction number R0at equilibrium points is the simplest way to
investigate the disease dynamics described by a mathematical model. Setting the right-hand side of
the model equations to zero, the first equilibrium point η(E0,Ei
0,V0,N0,L0) in absence of the
SARS-CoV-2 virus of the model (2.1) is obtained as
(E0,Ei
0,V0,N0,L0)=(a1
a2
,0,0,d1
d2
,e3
e2
).(3.1)
Further, at endemic equilibrium, none of the state variables is identically zero in presence of the
virus within-host. In that case, the second equilibrium point η(E
0,Ei
0,V
0,N
0,L
0) for model (2.1) is
given by:
E=a1
a2+a3V
Ei=a1a3V
(a2+a3V)b1
V=c1Ei
c2+c3N+c4L
N=d1
d2
L=e3(n+V)
e2(n+V)e1V
(3.2)
The infection ability of a pathogen within-host is quantified by determining R0for the corresponding
model [37]. This number is employed to evaluate the average secondary infections resulting from an
infected cell. Mathematically, R0<1 implies that an infected cell (pathogen) might infect less than one
cell, resulting in an invasion of the disease from the host. In contrast, R0>1 suggests a suppression of
the host cells resulting in the progress of the disease within-host.
The concept of the next-generation matrix (NG M) [38] is used to determine the basic reproduction
number R0. The system (2.1) has three infected states E,Eiand V, and two uninfected states, Nand L.
Linearising the Eqs (1)–(3) of the system (2.1) as a subsystem of infection, the following transmission
matrix (T) and transition matrix (Q) are obtained:
T=
0 0 a3a1
a2
0 0 a3a1
a2
0 0 0
and Q=
a20 0
0b10
0c1α
.
where α=c2+c3d1
d2
+c4e3
e2
.
Since the first two columns of the transmission matrix (T) are zero, the next generation matrix [38]
will be K=ETT Q1Ewhere the axillary matrix Eis given by:
E=
1 0
0 1
0 0
.
Using the next-generation matrix K, the R0of the model (2.1) can be written as
R0=a1a3c1
a2b1α.(3.3)
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2624
3.1. Steady state analysis
The stability of model (2.1) at the virus-free equilibrium point η, is determined by the following
lemma:
Lemma 1. The disease-free equilibrium of model (2.1) is locally asymptotically stable for R0<1 and
unstable for R0>1.
Proof. The Jacobian matrix of the model at disease-free equilibrium point ηcan be written as
JE0=
a20a1a3
a2
0 0
0b1
a1a3
a2
0 0
0c1α0 0
0 0 0d20
0 0 e1e3
ne2
0e2
.
The characteristic equation of this Jacobean matrix is given by
(λa2) (λd2) (λe2)λ2+k1λ+k2=0,(3.4)
where k1=(b1+α)>0 and k2=b1α(a1a3c1)/(a2).
It is clear that the first three eigen-values obtained from Eq (3.4) are negative as a2>0, d2>0 and
e2>0. As per the Descarte’s sign rule [39], the rest of the eigen-values are negative for k1>0 and
k2>0. Rewriting the condition k2>0 in R0, it gives k2=1R0>0, that means, k2>0 if only if
R0<1. Thus all of the eigenvalues are negative. On the contrary, k2becomes negative if R0>1 which
provides at least one of the eigenvalues is positive. Hence, Lemma 1 follows.
The analytical computation of the characteristic equation at endemic equilibrium point ηproduced
a large expression and therefore, the determination of the sign of eigenvalues at ηbecomes very
complicated. Rather, the qualitative behaviour of the solution trajectories at ηwas investigated through
a series of numerical explorations.
4. Numerical simulation and discussion
A series of numerical simulations were performed using Python (V. 3.7) SciPy.integrate package
and odeint library to solve the ordinary non-linear dierential equations of the present model. The
model equations comprised 14 parameters and assumed 5 initial conditions. The parameter values were
estimated based on some homeostatic process, and available statistical data of SARS-CoV-2 infected
patients [21, 24]. Considering the physical significance of the parameters, numerical investigations
corresponding to the eects of external NK cell influx, anti-viral therapy, and their combined eect over
the SARS-CoV-2 growth projection have been analyzed under immune surveillance in section 4.1–4.3
respectively.
The initial values for healthy and infected epithelial cells, SARS-CoV-2 virus, and T-cell are based
on COVID-19 patient data analysis [21, 24], which may provide realistic results for numerical
simulations. The model assumes the initial conditions as follows: Ei(0) =2.59 (score) [24],
E(0) =25 Ei(0) =22.41 [24]), V(0) =0.061 (score) [24], L(0) =106 [21], N(0) =105 [40]. The
virus clearance rate c2was fixed and characterized as a process that is not linked with the immune
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2625
system. The parameter a1was estimated as [24]: a1=a2×E(0) =0.02241, and the other simulation
parameters are tabulated in Table 1. It is noted that the parameter values for epithelial cells and
SARS-CoV-2 were chosen from published literature based on chest radiograph score which is an
approximation, not actual infected data for pulmonary cells [24].
Table 1. Model parameters used in the simulations.
Parameters Description Value Reference
a1Constant regeneration of E.C 0.0224 (estimated) Li et al. [24]
a2Death rate of E.C (day1) 103
a3Infection rate of E.C by virus 0.55
(day1score1))
b1Death rate of infected E.C.(day1) 0.11
c1Production rate of virus from per 0.24
infected E.C.(day1)
c2Death rate of virus (day1) 5.36
c4Killing rate of virus by T-lymp. (day1) 1.89 ×106Hernandez et al. [21]
e1Proliferation rate of T-lymp. 0.194
e2Death rate of T-lymp. (day1) 0.1
e3Constant regeneration of T-lymp. 105
nHalf saturation constant of T-lymp. 1.26 ×105
c3Killing rate of virus by N.K (day1) 1.5×106(assumed) Present study
d1External influx of N.K. cell (day1cells) 3.2×103Kuznetsov et al. [41]
d2Death rate of N.K. cell (day1cells) 4.12 ×102de Pillis et al. [42]
4.1. External influx of natural killer (NK) cell
The impact of NK cell influx from an external source on the disease progression is assessed to
quantify the viral dynamics within-host. The immunity system works to clear the virus from the day
of symptom onset. However, the immunity strength depends on dierent factors including age and
co-morbidness of the patients. Figure 1 shows the eect of external NK cell influx over viral load.
As seen in the figure, the viral load increase very fast at the beginning of the infection due to slow
immune response and reaches the peak of 0.35 (chest score) on day 10 after symptom onset where
the viral load is expressed as a chest radiograph score. A small increase in the external influx of NK
cell as d1=d1+j1000 (0 j5) results in a reduction of viral load about 0.3 to 1.5% in
each loop iteration. The maximum viral load (chest score) for the corresponding loop iterations are
v=0.3501,v=0.3492,v=0.3475,v=0.3450,and v=0.3417 respectively. Because the viral
loads corresponding to d=3200,d=4200,and d=6200 are so similar, Figure 1 contains plots
corresponding to d=3200, d=9200, and d=13200, which illustrate the intensity of viral load relative to
the change in external NK cell influx.
Figure 1(d) shows the basic reproduction number R0which gradually decreases subject to the
external influx of the NK cell. The large influx of NK cells strengthens the immune system, helps to
restrict viral proliferation. Diminishing R0from 3.65 to 3.33 (approx.) implies the invasion of viral
replication within the host during the disease. A simulation was also run with higher initial values of
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2626
the SARS-CoV-2 virus to investigate the initial viral load over the disease progression. A significant
rise was observed in the chest radiograph score (viral load) which indicates a symptom severity in
patients.
Figure 1. Viral load (V) and reproduction number (R0) relative to NK cell influx (d1).
4.2. Anti-viral therapy
The role of possible anti-viral therapy can reduce the SARS-CoV-2 virus replication rate. A high
replication rate c1of the SARS-CoV-2 implies a rapid invasion of the susceptible epithelial lung
cells (E). The SARS-CoV-2 replication rate on viral load dynamics is portrayed in Figure 2. The
maximum viral load of 0.35 (score) occurs in Figure 2(a), while the minimum viral load of 0.003
(score) ensures in Figure 2(e). For each loop decrement (c1=c1j0.02) in the value of c1, viral
load was decreased by approximately 14 to 31%. For a lower replication rate, SARS-CoV-2 growth
dynamics are found less steep than for a higher replication rate (Figure 2), which indicates that an
anti-viral therapy can more eectively combat the virus. In addition, the SARS-CoV-2 virus cannot
grow within-host under immune surveillance if the replication rate is less than or equal to 0.04 as seen
in Figure 2(e). The threshold value of c1was found 0.04 for the defined loop iterations. The actual
threshold value of c1for model (2.1) was determined as 0.09 implying that SARS-CoV-2 cannot grow
in the host with a replication rate less than 0.09.
Figure 2(f) depicts the graph of basic reproduction number R0relative to the SARS-CoV-2 virus
replication rate. The number R0shows a positive linear trend with virus replication rate c1. A higher
replication rate clearly contributes to an increased symptom and disease prevalence in patients,
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2627
resulting in community disease transmission and lung cell destruction. This finding suggests that
prescribing appropriate antiviral drugs to a patient sooner may reduce the symptom.
Figure 2. Viral load (V) and reproduction number (R0) relative to SARS-CoV-2 replication
rate (c1).
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2628
Figure 3. Viral load (V) and reproduction number (R0) relative to the combined eect of (c1)
and (d1).
4.3. Combination of anti-viral therapy and influx of external NK cell
Combinations of dierent therapeutic treatments have become popular in many viral disease
management protocols. For instance, monoclonal antibodies with other anti-viral medicine showed an
important role in Ebola virus infection [12]. In this section, the combined impact of anti-viral therapy
and the eternal influx of NK over the SARS-CoV-2 virus growth projection has been demonstrated.
The combined eect of possible anti-viral drugs and influx of external NK cells is shown in Figure 3.
The influx of NK cells strengthens the patient’s immune system, while antiviral therapy reduces virus
replication. It is found that administering this combined therapy results in a quick reduction of viral
load from the initial peak. Such a substantial reduction in viral load causes symptom relief and
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2629
disease invasion in the patient.
The basic reproduction number plot Figure 3(f) shows a sharp fall due to the combined eect of
NK cell influx and anti-viral drug. This reflection implies that the combined eect is more eective in
suppressing the viral load within the host under immune surveillance.
5. Sensitivity analysis
The sensitivity of a specific parameter identifies how a small change in numeric value causes a large
impact on the model output. This analysis may assist to establish the possible treatment protocol for
the SARS-CoV-2 disease. The sensitivity indices of the basic reproduction number R0relative to the
parameters is defined as [12]:
Θa1=r
R0
Ro
r,
where ris the model parameter. The estimated sensitivity indices of the model (2.1) are summarized
in Table 2.
Table 2. Sensitivity of R0with respect to the parameters.
Sensitivity index Index value
Θa11
Θa31
Θc11
Θd20.01581549
Θe20.25656672
Θa21
Θb11
Θc20.72761779
Θc30.01581549
Θd10.01581549
Θc40.25656672
Θe30.25656672
From the sensitivity indices, it is obvious that a1,a3,c1,d2, and e2have a positive impact on R0
while the rest of the parameters shows a negative impact. The negative impact implies the invasion of
viral load whereas the positive impact indicates a progression of virus load within-host. The indices
Θa1,Θa3, and Θc1are being equal to 1 (most sensitive) reveals that a 10% increase in the corresponding
parameter values causes a 10% growth projection in R0. Similarly, a2and b1show reverse correlation
to R0, i.e., 10% increase in a2and b1(least sensitive) results in decreasing the value of R0by 10%.
These results fairly agree with the numerical results (Figures 1 and 2). The constant regeneration a1
shows positive sensitivity to R0as in Table 2. It means that a host with higher cells of pulmonary
epithelial is more likely to become infected, and provides clarification why especially babies having
lower epithelial cells are not most expected to get infected.
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2630
6. Conclusions
A mathematical model was developed to explore the interaction between the immune system and
SARS-CoV-2 within the host by integrating the function of NK cells and T-cells. A series of
simulations were performed to investigate the qualitative behaviour of the model. The numerical
results complied with the analytical results of the model where the solution trajectories were found
bounded in the positive quadrant. A sensitivity analysis was also carried out for basic reproduction
number R0to analyse the impact of the parameters on the disease progression. The model is found to
be advantageous in assessing the antiviral medication ecacy and NK cell influx into the host body,
and with the value of R0across the time of infection period, the model can be used in forecasting
disease severity. The numerical analysis showed that the use of anti-viral therapy and influx of NK
cells causes a fall in the value of R0. In addition, SARS-CoV-2 cannot grow within-host if the virus
replication rate (c1) is under a threshold limit of 0.09. The combined use of anti-viral therapy and
influx of external NK cells together played an important role to control the SARS-CoV-2 viral growth
within-host. If the proposed anti-viral combinations are capable to control or reduce the SARS-CoV-2
replication rate, it may be a good attempt to use those from the very beginning of the disease. The
present model can be used further to investigate the role of antibody development and cross-reaction.
Furthermore, the present model can be extended to study the innate immune response to dierent
SARS-CoV-2 mutations. Each new variety must have its own replication rate, which has an impact on
the severity of the disease and its propagation in the population. This idea would help us to develop a
new mathematical model in the future study.
Acknowledgements
The authors extend their appreciation to the Deanship of Scientific Research at King Khalid
University for funding this work through the research groups program under grant number
RGP.1/327/42.
Conflict of interest
The authors declare that they have no conflict of interest.
References
1. A. Sharma, S. Tiwari, M. K. Deb, J. L. Marty, Severe acute respiratory syndrome coronavirus-2
(SARS-CoV-2): A global pandemic and treatment strategies, Int. J. Antimicrob. Ag.,56 (2020),
106054. doi: 10.1016/j.ijantimicag.2020.106054.
2. N. Ramadan, H. Shaib, Middle East respiratory syndrome coronavirus (MERS-CoV): A review,
Germs,9(2019), 35–42. doi: 10.18683/germs.2019.1155.
3. H. Ouassou, L. Kharchoufa, M. Bouhrim, N. E. Daoudi, H. Imtara, N. Bencheikh, et al., The
pathogenesis of coronavirus disease 2019 (COVID-19): Evaluation and prevention, J. Immunol.
Res.,2020 (2020), 1357983. doi: 10.1155/2020/1357983.
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2631
4. M. Mofijur, I. M. R. Fattah, M. A. Alam, A. B. M. S. Islam, H. C. Ong, S. M. A. Rahman,
et al., Impact of COVID-19 on the social, economic, environmental and energy domains:
Lessons learnt from a global pandemic, Sustain. Prod. Consump.,26 (2021), 343–359. doi:
10.1016/j.spc.2020.10.016.
5. S. F. Ahmed, A. A. Quadeer, M. R. McKay, Preliminary identification of potential vaccine targets
for the COVID-19 coronavirus (SARS-CoV-2) based on SARS-CoV immunological studies,
Viruses,12 (2020), 254. doi: 10.3390/v12030254.
6. Q. Nie, X. G. Li, W. Chen, D. H. Liu, Y. Y. Chen, H. T. Li, et al., Phylogenetic and phylodynamic
analyses of SARS-CoV-2, Virus Res.,287 (2020), 198098. doi: 10.1016/j.virusres.2020.198098,
7. F. Krammer, SARS-CoV-2 vaccines in development, Nature,586 (2020), 516–527. doi:
10.1038/s41586-020-2798-3.
8. X. L. Lai, X. F. Zou, Modeling cell-to-cell spread of HIV-1 with logistic target cell growth, J. Math.
Anal. Appl.,426 (2015), 563–584. doi: 10.1016/j.jmaa.2014.10.086.
9. F. Li, J. L. Wang, Analysis of an HIV infection model with logistic target-cell growth and cell-to-
cell transmission, Chaos, Soliton. Fract.,81 (2015), 136–145. doi: 10.1016/j.chaos.2015.09.003.
10. J. L. Wang, J. Y. Lang, X. F. Zou, Analysis of an age structured HIV infection model with virus-
to-cell infection and cell-to-cell transmission, Nonlinear Anal.-Real,34 (2017), 75–96.
11. F. Graw, A. S. Perelson, Modeling viral spread, Annu. Rev. Virol.,3(2016), 555–572. doi:
10.1146/annurev-virology-110615-042249.
12. V. K. Nguyen, E. A. Hernandez-Vargas, Windows of opportunity for Ebola virus infection
treatment and vaccination, Sci. Rep.,7(2017), 8975. doi: 10.1038/s41598-017-08884-0.
13. V. K. Nguyen, S. C. Binder, A. Boianelli, M. Meyer-Hermann, E. A. Hernandez-Vargas, Ebola
virus infection modelling and identifiability problems, Front. Microbiol.,6(2015), 257. doi:
10.3389/fmicb.2015.00257.
14. C. Hadjichrysanthou, E. Cau¨
et, E. Lawrence, C. Vegvari, F. de Wolf, R. M. Anderson,
Understanding the within-host dynamics of influenza A virus: From theory to clinical implications,
J. R. Soc. Interface,13 (2016), 20160289. doi: 10.1098/rsif.2016.0289.
15. E. A. Hernandez-Vargas, E. Wilk, L. Canini, F. R. Toapanta, S. C. Binder, A. Uvarovskii, et
al., Eects of aging on influenza virus infection dynamics, J. Virol.,88 (2014), 4123–4131. doi:
10.1128/JVI.03644-13.
16. R. M. Anderson, H. Heesterbeek, D. Klinkenberg, T. D. Hollingsworth, How will country-based
mitigation measures influence the course of the COVID-19 epidemic? The lancet,395 (2020),
931–934. doi: 10.1016/S0140-6736(20)30567-5.
17. N. G. D. Davies, A. J. Kucharski, R. M. Eggo, A. Gimma, W. J. Edmunds, T. Jombart, et al., Eects
of non-pharmaceutical interventions on COVID-19 cases, deaths, and demand for hospital services
in the UK: A modelling study, The Lancet Public Health,5(2020), 375–385. doi: 10.1016/S2468-
2667(20)30133-X.
18. L. Ferretti, C. Wymant, M. Kendall, L. Zhao, A. Nurtay, L. Abeler-D¨
orner, et al., Quantifying
SARS-CoV-2 transmission suggests epidemic control with digital contact tracing, Science,368
(2020), eabb6936. doi: 10.1126/science.abb6936.
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2632
19. Y. N. Kyrychko, K. B. Blyuss, I. Brovchenko, Mathematical modelling of the dynamics and
containment of COVID-19 in Ukraine, Sci. Rep.,10 (2020), 19662. doi: 10.1038/s41598-020-
76710-1.
20. S. Q. Du, W. M. Yuan, Mathematical modeling of interaction between innate and adaptive immune
responses in COVID-19 and implications for viral pathogenesis, J. Med. Virol.,92 (2020), 1615–
1628. doi: 10.1002/jmv.25866.
21. E. A. Hernandez-Vargas, J. X. Velasco-Hernandez, In-host mathematical modelling of COVID-19
in humans, Annu. Rev. Control,50 (2020), 448–456. doi: 10.1016/j.arcontrol.2020.09.006.
22. S. P. Wang, Y. Pan, Q. Y. Wang, H. Y. Miao, A. N. Brown, L. B. Rong, Modeling the viral dynamics
of SARS-CoV-2 infection, Math. Biosci.,328 (2020), 108438.
23. J. Fajnzylber, J. Regan, K. Coxen, H. Corry, C. Wong, A. Rosenthal, et al., SARS-CoV-2 viral load
is associated with increased disease severity and mortality, Nature Commun.,11 (2020), 5493. doi:
10.1038/s41467-020-19057-5.
24. C. T. Li, J. H. Xu, J. W. Liu, Y. C. Zhou, The within-host viral kinetics of SARS-CoV-2, Math.
Biosci. Eng.,17 (2020), 2853–2861. doi: 10.3934/mbe.2020159.
25. F. Pan, T. H. Ye, P. Sun, S. Gui, B. Liang, L. L. Li, et al., Time course of lung changes on chest
CT during recovery from 2019 novel coronavirus (COVID-19) pneumonia, Radiology,295 (2020),
715–721. doi: 10.1148/radiol.2020200370.
26. R. W¨
olfel, V. M. Corman, W. Guggemos, M. Seilmaier, S. Zange, M. A. M¨
uller, et al.,
Virological assessment of hospitalized patients with COVID-2019, Nature,581 (2020), 465–469.
doi: 10.1038/s41586-020-2196-x.
27. L. Zou, F. Ruan, M. Huang, L. Liang, H. Huang, Z. Hong, et al., SARS-CoV-2 viral load in
upper respiratory specimens of infected patients, N. Engl. J. Med.,382 (2020), 1177–1179. doi:
10.1056/NEJMc2001737.
28. W. J. Guan, Z. Y. Ni, Y. Hu, W. H. Liang, C. Q. Ou, J. X. He, et al., Clinical characteristics of
coronavirus disease 2019 in China, N. Engl. J. med.,382 (2020), 1708–1720.
29. X. B. Yang, Y. Yu, J. Q. Xu, H. Q. Shu, J. A. Xia, H. Liu, et al., Clinical course and outcomes
of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: A single-centered,
retrospective, observational study, The Lancet Resp. Med.,8(2020), 475–481. doi: 10.1016/S2213-
2600(20)30079-5.
30. Y. D. Zhou, Y. Hou, J. Y. Shen, Y. Huang, W. Martin, F. X. Cheng, Network-based drug repurposing
for novel coronavirus 2019-nCoV/SARS-CoV-2, Cell Discov.,6(2020), 14. doi: 10.1038/s41421-
020-0153-3.
31. K. Frank, S. Paust, Dynamic natural killer cell and T cell responses to influenza infection, Front.
Cell. Infect. Microbiol.,10 (2020), 425. doi: 10.3389/fcimb.2020.00425.
32. I. Gutcher, B. Becher, APC-derived cytokines and T cell polarization in autoimmune inflammation,
J. Clin. Invest.,117 (2007), 1119–1127. doi: 10.1172/JCI31720.
33. Z. Varga, A. J. Flammer, P. Steiger, M. Haberecker, R. Andermatt, A. S. Zinkernagel, et al.,
Endothelial cell infection and endotheliitis in COVID-19, The Lancet,395 (2020), 1417–1418.
doi: 10.1016/S0140-6736(20)30937-5.
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
2633
34. G. M. Kuster, O. Pfister, T. Burkard, Q. Zhou, R. Twerenbold, P. Haaf, et al., SARS-CoV2: Should
inhibitors of the renin–angiotensin system be withdrawn in patients with COVID-19? Eur. Heart
J.,41 (2020), 1801–1803. doi: 10.1093/eurheartj/ehaa235.
35. Z. Xu, L. Shi, Y. Wang, J. Zhang, L. Huang, C. Zhang, et al., Pathological findings of COVID-19
associated with acute respiratory distress syndrome, The Lancet Resp. Med.,8(2020), 420–422.
doi: 10.1016/S2213-2600(20)30076-X.
36. W. E. Boyce, R. C. Diprima, Elementary dierential equations and boundary value
problems, 9 Eds. Available from: https://s2pnd-matematika.fkip.unpatti.ac.id/wp-
content/uploads/2019/03/Elementary-Dirential-Aquation-and-Boundary-Value-Problem-Boyce-
DiPrima.pdf.
37. J. M. Heernan, R. J. Smith, L. M. Wahl, Perspectives on the basic reproductive ratio, J. R. Soc.
Interface,2(2005), 281–293. doi: 10.1098/rsif.2005.0042.
38. O. Diekmann, J. A. P. Heesterbeek, M. G. Roberts, The construction of next-generation
matrices for compartmental epidemic models, J. R. Soc. Interface,7(2010), 873–885. doi:
10.1098/rsif.2009.0386.
39. V. P. Kostov, B. Shapiro, New aspects of descartes’ rule of signs, In: Polynomials-theory and
application, IntechOpen, 2018.
40. K. J. Mahasa, R. Ouifki, A. Eladdadi, L. de Pillis, Mathematical model of tumor–immune
surveillance, J. Theor. Biol.,404 (2016), 312–330. doi: 10.1016/j.jtbi.2016.06.012.
41. V. A. Kuznetsov, I. A. Makalkin, M. A. Taylor, A. S. Perelson, Nonlinear dynamics of
immunogenic tumors: parameter estimation and global bifurcation analysis, B. Math. Biol.,56
(1994), 295–321. doi: 10.1016/S0092-8240(05)80260-5.
42. L. G. de Pillis, W. Gu, A. F. Radunskaya, Mixed immunotherapy and chemotherapy of tumors:
modeling, applications and biological interpretations, J. Theor. Biol.,238 (2006), 841–862. doi:
10.1016/j.jtbi.2005.06.037.
c
2022 the Author(s), licensee AIMS Press. This
is an open access article distributed under the
terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0)
AIMS Mathematics Volume 7, Issue 2, 2618–2633.
... The full within-host dynamics is given as: Production rate of neutrophils in I C 3.2 × 10 2 cell day −1 Estimated from [13] π2 ...
... Production rate of neutrophils in I M 3.2 × 10 2 cell day −1 Estimated from [13] π3 ...
... Production rate of neutrophils in I CM 3.2 × 10 2 cell day −1 Estimated from [13] Remark 2.2.1. In the within-host scale, we consider three distinct populations of uninfected cells denoted as E 1 , E 3 , and E 5 . ...
Article
Full-text available
Methicillin-Resistant Staphylococcus Aureus (MRSA) infection can occur alongside or following COVID-19, which is a concern in healthcare settings. The effectiveness of antiviral treatments for COVID-19 depends on a functioning immune response, but antibiotics used for bacterial infections like MRSA can disrupt the immune response and reduce the effectiveness of antiviral treatments. The emergence of MRSA due to excessive antibiotic usage has led to the widespread use of vancomycin as an alternative treatment. Immunomodulatory antibiotics like azithromycin may also be considered. To study the dynamics of these coinfections, a multiscale model was developed. Parameter estimation and sensitivity analysis were performed, revealing influential parameters affecting the reproduction number. Numerical simulations showed that methicillin may increase the population of co-infected cells, while azithromycin can improve the host immune response but has limited impact on MRSA proliferation. Increased efficacy of vancomycin can lead to MRSA eradication. Combination of immunomodulatory antibiotics and vancomycin has minimal effect on co-infected cell population, but increased vancomycin efficacy can reduce coinfection severity. This study emphasizes the importance of continuous research, surveillance, and the development of effective strategies to combat the complexities of COVID-19 and MRSA coinfection.
... First is the immune response to SARS-CoV-2 that involves both innate and adaptive components. The innate immune response involves activation of macrophages, natural killer cells, as well as the production of inflammatory cytokines in defense against virus [14,15] and it has been studied through mathematical models as well [16][17][18]. The adaptive immune response involves the activation of T cells and B cells, which produce antibodies and target infected cells [19]. ...
... Numerous mathematical models have been developed to describe in-host dynamics of infectious diseases such as HIV [25][26][27], MALARIA [28], EBOLA [29,30], and INFLUENZA [31][32][33][34][35]. In the case of COVID-19, studies have developed models to investigate viral replication within host [36,37], interaction of the immune system with the viral kinetics of SARS-CoV-2 [16,18,20,[38][39][40][41], interaction of immune responses with the effects of antiviral therapy [17,[42][43][44], and multiscale modeling to examine the coupled within-host and between-host aspects of COVID-19 [45][46][47]. Additionally, age structured and network models [48][49][50] involving social and non-social interventions have been proposed to understand the impact of media and age distribution of population. ...
... Wang et al. [20] and Ghosh et al. [17] integrated immune response with virus dynamics through linear kinetics approach to discuss pathogenic features of SARS-CoV-2 infection besides evaluating various prospective therapies for COVID-19 using model simulations. Chowdhury et al. [18] developed a mathematical model to investigate the interaction between immune system and virus particles while also incorporating the roles of natural killer cells and T-lymphocytes. Other than virus and T cell interaction that is modeled through saturating kinetics, they have assumed all the other interactions to be linear. ...
... Since the disease transmission rate is uncertain, we develop a mathematical model to present the spread of the COVID-19 in India and worldwide. This model is helpful for further modelling studies [28][29][30], and we can compare our model with previous mathematical models with actual data values (MoHFW India) (Corona virus disease (COVID-19) 2020) for the authentication of the model. The paper is organised as follows: Section 2 is related to the mathematical model formulation and some basic definitions and theorems. ...
Article
Full-text available
The novel coronavirus SARS-Cov-2 is a pandemic condition and poses a massive menace to health. The governments of different countries and their various prohibitory steps to restrict the virus's expanse have changed individuals' communication processes. Due to physical and financial factors, the population's density is more likely to interact and spread the virus. We establish a mathematical model to present the spread of the COVID-19 in worldwide. In this article, we propose a novel mathematical model (‘ $ \mathbb {S}\mathbb {L}\mathbb {I}\mathbb {I}_{q}\mathbb {I}_{h}\mathbb {R}\mathbb {P} $ SLIIqIhRP’) to assess the impact of using hospitalization, quarantine measures, and pathogen quantity in controlling the COVID-19 pandemic. We analyse the boundedness of the model's solution by employing the Laplace transform approach to solve the fractional Gronwall's inequality. To ensure the uniqueness and existence of the solution, we rely on the Picard-Lindelof theorem. The model's basic reproduction number, a crucial indicator of epidemic potential, is determined based on the greatest eigenvalue of the next-generation matrix. We then employ stability theory of fractional differential equations to qualitatively examine the model. Our findings reveal that both locally and globally, the endemic equilibrium and disease-free solutions demonstrate symptomatic stability. These results shed light on the effectiveness of the proposed interventions in managing and containing the COVID-19 outbreak.
... The introduction of temporal delays in the model unveils its true dynamic behavior, which is contingent on the system's prior history [16][17][18]. Chowdhury et al. [19] developed a mathematical model for analyzing the interaction of the immune system with SARS-CoV-2 in the host. The model explores the dynamics of viral load within the host by considering the role of natural killer cells and T cells, and the developed model will contribute to the understanding of disease dynamics and help to establish a variety of possible COVID-19 therapeutic strategies. ...
Article
Full-text available
This paper explores a double stochastic network epidemic model within the constraints of limited medical resources and nonlinear contact rate. Initially, we investigate the dynamic behavior of this infectious disease model. Subsequently, we explore how the intensity of volatility and the speed of reversion influence the model’s dynamics. Moreover, we present the outcomes of simulations conducted on this double stochastic network model. These simulations shed light on the repercussions of various factors, such as the intensity of volatility, the speed of reversion, the intensity of white noise, the availability of limited medical resources, and the nonlinear contact rate on the dynamics of the epidemic. Lastly, we delve into the impact of the intensity of volatility and the speed of reversion on the spread of infectious diseases through a comprehensive examination of smooth distributions. In conclusion, our findings have unveiled the intrinsic mechanisms governing the dynamic changes in this double stochastic epidemic model. Furthermore, our study places a strong emphasis on the influence of volatility intensity and the speed of the reversion within the mean-reverting Ornstein–Uhlenbeck processes on the propagation of infectious diseases.
Article
Vaccination is widely recognized as a powerful tool in controlling the spread of diseases. However, it is important to acknowledge that some diseases are not completely preventable by vaccines, meaning that the protective effect of certain vaccines is not absolute. Additionally, the effectiveness of vaccines may diminish over time, which implies that even with completed vaccinations, there remains a potential risk of infection. Therefore, conducting comprehensive studies on how these imperfect vaccines impact the dynamics of disease transmission is crucial. In this study, we formulate and analyze a two-strain epidemic model with imperfect vaccination on complex networks. We derive the basic reproduction number and introduce the invasion reproduction numbers corresponding to each strain. Using the Lyapunov function method and LaSalle’s invariance principle, we demonstrate the global asymptotical stability of the disease-free equilibrium and dominant equilibrium. Furthermore, we apply persistence theory to verify the existence of coexistence equilibrium and the persistence of the disease. To validate our theoretical findings, we conduct numerical simulations. Through these simulations, we confirm that vaccination plays a significant role in controlling the spread of diseases, despite the imperfect efficacy of certain vaccines in specific cases. These findings highlight the importance of continued efforts to improve vaccine effectiveness and explore alternative strategies to enhance disease control measures.
Article
Leptospirosis is a bacteria infection prevalent in many tropical regions, caused by the genus Leptospira. Humans contract the disease by coming in contact with contaminated environments. This study proposes a deterministic mathematical model that links the within-host and between-host dynamics of leptospirosis and investigates its properties. The model's parameters were estimated by fitting it to real-life data using the "lsqcurvefit" package in MATLAB. The study employs global sensitivity analysis using Latin hypercube sampling with a partial rank correlation coefficient index and uses Pon-tryagin's maximum principle to identify cost-effective solutions for time-dependent intervention strategies to suppress the bacteria transmission within a specific period. The results of the study showed that bacteria replication within human and rodent hosts is the major driver of the overall dynamics of leptospirosis and therefore controlling the prevalence of the disease by focusing on the epidemiology components is necessary but will not be effective if the intra-host dynamics of the bacteria within rodent hosts and human hosts are not attentively considered. MSC 2020: 92B05; 92C37; 92D30
Article
Full-text available
The present study discussed a model to describe the SARS-CoV-2 viral kinetics in the presence of saturated antiviral responses. A discrete-time delay was introduced due to the time required for uninfected epithelial cells to activate a suitable antiviral response by generating immune cytokines and chemokines. We examined the system's stability at each equilibrium point. A threshold value was obtained for which the system switched from stability to instability via a Hopf bifurcation. The length of the time delay has been computed, for which the system has preserved its stability. Numerical results show that the system was stable for the faster antiviral responses of epithelial cells to the virus concentration, i.e., quick antiviral responses stabilized patients' bodies by neutralizing the virus. However, if the antiviral response of epithelial cells to the virus increased, the system became unstable, and the virus occupied the whole body, which caused patients' deaths.
Article
Full-text available
Introduction The immune responses play important roles in the course of disease initiation and progression upon virus infection such as SARS-CoV-2. As the tissues consist of spatial structures, the spatial dynamics of immune responses upon viral infection are essential to the outcome of infection. Methods A hybrid computational model based on cellular automata coupled with partial differential equations is developed to simulate the spatial patterns and dynamics of the immune responses of tissue upon virus infection with several different immune movement modes. Results Various patterns of the distribution of virus particles under different immune strengths and movement modes of immune cells are obtained through the computational models. The results also reveal that the directed immune cell wandering model has a better immunization effect. Several other characteristics, such as the peak level of virus density and onset time and the onset of the diseases, are also checked with different immune and physiological conditions, for example, different immune clearance strengths, and different cell-to-cell transmission rates. Furthermore, by the Lasso analysis, it is identified that the three main parameters had the most impact on the rate of onset time of disease. It is also shown that the cell-to-cell transmission rate has a significant effect and is more important for controlling the diseases than those for the cell-free virus given that the faster cell-to-cell transmission than cell-free transmission the rate of virus release is low. Discussion Our model simulates the process of viral and immune response interactions in the alveola repithelial tissues of infected individuals, providing insights into the viral propagation of viruses in two dimensions as well as the influence of immune response patterns and key factors on the course of infection.
Article
Full-text available
COVID-19 disease caused by the novel SARS-CoV-2 coronavirus has already brought unprecedented challenges for public health and resulted in huge numbers of cases and deaths worldwide. In the absence of effective vaccine, different countries have employed various other types of non-pharmaceutical interventions to contain the spread of this disease, including quarantines and lockdowns, tracking, tracing and isolation of infected individuals, and social distancing measures. Effectiveness of these and other measures of disease containment and prevention to a large degree depends on good understanding of disease dynamics, and robust mathematical models play an important role in forecasting its future dynamics. In this paper we focus on Ukraine, one of Europe's largest countries, and develop a mathematical model of COVID-19 dynamics, using latest data on parameters characterising clinical features of disease. For improved accuracy, our model includes age-stratified disease parameters, as well as age-and location-specific contact matrices to represent contacts. We show that the model is able to provide an accurate short-term forecast for the numbers and age distribution of cases and deaths. We also simulated different lockdown scenarios, and the results suggest that reducing work contacts is more efficient at reducing the disease burden than reducing school contacts, or implementing shielding for people over 60.
Article
Full-text available
The relationship between SARS-CoV-2 viral load and risk of disease progression remains largely undefined in coronavirus disease 2019 (COVID-19). Here, we quantify SARS-CoV-2 viral load from participants with a diverse range of COVID-19 disease severity, including those requiring hospitalization, outpatients with mild disease, and individuals with resolved infection. We detected SARS-CoV-2 plasma RNA in 27% of hospitalized participants, and 13% of outpatients diagnosed with COVID-19. Amongst the participants hospitalized with COVID-19, we report that a higher prevalence of detectable SARS-CoV-2 plasma viral load is associated with worse respiratory disease severity, lower absolute lymphocyte counts, and increased markers of inflammation, including C-reactive protein and IL-6. SARS-CoV-2 viral loads, especially plasma viremia, are associated with increased risk of mortality. Our data show that SARS-CoV-2 viral loads may aid in the risk stratification of patients with COVID-19, and therefore its role in disease pathogenesis should be further explored. In this study, Massachusetts Consortium for Pathogen Readiness (MassCPR) investigators assess the relationship between SARS-CoV-2 viral load and COVID-19 disease severity and report that the levels of detectable viral RNA, especially in plasma, correlates with severity of respiratory disease, inflammatory markers and predicted risk of death.
Article
Full-text available
COVID-19 has heightened human suffering, undermined the economy, turned the lives of billions of people around the globe upside down, and significantly affected the health, economic, environmental and social domains. This study aims to provide a comprehensive analysis of the impact of the COVID-19 outbreak on the ecological domain, the energy sector, society and the economy and investigate the global preventive measures taken to reduce the transmission of COVID-19. This analysis unpacks the key responses to COVID-19, the efficacy of current initiatives, and summarises the lessons learnt as an update on the information available to authorities, business and industry. This review found that a 72-hour delay in the collection and disposal of waste from infected households and quarantine facilities is crucial to controlling the spread of the virus. Broad sector by sector plans for socioeconomic growth as well as a robust entrepreneurship-friendly economy is needed for the business to be sustainable at the peak of the pandemic. The socioeconomic crisis has reshaped investment in energy and affected the energy sector significantly with most investment activity facing disruption due to mobility restrictions. Delays in energy projects are expected to create uncertainty in the years ahead. This report will benefit governments, leaders, energy firms and customers in addressing a pandemic-like situation in the future.
Article
Full-text available
Influenza viruses have perplexed scientists for over a hundred years. Yearly vaccines limit their spread, but they do not prevent all infections. Therapeutic treatments for those experiencing severe infection are limited; further advances are held back by insufficient understanding of the fundamental immune mechanisms responsible for immunopathology. NK cells and T cells are essential in host responses to influenza infection. They produce immunomodulatory cytokines and mediate the cytotoxic response to infection. An imbalance in NK and T cell responses can lead to two outcomes: excessive inflammation and tissue damage or insufficient anti-viral functions and uncontrolled infection. The main cause of death in influenza patients is the former, mediated by hyperinflammatory responses termed “cytokine storm.” NK cells and T cells contribute to cytokine storm, but they are also required for viral clearance. Many studies have attempted to distinguish protective and pathogenic components of the NK cell and T cell influenza response, but it has become clear that they are dynamic and integrated processes. This review will analyze how NK cell and T cell effector functions during influenza infection affect the host response and correlate with morbidity and mortality outcomes.
Article
Full-text available
Coronavirus Disease 2019 (COVID-19) has become a major health problem causing severe acute respiratory illness in humans. It has spread rapidly around the globe since its first identification in Wuhan, China, in December 2019. The causative virus is called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the World Health Organization (WHO) named the new epidemic disease Coronavirus Disease (COVID-19). The incidence of COVID-19 continues to increase with more than three million confirmed cases and over 244,000 deaths worldwide. There is currently no specific treatment or vaccine against COVID- 19. Therefore, in the absence of pharmaceutical interventions, the implementation of precautions and hygienic measures will be essential to control and to minimize human transmission of the virus. In this review, we highlight the epidemiology, transmission, symptoms, and treatment of this disease, as well as future strategies to manage the spread of this fatal coronavirus.
Article
Full-text available
The emergence and rapid spread of novel coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a potentially fatal disease is swiftly evolving public health crises worldwide. The origin of SARS-CoV-2 infection was first reported in people exposed to wet animal market in Wuhan City, China in December 2019. It was suggested that the infection is likely to be of zoonotic origin and transmitted to human through yet unknown intermediary. As of (22/05/2020), there are around 4,995,996 confirmed cases reported by WHO with 327,821 deaths. SARS-CoV-2 infection is transmitted via inhalation or direct contact of infected people's droplets. It has an incubation period ranging from 2 to 14 days or more. The rate of spread of SARS-CoV-2 is more than partially resembled coronavirus (SARS-CoV and MERS). The symptoms are similar to influenza like, breathlessness, sore throat and fatigue therefore, infected person is isolated and administrated with effective treatments. Infection is mild in most but in elderly (>50 years) and those with cardiac and respiratory disorder, it may progress to pneumonia, acute respiratory distress syndrome, and multi organ failure. People with strong immunity or those developed herd immunity are asymptomatic. Fatality rate ranges to 3-4% on case basis. Diagnosis of SARS-CoV-2 is recommended in respiratory secretions by special molecular tests like PCR, chest scan and common laboratory diagnosis. Currently, the existing treatment is essentially supportive and role of antiviral agents is yet to be established as there is no vaccination or therapy available. This review focuses on epidemiology, symptoms, transmission, pathogenesis, ongoing available treatments and future perspectives of SARS-CoV-2.
Article
----(Paper available until Feb 4, 2021 at https://authors.elsevier.com/c/1cFPX4syv1oKzU.)---- COVID-19 pandemic has underlined the impact of emergent pathogens as a major threat to human health. The development of quantitative approaches to advance comprehension of the current outbreak is urgently needed to tackle this severe disease. Considering different starting times of infection, mathematical models are proposed to represent SARS-CoV-2 dynamics in infected patients. Based on the target cell limited model, the within-host reproductive number for SARS-CoV-2 is consistent with the broad values of human influenza infection. The best model to fit the data was including immune cell response, which suggests a slow immune response peaking between 5 to 10 days post-onset of symptoms. The model with the eclipse phase, time in a latent phase before becoming productively infected cells, was not supported. Interestingly, model simulations predict that SARS-CoV-2 may replicate very slowly in the first days after infection, and viral load could be below detection levels during the first 4 days post infection. A quantitative comprehension of SARS-CoV-2 dynamics and the estimation of standard parameters of viral infections is the key contribution of this pioneering work. These models can serve for future evaluation of control theoretical approaches to tailor new drugs against COVID-19.
Article
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2019 in China and caused a coronavirus disease 2019 (COVID-19) pandemic. To mitigate the public health, economic and societal impacts of the virus, a vaccine is urgently needed. The development of SARS-CoV-2 vaccines was initiated in early January 2020 when the sequence of the virus became available and moved at record speed with one Phase I trial already starting in March 2020 and currently more than 180 vaccines in various stages of development. Phase I/II trial data is already available for several vaccine candidates and many have moved into Phase III trials. The data available so far suggests that effective and safe vaccines might become available within months rather than years.
Article
Coronavirus disease 2019 (COVID-19), an infectious disease caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is spreading and causing the global coronavirus pandemic. The viral dynamics of SARS-CoV-2 infection have not been quantitatively investigated. In this paper, we use mathematical models to study the pathogenic features of SARS-CoV-2 infection by examining the interaction between the virus, cells and immune responses. Models are fit to the data of SARS-CoV-2 infection in patients and non-human primates. Data fitting and numerical simulation show that viral dynamics of SARS-CoV-2 infection have a few distinct stages. In the initial stage, viral load increases rapidly and reaches the peak, followed by a plateau phase possibly generated by lymphocytes as a secondary target of infection. In the last stage, viral load declines due to the emergence of adaptive immune responses. When the initiation of seroconversion is late or slow, the model predicts viral rebound and prolonged viral persistence, consistent with the observation in non-human primates. Using the model we also evaluate the effect of several potential therapeutic interventions for SARS-CoV-2 infection. Model simulation shows that anti-inflammatory treatments or antiviral drugs combined with interferon are effective in reducing the duration of the viral plateau phase and diminishing the time to recovery. These results provide insights for understanding the infection dynamics and might help develop treatment strategies against COVID-19.
Article
To investigate the evolutionary and epidemiological dynamics of the current COVID-19 outbreak, a total of 112 genomes of SARS-CoV-2 strains sampled from China and 12 other countries with sampling dates between 24 December 2019 and 9 February 2020 were analyzed. We performed phylogenetic, split network, likelihood-mapping, model comparison, and phylodynamic analyses of the genomes. Based on Bayesian time-scaled phylogenetic analysis with the best-fitting combination models, we estimated the time to the most recent common ancestor (TMRCA) and evolutionary rate of SARS-CoV-2 to be 12 November 2019 (95% BCI: 11 October 2019 and 09 December 2019) and 9.90 × 10⁻⁴ substitutions per site per year (95% BCI: 6.29 × 10⁻⁴–1.35 × 10⁻³), respectively. Notably, the very low Re estimates of SARS-CoV-2 during the recent sampling period may be the result of the successful control of the pandemic in China due to extreme societal lockdown efforts. Our results emphasize the importance of using phylodynamic analyses to provide insights into the roles of various interventions to limit the spread of SARS-CoV-2 in China and beyond.