
Citation: Sezer Okay, Öznur Özge Özcan, Mesut Karahan. Nanoparticle-based delivery platforms for mRNA vaccine development[J]. AIMS Biophysics, 2020, 7(4): 323-338. doi: 10.3934/biophy.2020023
[1] | Yuto Omae, Yohei Kakimoto, Makoto Sasaki, Jun Toyotani, Kazuyuki Hara, Yasuhiro Gon, Hirotaka Takahashi . SIRVVD model-based verification of the effect of first and second doses of COVID-19/SARS-CoV-2 vaccination in Japan. Mathematical Biosciences and Engineering, 2022, 19(1): 1026-1040. doi: 10.3934/mbe.2022047 |
[2] | Mattia Zanella, Chiara Bardelli, Mara Azzi, Silvia Deandrea, Pietro Perotti, Santino Silva, Ennio Cadum, Silvia Figini, Giuseppe Toscani . Social contacts, epidemic spreading and health system. Mathematical modeling and applications to COVID-19 infection. Mathematical Biosciences and Engineering, 2021, 18(4): 3384-3403. doi: 10.3934/mbe.2021169 |
[3] | Fen-fen Zhang, Zhen Jin . Effect of travel restrictions, contact tracing and vaccination on control of emerging infectious diseases: transmission of COVID-19 as a case study. Mathematical Biosciences and Engineering, 2022, 19(3): 3177-3201. doi: 10.3934/mbe.2022147 |
[4] | Anichur Rahman, Muaz Rahman, Dipanjali Kundu, Md Razaul Karim, Shahab S. Band, Mehdi Sookhak . Study on IoT for SARS-CoV-2 with healthcare: present and future perspective. Mathematical Biosciences and Engineering, 2021, 18(6): 9697-9726. doi: 10.3934/mbe.2021475 |
[5] | Matthew Hayden, Bryce Morrow, Wesley Yang, Jin Wang . Quantifying the role of airborne transmission in the spread of COVID-19. Mathematical Biosciences and Engineering, 2023, 20(1): 587-612. doi: 10.3934/mbe.2023027 |
[6] | Kai Wang, Zhenzhen Lu, Xiaomeng Wang, Hui Li, Huling Li, Dandan Lin, Yongli Cai, Xing Feng, Yateng Song, Zhiwei Feng, Weidong Ji, Xiaoyan Wang, Yi Yin, Lei Wang, Zhihang Peng . Current trends and future prediction of novel coronavirus disease (COVID-19) epidemic in China: a dynamical modeling analysis. Mathematical Biosciences and Engineering, 2020, 17(4): 3052-3061. doi: 10.3934/mbe.2020173 |
[7] | Ryo Kinoshita, Sung-mok Jung, Tetsuro Kobayashi, Andrei R. Akhmetzhanov, Hiroshi Nishiura . Epidemiology of coronavirus disease 2019 (COVID-19) in Japan during the first and second waves. Mathematical Biosciences and Engineering, 2022, 19(6): 6088-6101. doi: 10.3934/mbe.2022284 |
[8] | Xinmiao Rong, Liu Yang, Huidi Chu, Meng Fan . Effect of delay in diagnosis on transmission of COVID-19. Mathematical Biosciences and Engineering, 2020, 17(3): 2725-2740. doi: 10.3934/mbe.2020149 |
[9] | A. D. Al Agha, A. M. Elaiw . Global dynamics of SARS-CoV-2/malaria model with antibody immune response. Mathematical Biosciences and Engineering, 2022, 19(8): 8380-8410. doi: 10.3934/mbe.2022390 |
[10] | Elodie Yedomonhan, Chénangnon Frédéric Tovissodé, Romain Glèlè Kakaï . Modeling the effects of Prophylactic behaviors on the spread of SARS-CoV-2 in West Africa. Mathematical Biosciences and Engineering, 2023, 20(7): 12955-12989. doi: 10.3934/mbe.2023578 |
As of April 2021, the coronavirus disease (COVID-19) continues to spread in Japan. On June 19, 2020, the Japanese government released the COVID-19 Contact-Confirming Application (COCOA), a smartphone app [1] that was developed to let COCOA users know whether they had come into contact with any COVID-19 infectors. The contact notification process comprises the following steps:
1. An individual with a pre-existing severe acute respiratory distress syndrome coronavirus 2 (COVID-19) infection and an uninfected individual both install COCOA on their smartphones (the infector is unaware of the infection because he/she has not undergone the reverse transcription polymerase chain reaction (RT-PCR test) for COVID-19).
2. The uninfected individual is in the proximity (<1 m) of the individual with a pre-existing COVID-19 infection for 15 minutes or longer. Through this process, the uninfected person becomes a new infector.
3. At a later date, the individual with the pre-existing COVID-19 infection who is the source of infection undergoes an RT-PCR test.
4. In the case of a positive result for COVID-19, the individual with a pre-existing COVID-19 infection who is the source of infection registers this result in the COCOA.
5. Then, individuals who came into contact with the individual with a pre-existing COVID-19 infection who is the source of infection within less than 2 weeks from the date of the positive result get a contact notification via COCOA.
The display of the COCOA is shown in Figure 1. If COCOA users click on the "check close contacts" button to the top-left, they receive information on whether they had any contact with the infectors. Infectors who become aware of a positive RT-PCR test result can register this result by using the lower "Register positive test result" button. Through these features, COCOA users can become aware of the possibility of infection through close contact with infectors with a pre-existing COVID-19 infection. If these exposed individuals decrease venturing outside and initiate self-quarantine, the number of total infectors in the community will decrease.
Worldwide, apps that are similar to COCOA have been developed (e.g., England [2], China, India, and Israel [3]) as various countries consider that the contact notification function is important for overcoming COVID-19. There are some important parameters that can decrease the number of new infectors as a function of COCOA. It is essential that the pre-existing infector and the close contact both use COCOA; therefore, the usage rate of COCOA is an important parameter (the term "close contact" refers to a person who has been in close contact with a pre-existing COVID-19 infector). Moreover, because the condition by which COCOA ascertains a positive contact with a pre-existing infector is contact for 15 minutes or longer at a distance of less than 1 m, the rate of new infectors who meet the abovementioned condition is important. Individuals who have undergone an RT-PCR test and have received positive results only could register their result in the COCOA. Therefore, the rate of PCR testing and the false-negative rate of PCR tests are important parameters. Furthermore, the duration of delay from the time of infection to undergoing an RT-PCR test is important. If the delay is too long, the close contact would not receive the "infector contact" notification from COCOA for a long duration. If the close contact becomes an infector through contact with pre-existing infectors, he/she may infect another individual. Moreover, COCOA cannot send the contact notification to a close contact if the infector who is the source of infection does not register a positive result in COCOA, even if he/she has received a positive result on the RT-PCR test and is aware of the infection. Therefore, the rate of registration of positive results is an important parameter. If the pre-existing infector who was the source of infection had maintained appropriate correspondence (using COCOA, getting the RT-PCR test done, registering positive results, and so on), the close contact can receive contact notification from COCOA. However, if a person in close contact does not undertake self-quarantine, the effectiveness of COCOA in decreasing the number of COVID-19 infectors does not occur. Therefore, the rate of self-quarantine of individuals who receive contact notifications from COCOA is another important parameter. Thus, there are some important parameters to estimate the effectiveness of COCOA.
Simulation is a desirable approach to clarify the effectiveness of COCOA. Many studies on infection have used simulation (e.g., [4,5,6,7]). Recently, COVID-19 infection simulations have been reported from target countries, such as China [8,9,10], India [11], The Netherlands [12], Korea, Italy, France [13], Nigeria [14], Arizona [15], Australia [16], South Carolina [17], Scandinavia [18], and Brazil [19,20]. Moreover, Niwa et al. reported the effectiveness of airport quarantine [21]. Kurahashi et al. reported the effectiveness of remote working and staggered commuting [22]. Vasconcelos et al. [23] and Macêdo et al. [24] analyzed the fatality curve of COVID-19 infectors by the simulation approach.
Recently, simulation studies have been conducted to estimate the effectiveness of contact-tracing apps. Various approaches [25,26,27,28,29] have been evaluated; however, the target of these researches were not COCOA. As an exception, Omae et al. [30] verified the effectiveness of COCOA using multi-agent simulation (MAS). However, because the calculation cost of MAS is high, this simulation scale is small (approximately 1000 agents), and undertaking trial and error in the various scenarios is difficult. Moreover, Omae et al. [30] did not consider the rate of RT-PCR test results and false negative rates. Kurita et al. [31] reported the effectiveness of COCOA in reducing the number of infectors; however, they did not consider the rate of registration of positive results and the false negative rate of RT-PCR tests.
Against this background, we developed a Susceptible-Infected-Removed (COCOA-SIR) model to estimate the effectiveness of COCOA in decreasing infectors to simulate infection spread in Japan. The COCOA-SIR model has the following important parameters for estimating the effect of COCOA.
1. the usage rate of COCOA
2. the rate of new infectors who are infected by contact with other infectors of less than 1 m and for 15 minutes or longer.
3. the rate of undergoing RT-PCR test
4. the delay from infection timing to undergoing an RT-PCR test
5. the false negative rate of RT-PCR test
6. the rate of registration of a COVID-19 positive result
7. the rate of self-quarantine of individuals who receive the contact notification from COCOA
The SIR model is used for infection epidemic simulation of COVID-19 (e.g., [32,33,34,35]) and, therefore, we adopted the SIR model as our research approach.
The basic SIR model [36] is represented as,
dS(t)dt=−β(t)S(t)I(t), | (2.1) |
dI(t)dt=β(t)S(t)I(t)−γI(t), | (2.2) |
dR(t)dt=γI(t), | (2.3) |
where S(t),I(t),R(t) are the number of susceptible (not infected), infected, and removed individuals at time t, respectively. "Removed persons" means individuals who are recovering from a COVID-19 infection or those who died of the infection. β(t) is the infectivity of time t, and γ is the recovery rate. If people in the community are careful about preventing infection, then the infectivity is low. However, if individuals in the community are not careful about infection, then the infectivity is high. The degree of care differs based on various situations. Therefore, β(t) is the function of time t.
Based on the above-described differential equations of Equations (2.1)-(2.3), we can understand the infection spread dynamics continuously. However, the number of new COVID-19 infectors is reported once a day in Japan. Therefore, it is desirable to express this parameter through a discrete-time equation rather than via a differential equation. Thus, we use,
St+1=St−β′tStIt, | (2.4) |
It+1=It+β′tStIt−γ′It, | (2.5) |
Rt+1=Rt+γ′It, | (2.6) |
as discrete-time equations for simulating infection spread. Where St, It, and Rt are the numbers of susceptible (not infected), infected, and removed persons on day t, respectively. β′t is the infectivity on day t, and γ′ is the recovery rate per day. Assuming that the infection period follows exponential distribution, the recovery rate γ′ is the reciprocal of the average infection period [37,38]. For example, if the average infection period is 7 days, γ′ is 1/7. Moreover, it is necessary to set S0, I0, and R0.
In Equation (2.4), the first term is the number of susceptible individuals on day t, and the second term represents the number of individuals who are susceptible to virus infection from day t to day t+1. In Equation (2.5), the first term is the number of infected persons on day t, the second term is the number of newly infected persons from day t to day t+1, and the third term represents the number of infected persons who are excluded due to recovery following virus infection (or death) from day t to day t+1. In Equation (2.6), the first term is the number of recovered individuals on day t, and the second term represents the number of newly recovered persons based on recovery from COVID-19 infection (or death) from day t to day t+1.
An effective reproduction number is an important index for the virus-spread simulation. The effective reproduction number refers to the number of additional persons infected by an infected person during the entire infection period [39]. In Equation (2.5), an infector infects β′tSt persons per day, and the average infection period is (γ′)−1. Therefore, we can define the effective reproduction number rt on day t as
rt=β′tStγ′. | (2.7) |
If the effective reproduction number rt>1, then β′tStIt−γ′It>0. Therefore, It+1 is larger than It. In contrast, if the effective reproduction number is rt<1, It+1 is less than It. For decreasing rt, it is required to decrease β′t. To decrease β′t, it is important to wear a mask, disinfect fingers, decrease going out, and so on.
With regard to the originality of our research, we propose the COCOA-SIR model, which can represent the effectiveness of the COCOA developed by Japan. After this section, we use two variables i,t to represent "day" i represents the timing of the occurrence of a new infector. t is a variable that represents the current day.
By the second term of Equation (2.5) of the basic SIR model, the number of new infectors occurring from day t to day t+1 is defined by the value obtained by multiplying the number of infectors It on day t and β′tSt. Thus, the infector It on day t generates new infectors. If COCOA is present in the community, we consider that a percentage of individuals who receive notification of contact with infectors from COCOA would undertake self-quarantine. Here, the number of infectors who receive notifications of contact with infectors through COCOA and undertake self-quarantine is defined as Inoticet. Then, the number of infectors that infect other persons on day t is It−Inoticet instead of It. Therefore, in the case of existing COCOA in society, the number of infectors on day t+1 is defined by
It+1=It+β′tSt(It−Inoticet)−γ′It. | (3.1) |
This means that the infectors It−Inoticet who did not receive a COCOA notification of contact with infectors (or did not take self-quarantine in spite of receiving it) would generate new infectors.
Herein, we consider the details of Inoticet. We explain the process flow until the new infectors receive a COCOA notification of contact, based on Figure 2. This is an example of how a newly infected person reproduces the infection by contact with an individual with a preexisting infection from days 3 to 4. In this case, to send the contact notification via COCOA, both individuals need to use COCOA and satisfy the condition of contact of COCOA ( < m and ≥ minutes). Moreover, an individual with a preexisting infection needs to undergo an RT-PCR test and to obtain a positive result, as well as input the fact into COCOA. In the case shown in Figure 2, on day 7, an individual with the preexisting infection fulfills these conditions. At this time, an individual who is newly infected from days 3 to 4 can receive a contact notification via COCOA. If a newly infected individual who receives the contact notification appropriately undertakes self-quarantine, he/she will not infect other individuals after day 7. With regard to the function of COCOA [1], the app retains the notification-related information for 14 days after the contact with an individual with a preexisting infection. Therefore, an individual who is newly infected from days 3 to 4 receives the notification via COCOA until day 17.
Based on the abovementioned discussions, it is necessary to fulfill various conditions to ensure the effectiveness of COCOA in decreasing new infectors. Furthermore, the timing when the infected individual receives the contact notification through COCOA is the time at which the individual with the preexisting infection undergoes the RT-PCR test, receives a positive test result, and inputs this result into COCOA. Therefore, the probability that a newly infected individual who appears on day i will receive a contact notification on day t, and will not infect others from day t to day t+1 is defined by
Pi,t={(puse)2×p15min×ppcri,t(1−pfalse)×pregist×phome(t≤i+14)0(otherwise). | (3.2) |
Where, puse is the usage rate of COCOA, p15min is the rate of satisfying the conditions of infective contact of more than 15 minutes at a distance less than 1 m, pfalse is the false-negative rate of the RT-PCR test. pregist is the rate of registration of a COVID-19-positive result, and phome is the rate of self-quarantine of a person who receives a contact notification via COCOA (shown in Table 1). There are two puse in Equation (3.2) because both an individual with a preexisting infection and a close contact use COCOA. The period of sending the contact notification was 14 days from the contact and time of infection [1]. Therefore, if the current day t is more than 14 days from day i, Pi,t is zero.
Variables | Meaning |
puse | Usage rate of COCOA |
p15min | The rate of new infectors infected by contact with other infectors less than 1 m and for 15 minutes or longer. |
ppcri,t | The rate of undergoing an RT-PCR test |
pfalse | false-negative rate of the RT-PCR test |
pregist | Rate of registration of the COVID-19-positive result |
phome | Rate of self-quarantine of an individual who receives a notification of contact through COCOA |
Next, we explain ppcri,t in Equation (3.2). As a function of COCOA, the timing of sending the contact notification to the close contact is not the timing of contact with the infected person who is the source of infection (it is not on day i). The timing when the close contact can receive the contact notification is when the infected person who is the source of infection registers a positive result on the RT-PCR test in COCOA. To represent the described delay time, we define the probability of the individual with the preexisting infection obtaining an RT-PCR test on day t (who is the source of a new infected person occurring on day i) as
ppcri,t=ppcrmax1+exp{−a(t−(i+1)−b)}, | (3.3) |
where, ppcrmax is the maximum probability of the individual undergoing an RT-PCR test. As it is a sigmoid function, ppcri,t∈(0,ppcrmax). where the parameter a is the gradient of the sigmoid function and the parameter b is the number of days that half of the infected individuals of ppcrmax undergo the RT-PCR test. For example, we show the results for ppcrmax=0.2 and (a,b)=(1.25,3),(4.25,3),(1.25,6),(4.25,6) in Figure 3. The horizontal axis t−(i+1) represents the number of days elapsed from the infection timing (day i) of individuals in close contact. Because t=i+1 is the next day of the infection timing by contact with infectors, ppcri,t=i+1 is a small value. The reason is that we assume the rate of undergoing RT-PCR test gradually increases depending on the elapsed time. We can understand that the larger the a is, the larger is the gradient of ppcri,t. Moreover, b is the number of days that half of the infected persons of ppcrmax undergo the RT-PCR test. As the parameters a,b represent the delay time from infection timing to the RT-PCR test, we call a,b the delaying parameters. For your information, ppcri,t is not more than ppcrmax. Therefore, in the case of Figure 3 (ppcrmax=0.2), 80% of individuals with preexisting infection who were the source of infection did not undergo an RT-PCR test regardless of how much time had elapsed. Moreover, there is a problem in that the RT-PCR test is negative even if the individual is infected (false negative). In this case, because infected persons cannot be aware of infection, they cannot register positive results into COCOA even if they are infected. As the false negative rate is pfalse, the 1−pfalse in Equation (3.2) is the correct positive rate.
We consider Inoticet, which is the number of infectors who receive contact notifications on day t based on the above-described discussions. We define the number of new infectors occurring from day i to day i+1 as I+i. As this is the second term of Equation (3.1), I+i is
I+i=β′iSi(Ii−Inoticei). | (3.4) |
Here, I+i, which is the infectors generated from day i to day i+1, and is recovered by the rate γ′ per day. Therefore, the number of unrecovered infected persons on day t is,
I+i(1−γ′)t−(i+1). | (3.5) |
In the case of COCOA existing in the community, some of I+i(1−γ′)t−(i+1) will receive a contact notification on COOCA. As it is the rate of satisfying the event, as shown in Figure 2, the number of infectors who have received contact notifications on COCOA is
Pi,tI+i(1−γ′)t−(i+1). | (3.6) |
In other words, the number of persons who were infected on day i and have not recovered from infection by day t and have received the contact notification is Pi,tI+i(1−γ′)t−(i+1).
These abovementioned discussions indicate only new infectors who are generated from day i to day i+1. As of day t, these new infectors exist for t−1 days. Therefore, as of day t, the number of new infectors who have not recovered from infection and have received the contact notification in COCOA and will not infect others by undertaking self-quarantine is
Inoticet=t−1∑i=0Pi,tI+i(1−γ′)t−(i+1) | (3.7) |
=t−1∑i=0Pi,tβ′iSi(Ii−Inoticei)(1−γ′)t−(i+1). | (3.8) |
Note that, by using Equation (3.2), the summation range of Equations (3.7) and (3.8) can also be from t−14 to t−1 instead of from 0 to t−1 when satisfying t≥14. In Equation (3.8), Inoticet includes Inotice{0,⋯,t−1}. Therefore, we cannot calculate Inotice0. As Inotice0 denotes the number of infectors who have received contact notification on COCOA on day 0, we set
Inotice0=const. | (3.9) |
In other words, we can set Inotice0 to any number on the simulation starting time. If there is no particular reason, set Inotice0 to 0. This means that there are no individuals who have received contact notifications at the simulation starting time. By substituting Inoticet calculated by Equation (3.8) for Equation (3.1), we can calculate the infectors It+1 on day t+1 by considering the effect of COCOA on decreasing the number of infectors.
Thus, we arrange Equation (3.1) as,
It+1=It+β′tStIt−γ′It−β′tStInoticet. | (3.10) |
Comparing Equation (2.5) of the basic SIR model and Equation (3.10) of the proposed SIR model, the first, second, and third terms are the same, and the fourth term is then added. In other words, we consider the fourth term β′tStInoticet of Equation (3.10) as the effect of COCOA on decreasing the number of new infectors. Therefore, we define the effect of COCOA in decreasing the number of new infectors on day t to day t+1 as
Eappt=β′tStInoticet | (3.11) |
=β′tStt−1∑i=0Pi,tβ′iSi(Ii−Inoticei)(1−γ′)t−(i+1). | (3.12) |
This indicates that the effect of COCOA is affected not only by Pi,t but also by various variables.
Based on the above discussion, to estimate the effect of COCOA in decreasing the number of infectors, we propose the COCOA-SIR model, defined as
St+1=St−β′tStIt+Eappt, | (3.13) |
It+1=It+β′tStIt−γ′It−Eappt, | (3.14) |
Rt+1=Rt+γ′It. | (3.15) |
Equation (3.14) to estimate the number of infectors It+1 is obtained by substituting Eappt defined by Equation (3.11) for Equation (3.10). Moreover, because the number of new infectors decreases by Eappt, Equation (3.13) to estimate the number of susceptible persons St+1 has the term Eappt. The number of recovered persons Rt+1 is the same as in the basic SIR model. Here, when we set the coefficient Pi,t,∀i,t affects the effect of COCOA as 0, Eappt=0 by Equation (3.12). In this case, the COCOA-SIR model defined by Equations (3.13), (3.14), and (3.15) are in the same form as the basic SIR model defined by Equations (2.4), (2.5), and (2.6). Therefore, we consider the COCOA-SIR model an extension of the basic SIR model.
The simulation results of the basic SIR and COCOA-SIR models are shown in Figure 4. The simulation parameters are rt=2 as the effective reproduction number, S0=9999,I0=1,R0=0 as the number of initial susceptible, infected, and recovered individuals, γ′=1/7 as the recovery rate (the average recovery period is 7 days), and the simulation period is 200 days. We set puse=1, p15min=0.5, ppcrmax=0.5, pfalse=0.1, pregist=1 and phome=1 for parameters of COCOA. Moreover, the delaying parameters were adopted as (a,b)=(4,3). From Figure 4, it was confirmed that the number of infectors decreased.
In this section, we compare the simulations in the case of the presence and absence of COCOA using the COCOA-SIR model and actual infector-open data in Japan [40]. COCOA was released on June 19, 2020 [41] in Japan. Open data [40] obtained after June 19, 2020 cannot be used because it is impossible to assume a Japan where the COCOA does not exist. Moreover, there are some days in which new infectors were reported to be zero, before February 12, 2020. This means that fluctuations in the number of newly infected people are unstable. Therefore, we use the open data [40] of reported new infectors in Japan from February 12, 2020 to June 18, 2020 for simulations (the maximum simulation period: tmax=128 days).
The open data [40] of the number of new infectors reported by Japan is only the number of infectors who were aware of infection based on the results of the RT-PCR test. There will be many infectors who are unaware of the infection. Therefore, we define the number of new infectors on day t, considering the amount of both individuals who are unaware and aware of infection as
I+,allt=CI+,findt, | (4.1) |
where I+,findt is the number of new infectors on day t reported in Japan, and C is a non-finding coefficient. Tsuchiya estimated that the number of new infectors, considering the amount of being unaware and aware of infection, was 23 times larger than that reported by Japan [39]. Therefore, we adopted C=23.
Kobayashi et al.[32] reported that γ′ ranges from 0.13 to 0.17 as depicting the spreading of COVID-19 infection in Japan; therefore, we adopted γ′=0.15, which is the median. In this study, because we assume that the infection period follows an exponential distribution according to reports from previous research [37] of the SIR model, we set γ′−1=1/0.15≃6.67 days as the average infection period. The function of the exponential distribution in the case of setting t as the infective period is
f(t)=γ′exp(−γ′t). | (4.2) |
By integrating the exponential distribution, the probability of recovering from 0 to x days is:
Pr(x)=∫x0f(t)dt=1−exp(−γ′x). | (4.3) |
For example, when adopting γ′=0.15, we obtain Pr(x=20)≃0.950. This means that approximately 95% of COVID-19 infectors recover within 20 days.
To perform simulations using the SIR model, it is necessary to set the numbers of initial susceptible, infected, and recovered individuals (S0, I0, and R0). As mentioned above, the starting time of the simulation was February 12, 2020, and the average infection period was 6.67 days. Therefore, we calculated the total number of new infectors reported in Japan for 7 days (from February 5 to February 11) prior to February 12, 2020. Subsequently, we multiplied the non-finding coefficient C to the total new infectors, and we adopted it as I0. As a result, the initial infectors I0 were 46 individuals in Japan. Moreover, because the total population of Japan is approximately 1.2×108 persons, we adopted the value of subtracting I0 from 1.2×108 as the initial number of susceptible (non-infected) individuals S0. Because of the starting time of the simulation, we assumed the initial number of recovered individuals to be zero (i.e., R0=0). In conclusion, we set S0+I0+R0=1.2×108, which is the total population of Japan.
To perform the simulation using the COCOA-SIR model, it is necessary to set the infectivity β′t. As mentioned above, we used the open data of periods without COCOA. Therefore, we searched for infectivity β′t that minimizes the absolute error between the new infectors β′tStIt using the basic SIR model shown in Equation (2.5) and the new infectors I+,allt reported by Japan. Thus, we adopted the optimal infectivity β′optt as,
β′optt=Argminβ′t|β′tStIt−I+,allt|. | (4.4) |
Moreover, by using Equation (2.7), we calculated the optimal effective reproduction number roptt as
roptt=β′opttStγ′. | (4.5) |
The line charts of new infectors in Japan I+,allt and new infectors reproduced by the SIR model using the optimal reproduction number roptt are shown in Figure 5. The black line on the upper side is I+,allt, and the yellow line on the upper side represents new infectors reproduced by the SIR model. As they are very similar, we adopted β′optt to simulate the spread of infection and to verify the effect of COCOA in reducing infectors in Japan. The blue line at the bottom of Figure 5 indicates the optimal reproduction number roptt solved by Equation (4.5).
According to the Japanese government, the number of COCOA users was 23.1 million on January 8, 2021, in Japan[41]. This means that the usage rate of COCOA in Japan is 0.1925 because the total population in Japan is 1.2×108. Therefore, we adopted puse=0.1925. Moreover, we adopted puse=0.6,0.9 to perform simulations in the case of the successful promotion of COCOA for the Japanese population.
Tsuchiya [39] reported that the estimated number of total infectors in Japan may be 23 times the number of infected individuals. In other words, the rate of RT-PCR testing in Japan may be approximately 1/23≃0.0435 for all infectors. Therefore, we adopted ppcrmax=0.0435 as the maximum rate of the PCR test. Next, we consider the delaying parameters of undergoing the RT-PCR test. It is difficult to determine the evidence-based values for the delaying parameters a,b in Equation (3.3). Therefore, we adopted (a,b)=(1.25,3) as the optimistic scenario and (a,b)=(1.25,6) as the pessimistic scenario, respectively (these forms are shown in Figure 3). This means that half of ppcrmax underwent the RT-PCR test by day 3 or 6. Moreover, we adopted ppcrmax=0.3,0.6,0.9 to perform simulations in the case of successful PCR tests in the Japanese population.
The total number of registrations of positive results on the RT-PCR test to COCOA is 5297 individuals on December 25, 2020, and 6929 individuals on January 8, 2021, [41]. Therefore, the number of registrations of positive results for these 2 weeks is 6929−5297=1632 individuals. Moreover, the number of new infectors for 2 weeks from December 25, 2020, to January 8, 2021, was 63383 individuals [40]. As the usage rate of COCOA puse is 0.1925, the estimated total number of infectors who use COCOA may be approximately 63383×0.1925≃12201 individuals for these 2 weeks. In this case, we can estimate pregist=1632/12201≃0.1337 as the registration rate of the positive result inputted to COCOA. Moreover, we adopted pregist=0.3,0.6,0.9 to perform simulations in the case of successful promotion of the registration of RT-PCR positive results to the Japanese population.
Kucirka et al. [42] reported a false-negative rate of the COVID-19 RT-PCR test. These authors indicated that the false negative rate depended on the days elapsed from the time of infection, and the median value was 0.38. Therefore, we adopted pfalse=0.38 as the false negative rate.
The conclusive condition of the COCOA of contact with infectors is that individuals are in proximity to the infectors by less than 1 m and for more than 15 minutes. However, it is difficult to know what percentage of infected people have been infected by satisfying these conditions. In other words, it is difficult to set the evidence-based value to p15min. Therefore, we adopted p15min=0.6 as the optimistic scenario and p15min=0.3 as the pessimistic scenario. Moreover, it is difficult to know what percentage of close contacts who receive a contact notification from COCOA undertake self-quarantine appropriately. Therefore, we adopted phome=0.3,0.6,0.9 to obtain the results for various scenarios.
We calculated the COCOA-SIR model, which consists of Equations (3.13), (3.14) and (3.15), using Python 3.7.3 [43], Numpy 1.18.5 [44]. The simulation results of the optimistic and pessimistic scenarios are presented in Tables 2 and 3, respectively. The optimistic and pessimistic scenarios indicate that the simulations adopted (a,b,p15min)=(1.25,3,0.6) and (a,b,p15min)=(1.25,6,0.3), respectively. Column 1 means scenario ID. snoapp represents Japan, which is the absence of COCOA, which means that the actual number of infectors are considered to be infectors who were unaware of the infection. s1actual,⋯,s3actual are the scenarios wherein parameters close to reality are adopted in the case of COCOA in Japan. s160,90,⋯,s1260,90 are the scenarios for the successful promotion of COCOA (puse=0.6,0.9). Thus, s160,90,⋯,s360,90 are the scenarios wherein parameters close to reality are adopted, with the exception of the usage rate of COCOA, and s460,90,⋯,s1260,90 are the scenarios in which all parameters improved. Moreover, sbest is the best scenario for decreasing infectors. "total infectors" in Table 2 and 3 is the total number of infectors at the ending time of simulation (t=tmax). "reductions" in Table 2 and 3 means that the number of decreasing infectors in comparison with the scenario snoapp and the "reduction rate" represents that rate.
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 405986 | 60 | 0.01 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 405925 | 121 | 0.03 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 405865 | 181 | 0.04 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405458 | 588 | 0.14 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 404870 | 1176 | 0.29 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 404284 | 1762 | 0.43 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 397030 | 9016 | 2.22 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 388199 | 17847 | 4.40 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 379549 | 26497 | 6.53 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 371079 | 34968 | 8.61 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 338913 | 67133 | 16.53 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 309339 | 96707 | 23.82 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 331284 | 74762 | 18.41 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 269415 | 136632 | 33.65 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 218349 | 187697 | 46.23 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 404724 | 1323 | 0.33 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 403405 | 2641 | 0.65 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 402091 | 3956 | 0.97 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 386019 | 20027 | 4.93 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 366909 | 39137 | 9.64 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 348677 | 57370 | 14.13 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 331284 | 74762 | 18.41 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 269415 | 136632 | 33.65 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 218349 | 187697 | 46.23 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 255710 | 150337 | 37.02 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 158220 | 247827 | 61.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 95976 | 310070 | 76.36 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 31714 | 374333 | 92.19 |
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 406032 | 14 | 0.00 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 406017 | 29 | 0.01 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 406002 | 44 | 0.01 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405904 | 142 | 0.04 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 405762 | 284 | 0.07 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 405620 | 426 | 0.11 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 403850 | 2196 | 0.54 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 401664 | 4382 | 1.08 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 399488 | 6558 | 1.62 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 397322 | 8724 | 2.15 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 388757 | 17290 | 4.26 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 380347 | 25699 | 6.33 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 386640 | 19406 | 4.78 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 368020 | 38026 | 9.37 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 350158 | 55888 | 13.76 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 405726 | 320 | 0.08 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 405407 | 640 | 0.16 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 405087 | 959 | 0.24 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 401119 | 4927 | 1.21 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 396243 | 9803 | 2.41 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 391417 | 14630 | 3.60 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 386640 | 19406 | 4.78 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 368020 | 38026 | 9.37 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 350158 | 55888 | 13.76 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 363485 | 42562 | 10.48 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 324726 | 81321 | 20.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 289464 | 116582 | 28.71 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 226108 | 179938 | 44.31 |
First, we compare snoapp and s1actual,⋯,s3actual. In both the optimistic and pessimistic scenarios, the rate of the number of decreasing infectors was only 0.00 % to 0.04 %. Therefore, we consider that the effect of COCOA on decreasing infectors is small in the actual conditions on January 2021 in Japan. Next, we check the cases of successful increase in the usage rate of COCOA to 60% (s160,⋯,s1260). In these cases, where ppcrmax,pregist,phome are high rates and the optimistic scenarios, the total infectors decrease by 46.23% (e.g., the scenario of s1260). However, even if the usage rate of COCOA is 60%, in these cases, ppcrmax,pregist,phome are low (e.g., scenarios s1,2,360), the effect of COCOA on decreasing infectors is insufficient. In the case of a usage rate of 90% (s1,⋯,1290), there are similar trends. In previous studies [30], the objective usage rate of the contact tracing app was 60% or higher. However, we found that ppcrmax,pregist,phome are also important parameters. The reasons for the different results from the previous studies are discussed. Omae et al.[30] verified the effect of COCOA on decreasing infectors using a multi-agent simulation. However, there were unrealistic assumptions, such as one assumption that all infectors underwent PCR tests and the false negative was zero [30].
We consider the results described in this report to reflect the specifications of COCOA as much as possible, and the conditions were set to be realistic. Therefore, the results shown in Tables 2 and 3 appear to be reliable. As for which is right, the optimistic or pessimistic scenario, it is difficult to decide this conclusively. However, it is important to promote not only the usage rate of COCOA, but also the rate of RT-PCR testing, the registration of positive results, and of self-quarantine to improve the effect of COCOA on decreasing the total number of infectors.
The line charts of some scenarios are shown in Figure 6 and 7. Figure 6 and 7 mean the optimistic and pessimistic scenarios, respectively. These indicate that snoapp is very similar to s1actual. Therefore, we consider that there will be little effect of COCOA on decreasing the number of infectors in the case of a small usage rate of COCOA. However, in the case of success in improving the rate of usage of COCOA, usage of RT-PCR tests, registration of positive results, and undertaking of self-quarantine (the scenarios: s1260, s1290, sbest), Figure 6, and 7 indicate that COCOA has a significant effect on decreasing the number of infectors. We consider the COCOA developed by Japan to be very important in overcoming the spread of COVID-19.
Caution must be exercised when explaining the explained simulations. The SIR model implies complete mixing of the population. However, this premise is not a realistic assumption because Japan is a sufficiently large country. For this point, we consider that this is not a problem because the COVID-19 outbreak in different locations in Japan started at approximately the same time (Postnikov [45] discussed the relationship between estimating infectors of the country as a whole and local disjoint sub-populations by the SIR model).
In this research, we proposed a COCOA-SIR model to verify the effect of the contact tracing app COCOA that was developed by Japan. The important parameters puse,p15min,ppcri,t,pfalse,pregist,phome shown in Table 1 were included in the COCOA-SIR model. Moreover, we performed simulations of various scenarios assumed for Japan. The results improved an understanding of the effect of COCOA on decreasing the number of infectors.
We show the future directions for research below. We have used the assumed numbers (e.g., a,b,p15min) to perform the simulation and, therefore, we will analyze and find more desirable numbers. In this research, we used an SIR model. However, we consider the SEIR model, which can represent the incubation period, may be a more desirable model for such a simulation. Therefore, we plan to propose and develop an SEIR model-based simulation to clarify the effect of COCOA on decreasing the number of infectors.
This work was supported in part by the Telecommunications Advancement Foundation (Grant No. 20203002 to Y. Omae) and a JSPS Grant-in-Aid for Scientific Research (C) (Grant No. 21K04535 to J. Toyotani].
The authors have no conflict of interest.
[1] | Sharp PA (2009) The centrality of RNA. Cell 136: 577-580. |
[2] | Cooper TA, Wan L, Dreyfuss G (2009) RNA and disease. Cell 136: 777-793. |
[3] | Fry LE, Patrício MI, Williams J, et al. (2019) Association of messenger RNA level with phenotype in patients with choroideremia: potential implications for gene therapy dose. JAMA Ophthalmol 138: 128-135. |
[4] | Li B, Zhang X, Dong Y (2019) Nanoscale platforms for messenger RNA delivery. Wires Nanomed Nanobi 11: e1530. |
[5] | Midoux P, Pichon C (2015) Lipid-based mRNA vaccine delivery systems. Expert Rev Vaccines 14: 221-234. |
[6] | Dannull J, Haley NR, Archer G, et al. (2013) Melanoma immunotherapy using mature DCs expressing the constitutive proteasome. J Clin Invest 123: 3135-3145. |
[7] | Van Lint S, Heirman C, Thielemans K, et al. (2013) mRNA: From a chemical blueprint for protein production to an off-the-shelf therapeutic. Hum Vacc Immunother 9: 265-274. |
[8] | Yang J, Arya S, Lung P, et al. (2019) Hybrid nanovaccine for the co-delivery of the mRNA antigen and adjuvant. Nanoscale 11: 21782-21789. |
[9] | Le TT, Andreadakis Z, Kumar A, et al. (2020) The COVID-19 vaccine development landscape. Nat Rev Drug Discov 19: 305-306. |
[10] | Geall AJ, Mandl CW, Ulmer JB (2013) RNA: the new revolution in nucleic acid vaccines. Semin Immunol 25: 152-159. |
[11] | Pardi N, Hogan MJ, Porter FW, et al. (2018) mRNA vaccines—a new era in vaccinology. Nat Rev Drug Discov 17: 261-279. |
[12] | Pascolo S (2015) The messenger's great message for vaccination. Expert Rev Vaccines 14: 153-156. |
[13] | Deering RP, Kommareddy S, Ulmer JB, et al. (2014) Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv 11: 885-899. |
[14] | Liu MA (2010) Immunologic basis of vaccine vectors. Immunity 33: 504-515. |
[15] | Jäschke A, Helm M (2003) RNA sex. Chem Biol 10: 1148-1150. |
[16] | Pollard C, Rejman J, De Haes W, et al. (2013) Type I IFN counteracts the induction of antigen-specific immune responses by lipid-based delivery of mRNA vaccines. Mol Ther 21: 251-259. |
[17] | Vallazza B, Petri S, Poleganov MA, et al. (2015) Recombinant messenger RNA technology and its application in cancer immunotherapy, transcript replacement therapies, pluripotent stem cell induction, and beyond. Wiley Interdiscip Rev RNA 6: 471-499. |
[18] | Gómez-Aguado I, Rodríguez-Castejón J, Vicente-Pascual M, et al. (2020) Nanomedicines to deliver mRNA: State of the art and future perspectives. Nanomaterials 10: 364. |
[19] | Versteeg L, Almutairi MM, Hotez PJ, et al. (2019) Enlisting the mRNA vaccine platform to combat parasitic infections. Vaccines 7: 122. |
[20] | Hekele A, Bertholet S, Archer J, et al. (2013) Rapidly produced SAM® vaccine against H7N9 influenza is immunogenic in mice. Emerg Microbes Infect 2: e52. |
[21] | Lindgren G, Ols S, Liang F, et al. (2017) Induction of robust B cell responses after influenza mRNA vaccination is accompanied by circulating hemagglutinin-specific ICOS+ PD-1+ CXCR3+ T follicular helper cells. Front Immunol 8: 1539. |
[22] | Luo F, Zheng L, Hu Y, et al. (2017) Induction of protective immunity against Toxoplasma gondii in mice by nucleoside triphosphate hydrolase-II (NTPase-II) self-amplifying RNA vaccine encapsulated in lipid nanoparticle (LNP). Front Microbiol 8: 605. |
[23] | Michel T, Golombek S, Steinle H, et al. (2019) Efficient reduction of synthetic mRNA induced immune activation by simultaneous delivery of B18R encoding mRNA. J Biol Eng 13: 40. |
[24] | Appay V, Douek DC, Price DA (2008) CD8+ T cell efficacy in vaccination and disease. Nat Med 14: 623-628. |
[25] | Pardi N, Hogan MJ, Naradikian MS, et al. (2018) Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med 215: 1571-1588. |
[26] | Zarghampoor F, Azarpira N, Khatami SR, et al. (2019) Improved translation efficiency of therapeutic mRNA. Gene 707: 231-238. |
[27] | Kowalski PS, Rudra A, Miao L, et al. (2019) Delivering the messenger: Advances in technologies for therapeutic mRNA delivery. Mol Ther 27: 710-728. |
[28] | Reichmuth AM, Oberli MA, Jaklenec A, et al. (2016) mRNA vaccine delivery using lipid nanoparticles. Ther Deliv 7: 319-334. |
[29] | Lundstrom K (2009) Alphaviruses in gene therapy. Viruses 1: 13-25. |
[30] | Chira S, Jackson CS, Oprea I, et al. (2015) Progresses towards safe and efficient gene therapy vectors. Oncotarget 6: 30675-30703. |
[31] | Ku SH, Jo SD, Lee YK, et al. (2016) Chemical and structural modifications of RNAi therapeutics. Adv Drug Deliv Rev 104: 16-28. |
[32] | Presnyak V, Alhusaini N, Chen YH, et al. (2015) Codon optimality is a major determinant of mRNA stability. Cell 160: 1111-1124. |
[33] | Thess A, Grund S, Mui BL, et al. (2015) Sequence-engineered mRNA without chemical nucleoside modifications enables an effective protein therapy in large animals. Mol Ther 23: 1456-1464. |
[34] | Wojtczak BA, Sikorski PJ, Fac-Dabrowska K, et al. (2018) 5′-phosphorothiolate dinucleotide cap analogues: Reagents for messenger RNA modification and potent small-molecular inhibitors of decapping enzymes. J Am Chem Soc 140: 5987-5999. |
[35] | Li B, Luo X, Dong Y (2016) Effects of chemically modified messenger RNA on protein expression. Bioconjug Chem 27: 849-853. |
[36] | Li M, Zhao M, Fu Y, et al. (2016) Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal epithelial barrier via intra- and paracellular pathways. J Control Release 228: 9-19. |
[37] | Svitkin YV, Cheng YM, Chakraborty T, et al. (2017) N1-methyl-pseudouridine in mRNA enhances translation through eIF2a-dependent and independent mechanisms by increasing ribosome density. Nucleic Acids Res 45: 6023-6036. |
[38] | Oberg AL, Kennedy RB, Li P, et al. (2011) Systems biology approaches to new vaccine development. Curr Opin Immunol 23: 436-443. |
[39] | Auffan M, Rose J, Bottero JY, et al. (2009) Towards a definition of inorganic nanoparticles from an environmental, health and safety perspective. Nat Nanotechnol 4: 634-641. |
[40] | Treuel L, Jiang X, Nienhaus GU (2013) New views on cellular uptake and trafficking of manufactured nanoparticles. J R Soc Interface 10: 20120939. |
[41] | Ulkoski D, Bak A, Wilson JT, et al. (2019) Recent advances in polymeric materials for the delivery of RNA therapeutics. Expert Opin Drug Deliv 16: 1149-1167. |
[42] | Pérez-Ortín JE, Alepuz P, Chávez S, et al. (2013) Eukaryotic mRNA decay: Methodologies, pathways, and links to other stages of gene expression. J Mol Biol 425: 3750-3775. |
[43] | Pati R, Shevtsov M, Sonawane A (2018) Nanoparticle vaccines against infectious diseases. Front Immunol 9: 2224. |
[44] | Means TK, Hayashi F, Smith KD, et al. (2003) The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol 170: 5165-5175. |
[45] | Boraschi D, Italiani P, Palomba R, et al. (2017) Nanoparticles and innate immunity: new perspectives on host defence. Semin Immunol 34: 33-51. |
[46] | Chen YS, Hung YC, Lin WH, et al. (2010) Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide. Nanotechnology 21: 195101. |
[47] | Wang T, Zou M, Jiang H, et al. (2011) Synthesis of a novel kind of carbon nanoparticle with large mesopores and macropores and its application as an oral vaccine adjuvant. Eur J Pharm Sci 44: 653-659. |
[48] | Xu L, Liu Y, Chen Z, et al. (2012) Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. Nano Lett 12: 2003-2012. |
[49] | Tao W, Gill HS (2015) M2e-immobilized gold nanoparticles as influenza A vaccine: role of soluble M2e and longevity of protection. Vaccine 33: 2307-2315. |
[50] |
Li X, Deng X, Huang Z (2001) In vitro protein release and degradation of poly- |
[51] | Chahal JS, Fang T, Woodham AW, et al. (2017) An RNA nanoparticle vaccine against Zika virus elicits antibody and CD8+ T cell responses in a mouse model. Sci Rep 7: 252. |
[52] | Chahal JS, Khan OF, Cooper CL, et al. (2016) Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and toxoplasma gondii challenges with a single dose. Proc Natl Acad Sci U S A 113: E4133-E4142. |
[53] | Sharifnia Z, Bandehpour M, Hamishehkar H, et al. (2019) In-vitro transcribed mRNA delivery using PLGA/PEI nanoparticles into human monocyte-derived dendritic cells. Iran J Pharm Res 18: 1659-1675. |
[54] | Uchida S, Kinoh H, Ishii T, et al. (2016) Systemic delivery of messenger RNA for the treatment of pancreatic cancer using polyplex nanomicelles with a cholesterol moiety. Biomaterials 82: 221-228. |
[55] | Kaczmarek JC, Patel AK, Kauffman KJ, et al. (2016) Polymer-lipid nanoparticles for systemic delivery of mRNA to the lungs. Angew Chem Int Ed Engl 55: 13808-13812. |
[56] | Patel AK, Kaczmarek JC, Bose S, et al. (2019) Inhaled nanoformulated mRNA polyplexes for protein production in lung epithelium. Adv Mater 31: e1805116. |
[57] | Liu Y, Li Y, Keskin D, et al. (2019) Poly(β-amino esters): Synthesis, formulations, and their biomedical applications. Adv Healthc Mater 8: e1801359. |
[58] | Capasso Palmiero U, Kaczmarek JC, Fenton OS, et al. (2018) Poly(β-amino ester)-co-poly(caprolactone) terpolymers as nonviral vectors for mRNA delivery in vitro and in vivo. Adv Healthc Mater 7: e1800249. |
[59] | Palamà IE, Cortese B, D'Amone S, et al. (2015) mRNA delivery using non-viral PCL nanoparticles. Biomater Sci 3: 144-151. |
[60] | Lacroix C, Humanes A, Coiffier C, et al. (2020) Polylactide-based reactive micelles as a robust platform for mRNA delivery. Pharm Res 37: 30. |
[61] | Dong Y, Dorkin JR, Wang W, et al. (2016) Poly(glycoamidoamine) brushes formulated nanomaterials for systemic siRNA and mRNA delivery in vivo. Nano Lett 16: 842-848. |
[62] | Palmerston Mendes L, Pan J, Torchilin VP (2017) Dendrimers as nanocarriers for nucleic acid and drug delivery in cancer therapy. Molecules 22: 1401. |
[63] | Franiak-Pietryga I, Ziemba B, Messmer B, et al. (2018) Dendrimers as drug nanocarriers: the future of gene therapy and targeted therapies in cancer. Dendrimers: Fundamentals and Applications IntechOpen, 7. |
[64] | Islam MA, Xu Y, Tao W, et al. (2018) Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat Biomed Eng 2: 850-864. |
[65] | Hajam IA, Senevirathne A, Hewawaduge C, et al. (2020) Intranasally administered protein coated chitosan nanoparticles encapsulating influenza H9N2 HA2 and M2e mRNA molecules elicit protective immunity against avian influenza viruses in chickens. Vet Res 51: 37. |
[66] | McCullough KC, Bassi I, Milona P, et al. (2014) Self-replicating replicon-RNA delivery to dendritic cells by chitosan-nanoparticles for translation in vitro and in vivo. Mol Ther Nucleic Acids 3: e173. |
[67] | Maiyo F, Singh M (2019) Folate-targeted mRNA delivery using chitosan-functionalized selenium nanoparticles: potential in cancer immunotherapy. Pharmaceuticals (Basel) 12: 164. |
[68] | Son S, Nam J, Zenkov I, et al. (2020) Sugar-nanocapsules imprinted with microbial molecular patterns for mRNA vaccination. Nano Lett 20: 1499-1509. |
[69] | Siewert C, Haas H, Nawroth T, et al. (2019) Investigation of charge ratio variation in mRNA - DEAE-dextran polyplex delivery systems. Biomaterials 192: 612-620. |
[70] | Zeng C, Zhang C, Walker PG, et al. (2020) Formulation and delivery technologies for mRNA vaccines. Current Topics in Microbiology and Immunology Berlin: Springer. |
[71] | Scheel B, Teufel R, Probst J, et al. (2005) Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur J Immunol 35: 1557-1566. |
[72] | Schlake T, Thess A, Fotin-Mleczek M, et al. (2012) Developing mRNA-vaccine technologies. RNA Biol 9: 1319-1330. |
[73] | Fotin-Mleczek M, Duchardt KM, Lorenz C, et al. (2011) Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and provide antitumor activity. J Immunother 34: 1-15. |
[74] | Schnee M, Vogel AB, Voss D, et al. (2016) An mRNA vaccine encoding rabies virus glycoprotein induces protection against lethal infection in mice and correlates of protection in adult and newborn pigs. PLoS Negl Trop Dis 10: e0004746. |
[75] | Udhayakumar VK, De Beuckelaer A, McCaffrey J, et al. (2017) Arginine-rich peptide-based mRNA nanocomplexes efficiently instigate cytotoxic T cell immunity dependent on the amphipathic organization of the peptide. Adv Healthc Mater 6: 1601412. |
[76] | Coolen AL, Lacroix C, Mercier-Gouy P, et al. (2019) Poly(lactic acid) nanoparticles and cell-penetrating peptide potentiate mRNA-based vaccine expression in dendritic cells triggering their activation. Biomaterials 195: 23-37. |
[77] | Jekhmane S, De Haas R, Paulino da Silva Filho O, et al. (2017) Virus-like particles of mRNA with artificial minimal coat proteins: particle formation, stability, and transfection efficiency. Nucleic Acid Ther 27: 159-167. |
[78] | Li J, Sun Y, Jia T, et al. (2014) Messenger RNA vaccine based on recombinant MS2 virus-like particles against prostate cancer. Int J Cancer 134: 1683-1694. |
[79] | Sun S, Li W, Sun Y, et al. (2011) A new RNA vaccine platform based on MS2 virus-like particles produced in saccharomyces cerevisiae. Biochem Biophys Res Commun 407: 124-128. |
[80] | Zhitnyuk Y, Gee P, Lung MSY, et al. (2018) Efficient mRNA delivery system utilizing chimeric VSVG-L7Ae virus-like particles. Biochem Biophys Res Commun 505: 1097-1102. |
[81] | Kauffman KJ, Webber MJ, Anderson DG (2016) Materials for non-viral intracellular delivery of messenger RNA therapeutics. J Control Release 240: 227-234. |
[82] | Kulkarni JA, Cullis PR, Van Der Meel R (2018) Lipid nanoparticles enabling gene therapies: From concepts to clinical utility. Nucleic Acid Ther 28: 146-157. |
[83] | Dimitriadis GJ (1978) Translation of rabbit globin mRNA introduced by liposomes into mouse lymphocytes. Nature 274: 923-924. |
[84] | Moon JJ, Suh H, Bershteyn A, et al. (2011) Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses. Nat Mater 10: 243-251. |
[85] | Tyagi RK, Garg NK, Sahu T (2012) Vaccination strategies against malaria: novel carrier(s) more than a tour de force. J Control Release 162: 242-254. |
[86] | Adler-Moore J, Munoz M, Kim H, et al. (2011) Characterization of the murine Th2 response to immunization with liposomal M2e influenza vaccine. Vaccine 29: 4460-4468. |
[87] | Monslow MA, Elbashir S, Sullivan NL, et al. (2020) Immunogenicity generated by mRNA vaccine encoding VZV gE antigen is comparable to adjuvanted subunit vaccine and better than live attenuated vaccine in nonhuman primates. Vaccine 38: 5793-5802. |
[88] | Erasmus JH, Archer J, Fuerte-Stone J, et al. (2020) Intramuscular delivery of replicon RNA encoding ZIKV-117 human monoclonal antibody protects against Zika virus infection. Mol Ther Methods Clin Dev 18: 402-414. |
[89] | Freyn AW, da Silva JR, Rosado VC, et al. (2020) A multi-targeting, nucleoside-modified mRNA influenza virus vaccine provides broad protection in mice. Mol Ther 28: 1569-1584. |
[90] | Lo MK, Spengler JR, Welch SR, et al. (2020) Evaluation of a single-dose nucleoside-modified messenger RNA vaccine encoding Hendra virus-soluble glycoprotein against lethal Nipah virus challenge in Syrian hamsters. J Infect Dis 221: S493-S498. |
[91] | Yang T, Li C, Wang X, et al. (2020) Efficient hepatic delivery and protein expression enabled by optimized mRNA and ionizable lipid nanoparticle. Bioact Mater 5: 1053-1061. |
[92] | Moyo N, Wee EG, Korber B, et al. (2020) Tetravalent immunogen assembled from conserved regions of HIV-1 and delivered as mRNA demonstrates potent preclinical T-cell immunogenicity and breadth. Vaccines (Basel) 8: 360. |
[93] | Lou G, Anderluzzi G, Schmidt ST, et al. (2020) Delivery of self-amplifying mRNA vaccines by cationic lipid nanoparticles: The impact of cationic lipid selection. J Control Release 325: 370-379. |
[94] | Mai Y, Guo J, Zhao Y, et al. (2020) Intranasal delivery of cationic liposome-protamine complex mRNA vaccine elicits effective anti-tumor immunity. Cell Immunol 354: 104143. |
[95] | Eygeris Y, Patel S, Jozic A, et al. (2020) Deconvoluting lipid nanoparticle structure for messenger RNA delivery. Nano Lett 20: 4543-4549. |
[96] | Van Hoecke L, Verbeke R, De Vlieger D, et al. (2020) mRNA encoding a bispecific single domain antibody construct protects against influenza A virus infection in mice. Mol Ther Nucleic Acids 20: 777-787. |
[97] | Zhong Z, Mc Cafferty S, Combes F, et al. (2018) mRNA therapeutics deliver a hopeful message. Nano Today 23: 16-39. |
[98] | Bogers WM, Oostermeijer H, Mooij P, et al. (2015) Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis 211: 947-955. |
[99] |
Jackson LA, Anderson EJ, Rouphael NG, et al. (2020) An mRNA vaccine against SARS-CoV-2—preliminary report. N Engl J Med . doi: 10.1056/NEJMoa2022483
![]() |
[100] | Alberer M, Gnad-Vogt U, Hong HS, et al. (2017) Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet 390: 1511-1520. |
[101] | Bahl K, Senn JJ, Yuzhakov O, et al. (2017) Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol Ther 25: 1316-1327. |
[102] | Feldman RA, Fuhr R, Smolenov I, et al. (2019) mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials. Vaccine 37: 3326-3334. |
[103] | Ding Y, Jiang Z, Saha K, et al. (2014) Gold nanoparticles for nucleic acid delivery. Mol Ther 22: 1075-1083. |
[104] |
Liu J, Miao L, Sui J, et al. (2020) Nanoparticle cancer vaccines: Design considerations and recent advances. Asian J Pharm Sci . doi: 10.1016/j.ajps.2019.10.006
![]() |
[105] | Yeom JH, Ryou SM, Won M, et al. (2013) Inhibition of xenograft tumor growth by gold nanoparticle-DNA oligonucleotide conjugates-assisted delivery of BAX mRNA. PLoS One 8: e75369. |
[106] | Azmi F, Ahmad Fuaad AAH, Skwarczynski M, et al. (2014) Recent progress in adjuvant discovery for peptide-based subunit vaccines. Hum Vaccin Immunother 10: 778-796. |
[107] | Coffman RL, Sher A, Seder RA (2010) Vaccine adjuvants: Putting innate immunity to work. Immunity 33: 492-503. |
[108] | Reed SG, Orr MT, Fox CB (2013) Key roles of adjuvants in modern vaccines. Nat Med 19: 1597-1608. |
[109] | Oleszycka E, Lavelle EC (2014) Immunomodulatory properties of the vaccine adjuvant alum. Curr Opin Immunol 28: 1-5. |
[110] | Alving CR (2009) Vaccine adjuvants. Vaccines for Biodefense and Emerging and Neglected Diseases London: Elsevier, 115-129. |
[111] | Hussein WM, Liu TY, Skwarczynski M, et al. (2014) Toll-like receptor agonists: a patent review (2011–2013). Expert Opin Ther Pat 24: 453-470. |
[112] | Montomoli E, Piccirella S, Khadang B, et al. (2011) Current adjuvants and new perspectives in vaccine formulation. Expert Rev Vaccines 10: 1053-1061. |
[113] | Mamo T, Poland GA (2012) Nanovaccinology: The next generation of vaccines meets 21st century materials science and engineering. Vaccine 30: 6609-6611. |
[114] | Banchereau J, Briere F, Caux C, et al. (2000) Immunobiology of dendritic cells. Annu Rev Immunol 18: 767-811. |
[115] | Oyewumi MO, Kumar A, Cui ZR (2010) Nano-microparticles as immune adjuvants: correlating particle sizes and the resultant immune responses. Expert Rev Vaccines 9: 1095-1107. |
[116] | Marasini N, Skwarczynski M, Toth I (2014) Oral delivery of nanoparticle-based vaccines. Expert Rev Vaccines 13: 1361-1376. |
[117] | Kawai T, Akira S (2011) Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34: 637-650. |
[118] | Vasilichin VA, Tsymbal SA, Fakhardo AF, et al. (2020) Effects of metal oxide nanoparticles on Toll-like receptor mRNAs in human monocytes. Nanomaterials (Basel) 10: 127. |
[119] | Roy R, Kumar D, Sharma A, et al. (2014) ZnO nanoparticles induced adjuvant effect via toll-like receptors and Src signaling in Balb/c mice. Toxicol Lett 230: 421-433. |
[120] | De Temmerman M-L, Rejman J, Demeester J, et al. (2011) Particulate vaccines: on the quest for optimal delivery and immune response. Drug Discov Today 16: 569-582. |
[121] | Hafner AM, Corthésy B, Merkle HP (2013) Particulate formulations for the delivery of poly(I: C) as vaccine adjuvant. Adv Drug Deliv Rev 65: 1386-1399. |
Variables | Meaning |
puse | Usage rate of COCOA |
p15min | The rate of new infectors infected by contact with other infectors less than 1 m and for 15 minutes or longer. |
ppcri,t | The rate of undergoing an RT-PCR test |
pfalse | false-negative rate of the RT-PCR test |
pregist | Rate of registration of the COVID-19-positive result |
phome | Rate of self-quarantine of an individual who receives a notification of contact through COCOA |
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 405986 | 60 | 0.01 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 405925 | 121 | 0.03 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 405865 | 181 | 0.04 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405458 | 588 | 0.14 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 404870 | 1176 | 0.29 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 404284 | 1762 | 0.43 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 397030 | 9016 | 2.22 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 388199 | 17847 | 4.40 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 379549 | 26497 | 6.53 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 371079 | 34968 | 8.61 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 338913 | 67133 | 16.53 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 309339 | 96707 | 23.82 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 331284 | 74762 | 18.41 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 269415 | 136632 | 33.65 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 218349 | 187697 | 46.23 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 404724 | 1323 | 0.33 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 403405 | 2641 | 0.65 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 402091 | 3956 | 0.97 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 386019 | 20027 | 4.93 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 366909 | 39137 | 9.64 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 348677 | 57370 | 14.13 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 331284 | 74762 | 18.41 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 269415 | 136632 | 33.65 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 218349 | 187697 | 46.23 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 255710 | 150337 | 37.02 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 158220 | 247827 | 61.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 95976 | 310070 | 76.36 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 31714 | 374333 | 92.19 |
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 406032 | 14 | 0.00 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 406017 | 29 | 0.01 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 406002 | 44 | 0.01 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405904 | 142 | 0.04 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 405762 | 284 | 0.07 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 405620 | 426 | 0.11 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 403850 | 2196 | 0.54 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 401664 | 4382 | 1.08 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 399488 | 6558 | 1.62 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 397322 | 8724 | 2.15 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 388757 | 17290 | 4.26 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 380347 | 25699 | 6.33 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 386640 | 19406 | 4.78 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 368020 | 38026 | 9.37 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 350158 | 55888 | 13.76 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 405726 | 320 | 0.08 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 405407 | 640 | 0.16 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 405087 | 959 | 0.24 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 401119 | 4927 | 1.21 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 396243 | 9803 | 2.41 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 391417 | 14630 | 3.60 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 386640 | 19406 | 4.78 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 368020 | 38026 | 9.37 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 350158 | 55888 | 13.76 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 363485 | 42562 | 10.48 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 324726 | 81321 | 20.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 289464 | 116582 | 28.71 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 226108 | 179938 | 44.31 |
Variables | Meaning |
puse | Usage rate of COCOA |
p15min | The rate of new infectors infected by contact with other infectors less than 1 m and for 15 minutes or longer. |
ppcri,t | The rate of undergoing an RT-PCR test |
pfalse | false-negative rate of the RT-PCR test |
pregist | Rate of registration of the COVID-19-positive result |
phome | Rate of self-quarantine of an individual who receives a notification of contact through COCOA |
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 405986 | 60 | 0.01 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 405925 | 121 | 0.03 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 405865 | 181 | 0.04 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405458 | 588 | 0.14 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 404870 | 1176 | 0.29 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 404284 | 1762 | 0.43 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 397030 | 9016 | 2.22 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 388199 | 17847 | 4.40 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 379549 | 26497 | 6.53 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 371079 | 34968 | 8.61 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 338913 | 67133 | 16.53 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 309339 | 96707 | 23.82 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 331284 | 74762 | 18.41 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 269415 | 136632 | 33.65 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 218349 | 187697 | 46.23 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 404724 | 1323 | 0.33 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 403405 | 2641 | 0.65 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 402091 | 3956 | 0.97 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 386019 | 20027 | 4.93 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 366909 | 39137 | 9.64 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 348677 | 57370 | 14.13 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 331284 | 74762 | 18.41 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 269415 | 136632 | 33.65 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 218349 | 187697 | 46.23 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 255710 | 150337 | 37.02 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 158220 | 247827 | 61.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 95976 | 310070 | 76.36 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 31714 | 374333 | 92.19 |
ID | puse | ppcrmax | pregist | phome | total infectors | reductions | reduction rate[%] |
snoapp | 0.000 | 0.000 | 0.000 | 0.000 | 406046 | 0 | 0.00 |
s1actual | 0.193 | 0.044 | 0.134 | 0.300 | 406032 | 14 | 0.00 |
s2actual | 0.193 | 0.044 | 0.134 | 0.600 | 406017 | 29 | 0.01 |
s3actual | 0.193 | 0.044 | 0.134 | 0.900 | 406002 | 44 | 0.01 |
s160 | 0.600 | 0.044 | 0.134 | 0.300 | 405904 | 142 | 0.04 |
s260 | 0.600 | 0.044 | 0.134 | 0.600 | 405762 | 284 | 0.07 |
s360 | 0.600 | 0.044 | 0.134 | 0.900 | 405620 | 426 | 0.11 |
s460 | 0.600 | 0.300 | 0.300 | 0.300 | 403850 | 2196 | 0.54 |
s560 | 0.600 | 0.300 | 0.300 | 0.600 | 401664 | 4382 | 1.08 |
s660 | 0.600 | 0.300 | 0.300 | 0.900 | 399488 | 6558 | 1.62 |
s760 | 0.600 | 0.600 | 0.600 | 0.300 | 397322 | 8724 | 2.15 |
s860 | 0.600 | 0.600 | 0.600 | 0.600 | 388757 | 17290 | 4.26 |
s960 | 0.600 | 0.600 | 0.600 | 0.900 | 380347 | 25699 | 6.33 |
s1060 | 0.600 | 0.900 | 0.900 | 0.300 | 386640 | 19406 | 4.78 |
s1160 | 0.600 | 0.900 | 0.900 | 0.600 | 368020 | 38026 | 9.37 |
s1260 | 0.600 | 0.900 | 0.900 | 0.900 | 350158 | 55888 | 13.76 |
s190 | 0.900 | 0.044 | 0.134 | 0.300 | 405726 | 320 | 0.08 |
s290 | 0.900 | 0.044 | 0.134 | 0.600 | 405407 | 640 | 0.16 |
s390 | 0.900 | 0.044 | 0.134 | 0.900 | 405087 | 959 | 0.24 |
s490 | 0.900 | 0.300 | 0.300 | 0.300 | 401119 | 4927 | 1.21 |
s590 | 0.900 | 0.300 | 0.300 | 0.600 | 396243 | 9803 | 2.41 |
s690 | 0.900 | 0.300 | 0.300 | 0.900 | 391417 | 14630 | 3.60 |
s790 | 0.900 | 0.600 | 0.600 | 0.300 | 386640 | 19406 | 4.78 |
s890 | 0.900 | 0.600 | 0.600 | 0.600 | 368020 | 38026 | 9.37 |
s990 | 0.900 | 0.600 | 0.600 | 0.900 | 350158 | 55888 | 13.76 |
s1090 | 0.900 | 0.900 | 0.900 | 0.300 | 363485 | 42562 | 10.48 |
s1190 | 0.900 | 0.900 | 0.900 | 0.600 | 324726 | 81321 | 20.03 |
s1290 | 0.900 | 0.900 | 0.900 | 0.900 | 289464 | 116582 | 28.71 |
sbest | 1.000 | 1.000 | 1.000 | 1.000 | 226108 | 179938 | 44.31 |