1
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Animal Models for COVID-19 Therapeutic Development: Where We Are and Where We Need to Go

      discussion

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which has spread worldwide and reached a pandemic level within several months (Bedford et al., 2020; Li et al., 2020; Hu et al., 2021). Globally, scientists are trying to understand and defeat COVID-19 using animal studies to explore potential therapeutics. Vaccination remains the best choice to protect susceptible populations; therefore, many institutions and companies have developed vaccines against SARS-CoV-2 infection and many therapeutics are under clinical investigations. For these undertakings, it is essential to use suitable COVID-19 animal models (Caldera-Crespo et al., 2021; Munoz-Fontela et al., 2022). Essentials For Animal Models of COVID-19 Animal models are usually used in medical experimentation as an alternative to avoid potential hazards in humans (Liu and Fan, 2017). Appropriate COVID-19 animal models should be a very powerful tool in the study of the onset, development and culmination of this disease and provide helpful knowledge for its management. Making and choosing good models for COVID-19 requires taking full consideration of the following: (1) the animals should be susceptible to SARS-CoV-2, especially easily infected by the virus via the respiratory route; (2) the characteristics of the induced phenotypes or manifestations are similar to those of human COVID-19 patients, such as fever, coughing and myalgia or fatigue, pneumonia detected by computed tomography (CT) examination, along with sex- or age-related differences in severity (Huang et al., 2020); (3) immune responses should be similar to those in human coronavirus infections; and (4) the animals should have an appropriate cost. When testing vaccine protection effects, recovered infected animals should gain immunity to resist reinfection. These principles should be used for creating new models or evaluating candidate models for the development of COVID-19 therapeutics. Due to the similarity of SARS-CoV-2 to other two beta coronaviruses, SARS-CoV-1 and Middle Eastern respiratory syndrome coronavirus (MERS-CoV), lessons from SARS-CoV-1 vaccine development can be helpful for accelerating SARS-CoV-2 vaccine development (Amanat and Krammer, 2020). Scientists have designed several SARS-CoV-2 vaccines to prevent COVID-19. Although the SARS-CoV-2 vaccine did not catch up with the first wave of the pandemic, the application of the vaccine has reduced the severe incidence of COVID-19 patients. Several faithful animal models for studying the pathogenesis of SARS-CoV-2 and its vaccine development have recently been created by scientists and have shed light on COVID-19 research. Here, we provide a brief review of these models, summarize what we have learned, and discuss their usefulness for testing the immunological response of vaccine injection and post-challenge safety. Non-Human Primate Models of COVID-19 Non-human primates (the rhesus macaque or Macaca mulatta, cynomolgus monkey, or Macaca fascicularis, common marmoset or Callithrix jacchus and African green monkeys or Chlorocebus aethiops) have been used to explore the pathogenesis and test the potential therapies of SARS-CoV-2 (Hartman et al., 2020; Lu et al., 2020; Rockx et al., 2020; Yu et al., 2020; Woolsey et al., 2021). All the above mentioned primates can be infected by experimental inoculation with SARS-CoV-2. The rhesus macaque is the most susceptible to SARS-CoV2 infection, followed by the cynomolgus monkey and the common marmoset (Lu et al., 2020). The viral replication state of nasopharyngeal swabs, anal swabs and lungs in old rhesus macaques was more active than that in young monkeys for 14 days after SARS-CoV-2 challenge (Yu et al., 2020). In SARS-CoV-2-infected macaques, viruses were discharged from the nose and throat in the absence of clinical symptoms and could be detected in both type I and II pneumocytes in the foci of diffuse alveolar damage and ciliated epithelial cells of the nasal, bronchial, and bronchiolar mucosae (Rockx et al., 2020). SARS-CoV-2 also causes pulmonary infiltrates in these monkeys, a hallmark of human disease shown in CT scans, which can be detected by chest radiographs in all infected rhesus macaques (Munster et al., 2020). Another important finding is that monkeys that have recovered from SARS-CoV-2 infection cannot be infected again in the near future (Yu et al., 2020). This finding also provided an important clue for antibody-mediated protection, suggesting that COVID-19 can be prevented through either natural infection or vaccination. Autopsy examinations revealed that the lungs, throat, bronchi, and spleen were infected in both rhesus monkeys and cynomolgus monkeys (Lu et al., 2020; Yu et al., 2020). These observations suggested that the rhesus monkey and cynomolgus monkey models can be used to evaluate vaccines and drugs to treat or prevent COVID-19. Gao et al. developed a purified inactivated SARS-CoV-2 virus vaccine candidate (PiCoVacc) and then conducted a challenge experiment in rhesus monkeys. They reported that vaccine immunization significantly reduced the pathological changes in the lungs of rhesus monkeys and the viral titers were also significantly decreased whereas no antibody-dependent enhancement was observed (Gao et al., 2020). A recent report showed that African green monkey (AGM), another non-human primate, had robust SARS-CoV-2 replication and developed pronounced respiratory disease (Hartman et al., 2020; Woolsey et al., 2021). Shedding of SARS-CoV-2 from both respiratory and gastrointestinal tracts was also documented in AGMs, which may mimic human COVID-19 cases (Hartman et al., 2020). Therefore, AGM may provide another option for COVID-19 therapeutics and vaccine evaluation. These data suggest that monkey models are useful to evaluate vaccines and therapeutics for treating or preventing COVID-19. The advantages and disadvantages of non-human primates compared with those of other animal models are summarized in Table 1. However, high costs and ethical issues may strictly limit the widespread use of non-human primate models in most institutions. Table 1 Comparison of animal models and human COVID-19 features. Human Primates Mouse (hACE2 Tg) Ferret Hamster ACE2R similarity with human Same High Low (wide-type) High High Susceptibility + + + + + Respiratory transmission + + + + + Pneumonia + + Mild + + Severity differences (sex-related) + NR NR NR NR Severity differences (age-related) + + NR NR NR Resistant to reinfection ± + NR NR NR Cost Extremely high Low Middle Low Animal size Big Small Middle Small Animal sources Limited Easy Limited Easy Widely used ± + – – Reproductive cycle Long short Long short NR, not reported. Rodent Models of COVID-19 The genetically manipulated mouse model is relatively cheap and easy to use (Korner et al., 2020). It has been reported that SARS-CoV-2 infection can occur through the angiotensin converting enzyme II receptor (ACE2R) for cellular entry (Zhou et al., 2020). Since 11 of the 29 amino acids in the critical region of the human ACE2R receptor are different from those of the mouse, SARS-CoV-2 is unable to naturally infect mice. Therefore, human ACE2R transgenic mice were developed and analyzed for potential application in the study of COVID-19 (Netland et al., 2008; Bao et al., 2020). Although hACE2R transgenic mice showed signs of weight loss and interstitial pneumonia after infection, the symptoms were mild and much different from those of humans (Bao et al., 2020). The hACE2R transgenic mice developed by Netland et al. also exhibited brain infection of SARS-CoV-2, which is rare in humans (Netland et al., 2008). These drawbacks may puzzle or mislead researchers for interpretation of their studies. The development of lung cell-specific expression of the hACE2R gene in transgenic or knock-in models without endogenous mouse ACE2R gene may help to broaden the use of mouse models in COVID-19 studies in the future. Except for modifying the mouse ACE2R for SARS-CoV-2 infecting host cells, another reverse genetics strategy was to design a recombinant mouse-adapted SARS-CoV-2, designated as SARS-CoV-2 MA, which can use mouse ACE2R for entry into cells (Dinnon et al., 2020; Leist et al., 2020). SARS-CoV-2 MA was reported to successfully infect and replicate in the upper and lower airways in both adult and aged BALB/c mice (Dinnon et al., 2020). SARS-CoV-2 MA caused more severe disease in aged mice, which resembles the human features of COVID-19, and exhibited more clinically relevant phenotypes than those seen in Hfh4-ACE2R transgenic mice (Dinnon et al., 2020). This model provided a powerful tool for the study of SARS-CoV-2 pathogenesis and has been used to evaluate vaccine and antiviral therapeutics performance (Leist et al., 2020). As rats are unsusceptible to SARS-CoV-2, they are not used to study of COVID-19. However, Syrian hamster ACE2R proteins are reported to be highly similar to human ACE2R with 3–4 mutations at the interface (Chan et al., 2020). After being infected with SARS-CoV-2, hamsters showed weight loss, drowsiness, hairiness, hunched back, and shortness of breath. Olfactory and taste dysfunctions are common in mildly symptomatic COVID-19 patients. The food-searching behavioral test showed that anosmia may happen in SARS-CoV-2-infected hamsters, and the level of anosmia was associated with olfactory epithelium damage (Reyna et al., 2022). In addition, high titers of SARS-CoV-2 were found in the hamster lungs and intestines. The phenotypes of human upper and lower respiratory tract infections are very similar to those of hamsters, so this model benefits scientists who like to use small animals to perform COVID-19 research or therapeutic screening (Sia et al., 2020). Other Animal Models of COVID-19 Ferrets, another small animal, have special value in the research of virus-related diseases. After being infected with respiratory viruses, ferrets can show symptoms similar to those of humans. For example, ferrets infected with influenza virus can even sneeze like humans, and can easily transmit the virus to other ferrets (Herfst et al., 2012). SARS-CoV-2 can infect ferrets through the respiratory tract and can replicate in infected cells (Shi et al., 2020). The temperature of the infected ferrets rises slightly and infection does not cause serious illness or death. Although fatalities were not observed, SARS-CoV-2-infected ferrets shed the virus in nasal washes, saliva, urine, and feces up to 8 days post-infection (Kim et al., 2020). Ferrets give birth once per year, with 6–8 l, so they do not breed as rapidly as mice. To a certain extent, the slow reproduction speed may limit the wide application of ferrets. Similar to ferrets, farmed minks are also susceptible to coronaviruses. On farms, minks are infected with SARS-CoV-2 and can transmit it to humans (Enserink, 2020; Oreshkova et al., 2020; Sharun et al., 2021). Laboratory evidence also showed SARS-CoV-2 can be transmitted among minks and resulted severe respiratory syndrome (Shuai et al., 2021). Among middle- or large-sized animals, it was reported that SARS-CoV-2 can replicate efficiently in cats, with younger cats being more permissive and perhaps more importantly, the virus can transmit between cats via the airborne route (Shi et al., 2020). Dogs exhibit low susceptibility, whereas livestock, including pigs, chickens, and ducks, are not susceptible to SARS-CoV-2 (Shi et al., 2020). Previous reviews also have discussed the potentials of these animal species in COVID-19 studies (Munoz-Fontela et al., 2020; Sharun et al., 2020; Shou et al., 2021). Discussion And Perspectives The currently reported COVID-19 animal models were summarized in Table 1. The advantages and disadvantages of the COVID-19 animal models that are currently available was discussed in this and previous study (Shou et al., 2021). To date, it seems that no animal model is perfect when evaluating potential drugs and vaccines (Munoz-Fontela et al., 2022). The most useful animal model is the rhesus macaque, but because of the substantial cost and specific facility requirements, the monkey model is unlikely to be widely used in common laboratories. The ferret model has special applicability in the study of respiratory viruses, but the low breeding rate and the lack of suitable facilities make it difficult for wide use in COVID-19 studies. The limitations of COVID-19 animal models are also apparent (Sharun et al., 2020). First, no model can fully mimic human COVID-19, and the findings of each model are needed to confirm with other animal models and to increase its credibility. Second, although the non-human primate model can better represent human COVID-19, it faces stricter ethical review and is not easy to be widely used. Mice are cheap and there are many genetically modified models available, but their immune system responses differ from those of humans. Humanized mouse models may become a feasible study tool for COVID-19 research in the future. At present, various genetically engineered mouse models are currently on the way, and hopefully, these new models will provide a unique research tool to fight against COVID-19 in the future. Author Contributions JF and EL designed this project. SZ wrote the paper. All authors contributed to the article and approved the submitted version. Funding This study was supported by grants from the Natural Science Foundation of Shaanxi Province (Grant Nos. 2020PT-004 and 2020PT-001). Conflict of Interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Publisher's Note All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China

          Summary Background A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by WHO and the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0–58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0–13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            A pneumonia outbreak associated with a new coronavirus of probable bat origin

            Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats 1–4 . Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans 5–7 . Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor—angiotensin converting enzyme II (ACE2)—as SARS-CoV.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Early Transmission Dynamics in Wuhan, China, of Novel Coronavirus–Infected Pneumonia

              Abstract Background The initial cases of novel coronavirus (2019-nCoV)–infected pneumonia (NCIP) occurred in Wuhan, Hubei Province, China, in December 2019 and January 2020. We analyzed data on the first 425 confirmed cases in Wuhan to determine the epidemiologic characteristics of NCIP. Methods We collected information on demographic characteristics, exposure history, and illness timelines of laboratory-confirmed cases of NCIP that had been reported by January 22, 2020. We described characteristics of the cases and estimated the key epidemiologic time-delay distributions. In the early period of exponential growth, we estimated the epidemic doubling time and the basic reproductive number. Results Among the first 425 patients with confirmed NCIP, the median age was 59 years and 56% were male. The majority of cases (55%) with onset before January 1, 2020, were linked to the Huanan Seafood Wholesale Market, as compared with 8.6% of the subsequent cases. The mean incubation period was 5.2 days (95% confidence interval [CI], 4.1 to 7.0), with the 95th percentile of the distribution at 12.5 days. In its early stages, the epidemic doubled in size every 7.4 days. With a mean serial interval of 7.5 days (95% CI, 5.3 to 19), the basic reproductive number was estimated to be 2.2 (95% CI, 1.4 to 3.9). Conclusions On the basis of this information, there is evidence that human-to-human transmission has occurred among close contacts since the middle of December 2019. Considerable efforts to reduce transmission will be required to control outbreaks if similar dynamics apply elsewhere. Measures to prevent or reduce transmission should be implemented in populations at risk. (Funded by the Ministry of Science and Technology of China and others.)
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Microbiol
                Front Microbiol
                Front. Microbiol.
                Frontiers in Microbiology
                Frontiers Media S.A.
                1664-302X
                24 June 2022
                2022
                24 June 2022
                : 13
                : 907406
                Affiliations
                [1] 1Laboratory Animal Center, Health Science Center of Xi'an Jiaotong University , Xi'an, China
                [2] 2Institute of Molecular Virology, Health Science Center of Xi'an Jiaotong University , Xi'an, China
                [3] 3Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi , Chuo, Japan
                Author notes

                Edited by: Anna Kramvis, University of the Witwatersrand, South Africa

                Reviewed by: Junki Maruyama, University of Texas Medical Branch at Galveston, United States; Khan Sharun, Indian Veterinary Research Institute (IVRI), India

                *Correspondence: Enqi Liu liuenqi@ 123456xjtu.edu.cn

                This article was submitted to Virology, a section of the journal Frontiers in Microbiology

                Article
                10.3389/fmicb.2022.907406
                9263605
                35814648
                e433d5d3-1b68-45ce-aff1-d6a0ba7a35c5
                Copyright © 2022 Zhao, Fan and Liu.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 29 March 2022
                : 30 May 2022
                Page count
                Figures: 0, Tables: 1, Equations: 0, References: 34, Pages: 4, Words: 3485
                Funding
                Funded by: Natural Science Foundation of Shaanxi Province, doi 10.13039/501100007128;
                Award ID: 2020PT-001
                Award ID: 2020PT-004
                Categories
                Microbiology
                Opinion

                Microbiology & Virology
                animal models,covid-19,therapeutics,vaccines,humanized mouse model
                Microbiology & Virology
                animal models, covid-19, therapeutics, vaccines, humanized mouse model

                Comments

                Comment on this article