Next Article in Journal
Outreach Efforts to Prevent Newcastle Disease Outbreaks in Southern California
Next Article in Special Issue
SARS-CoV-2 Production, Purification Methods and UV Inactivation for Proteomics and Structural Studies
Previous Article in Journal
Avian IRF1 and IRF7 Play Overlapping and Distinct Roles in Regulating IFN-Dependent and -Independent Antiviral Responses to Duck Tembusu Virus Infection
Previous Article in Special Issue
Neutralizing Antibody Response to Sarbecovirus Is Delayed in Sequential Heterologous Immunization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell and Animal Models for SARS-CoV-2 Research

by
Eloïne Bestion
1,2,3,
Philippe Halfon
1,2,3,
Soraya Mezouar
1,2,3,*,† and
Jean-Louis Mège
1,2,*,†
1
Microbe Evolution Phylogeny Infection, Institut pour la Recherche et le Developpement, Assistance Publique Hopitaux de Marseille, Aix-Marseille University, 13005 Marseille, France
2
Institue Hospitalo, Universitaire Mediterranée Infection, 13005 Marseille, France
3
Genoscience Pharma, 13005 Marseille, France
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2022, 14(7), 1507; https://doi.org/10.3390/v14071507
Submission received: 24 May 2022 / Revised: 29 June 2022 / Accepted: 5 July 2022 / Published: 9 July 2022
(This article belongs to the Special Issue SARS-CoV-2 and Other Coronaviruses)

Abstract

:
During the last two years following the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, development of potent antiviral drugs and vaccines has been a global health priority. In this context, the understanding of virus pathophysiology, the identification of associated therapeutic targets, and the screening of potential effective compounds have been indispensable advancements. It was therefore of primary importance to develop experimental models that recapitulate the aspects of the human disease in the best way possible. This article reviews the information concerning available SARS-CoV-2 preclinical models during that time, including cell-based approaches and animal models. We discuss their evolution, their advantages, and drawbacks, as well as their relevance to drug effectiveness evaluation.

1. Introduction

In December 2019, the emergence of the coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), rapidly evolved into an unexpected pandemic [1]. COVID-19 is a still-expanding global disease involving more than 539,000,000 cases, including over 6,300,000 deaths worldwide [2]. SARS-CoV-2, targets the respiratory tract, focusing on epithelial cells and pneumocytes [3]. Transmission occurs through direct, indirect, or close contact with infected people via saliva, respiratory droplets, or secretions [4]. The SARS-CoV-2 spike (S) protein engages angiotensin-converting enzyme-2 (ACE2) as an entry receptor and is then primed by the transmembrane protease serine-2 (TMPRSS2) [5], two major factors enabling viral entry. Symptoms associated with SARS-CoV-2 broadly include dry cough, fever, headache, and weakness. The respiratory symptoms are remarkably heterogeneous, ranging from almost asymptomatic to acute respiratory distress syndrome that may lead to patient death [6]. A significant portion of patients experience gastrointestinal manifestations, such as diarrhea, vomiting, and loss of appetite [7]. COVID-19 affects the lung, digestive tract, and nervous system and causes multi-organ pathologies associated with immune system dysregulation [8,9]. Clinical reports have further disclosed prolonged and persistent clinical consequences after SARS-CoV-2 infection, including pulmonary fibrosis, thromboembolic events, chest pain, or neurological dysfunctions [10]. The world is thus facing an emergency situation, with medical and societal-associated impacts being increasingly appreciated but still not adequately managed.
To tackle this exceptional situation, a drug-repurposing approach was first attempted before giving way to innovative solutions. In both cases, relevance of the preliminary preclinical findings have been evaluated in more than 6800 clinical trials [11] through the end of November 2021. Trying to curb the COVID-19 health crisis, the clinical translation phase has thus been achieved in record time, shaking up research and medical habits. Many independent and diverse preclinical models have therefore been employed to understand the SARS-CoV-2 infection. In vitro models are compulsory for studying virus biology under highly controlled conditions. They further allow accomplishing substantial drug screening assays. Nevertheless, in vitro models have failed to demonstrate the complexity of the human body and whole cell interactions [12]. In contrast, in vivo models support the elucidation of the COVID-19 pathogenesis, reflecting the diversity of clinical manifestations. These models further elucidate the details of the host-immune response [13]. Extensive preclinical exploration can identify a lead candidate, select the best formulation, and determine the route, duration, and frequency of administration, which aids in supporting the upcoming clinical trial design. To date, disappointing outcomes have been reported regarding COVID-19 trials, as they have been failing to benefit patients [11,14]. An increased failure rate could be attributed to the limitations of time and means associated with the pandemic period. Indeed, several repurposed drug candidates have entered clinical trials even before showing efficacy in animal models [15,16,17]. Some preclinical models were missing SARS-CoV-2 receptors/mechanisms [5], contributing to a partial or inadequate demonstration of the disease and leading to impaired conclusions [18,19,20]. The lack of relevant models and absence of consensus regarding the endpoints monitored [21] reaffirms how important the proper characterization of the preclinical forces deployed is.
In this review, we provide an overview of the different models employed so far for SARS-CoV-2 infection comprehension and therapeutic testing. We discuss their strengths and limitations, especially their involved receptors and tissue origins, as well as their correct recapitulation of human clinical signs. Emerging techniques applied to SARS-CoV-2 research are discussed. We also discuss the importance of the identification of animal reservoirs to guarantee effective disease control. Finally, we outline the indispensable necessity of compiling models to correctly summarize SARS-CoV-2 pathophysiology and to consequently limit clinical trial failures.

2. Cell-Based Approaches

2.1. Cell Models

2.1.1. Immortalized Cell Lines

Similarly to SARS-CoV [22], the ACE2 receptor was found to be necessary for SARS-CoV-2 entry [5]. A group of cell lines known to express high ACE2 levels were put forward from the outset of the pandemic (Table 1). One such cell line is Vero, which has been broadly used to isolate, propagate, and investigate clinical isolates [23,24,25,26]. This cell line has indeed been proven to be highly susceptible and permissive to SARS-CoV-2 replication [27,28,29]. In contrast to Vero WT cells, the Caco-2 cell line also expresses the SARS-CoV-2 co-receptor TMPRSS2 endogenously, although the TMPRSS2-overexpressing Vero cell model successfully led to an increase in SARS-CoV-2 replication compared to wildtype [30,31].
Vero cells nevertheless do not well-recapitulate the pathophysiological properties of SARS-CoV-2, as they originate from the kidney epithelial cells of an African green monkey [25]. More interestingly, Caco-2 and Calu-3 cells were obtained from human colorectal and lung adenocarcinomas, respectively. They are reported to strongly express both ACE2 and TMPRSS2 receptors [69,70] and to present a strong titer of SARS-CoV-2 particles [13,28] compared to other cells lines originating from the lung and bronchus [29,35,49,71,72] (Table 1). Caco-2 and Calu-3 cells have therefore been widely used and have revealed relevant pathways for SARS-CoV-2 infection [29,37]. The Calu-3 cells are additionally able to polarize, potentially explaining their enhanced susceptibility to SARS-CoV-2 infection [73,74].
It has been found that 2D cell-based models are relevant in understanding the physiopathology and are suitable for high-throughput screening of drugs against SARS-CoV-2. The most frequently reported criterion to quantify the in vitro antiviral potency of compounds is the EC50; i.e., the drug concentration in cell culture media that provides 50% maximal antiviral activity. It should be noted that many experimental factors can affect the estimation of EC50, including viral strain, multiplicity of infection, drug duration, serum percentage, and cell line model chosen. Chloroquine (CQ), an anti-malarial drug, and its derivative hydroxychloroquine (HCQ) were the first drugs evaluated for their efficacy against SARS-CoV-2 infection. Comparison of the various preclinical studies and clinical outcomes rapidly spotlighted the importance of the features of each investigated model. CQ and HCQ first obtained very attractive EC50 results in Vero cells and thus were widely adopted to treat COVID-19 patients [75,76,77]. They were registered in 90 clinical trials worldwide during 2020, sometimes in combination with other drugs [78,79]. However, in the beginning of summer 2020, Hoffmann et al. were the first to demonstrate that CQ does not inhibit SARS-CoV-2 infection of human lung cells [32]. They revealed that CQ efficiently blocked SARS-CoV-2 infection in Vero cells [76,80,81], but not in Calu-3 and Vero/TMPRSS2+ cells. CQ inhibition of viral entry was inefficient and absent, suggesting that CQ is unhelpful in ACE2+/TMPRSS2+ models, and therefore highlighting the importance of the selected model in evaluating compounds of interest. This was followed by preclinical evidence that did not support broad use of HCQ in COVID-19 [82,83].
Cell line models provided valuable insights regarding virus replication kinetics and associated cellular tropism and interactors. A limitation to the use of immortalized cell lines is that they might not entirely reflect the SARS-CoV-2 infectious mechanism and infection rate, lacking population heterogeneity and for some, polarization. Their proliferation rate may also affect virus transfection efficiency. It is also well-established that most of the available cell lines display common mutations including TP53, KRAS, or EGFR, considered to regulate SARS-CoV replication [84,85,86]. Cell lines are nonetheless an essential study model in the SARS-CoV-2 compound screening step, with high throughput and reproducible results.

2.1.2. Primary Cells

Concerning pathogens that induce respiratory symptoms, primary cells originating from human airway epithelia (HAE) have historically been exploited for coronavirus isolation and cultivation [87,88], as well as to examine their tropism. Indeed, upon HAE cultivation at an airway-liquid interface (HAE-ALI), cells differentiated on the porous support exhibiting basal and apical sides. The tissue created is composed of basal, goblet, and ciliated cells, as in physiological conditions [89]. Hence, Hou et al. were able to demonstrate that human nasal/bronchial/alveolar epithelial cultures are permissive to SARS-CoV-2 infection [90]. The investigation of SARS-CoV-2 titers and ACE2 receptor expression from each cell type moreover revealed that the upper respiratory tract is more susceptible to infection than the lower one. Zhu et al. additionally showed that both ciliated and secretory cells from HAE are permissive to SARS-CoV-2 [91]. They also observed that the virus caused a plaque-like cytopathic effect in HAE cultures associated with apoptosis, destruction of epithelium integrity, and beaded changes, as well as lack of ciliary movement. Moreover, the SARS-CoV-2 infection in this model was long-lasting, with recurrent replication peaks [54].
The characterization of the antiviral activity of lead compounds on primary cells remains crucial and highly informative compared to immortalized cell line. Pruijssers et al. showed that remdesivir strongly inhibits SARS-CoV-2 replication in HAE cultures without causing cytotoxicity [92]. In contrast, favorable antiviral results with HCQ in Vero cells were invalidated in the HAE model cultured in an ALI [93]. The study additionally demonstrated that HCQ did not protect the integrity of epithelial tissue during infection. Since such primary cell cultures reproduce major structural features of human respiratory epithelia, drug screening outcomes are more representative.
Another limitation of the model is the absence of immune cells. Considering host-pathogen interactions and immune response following SARS-CoV-2 infection, standard and co-culture systems with induced pluripotent stem cell (iPSC)-derived cells seemed an appropriate strategy [94,95]. Interestingly, studies on the iPSC-derived human lung epithelial system showed that SARS-CoV-2 infection and propagation lead to transcriptional alterations in infected cells [96,97], characterized by a shift to an inflammatory phenotype and changes in cell function [98]. Indeed, the transcriptome of iPSC-derived cells is comparable to their respective primary homologs, allowing them to respond to immune stimuli in a similar way [84]. Histocompatibility issues occurring when examining human immune cells with other cell types were tackled by Duan et al. while co-culturing lung and macrophage-differentiated PSC [99]. Their modeling of lung cell and macrophage crosstalk during SARS-CoV-2 infection revealed synergistic effects of anti-inflammatory macrophages with ACE2 inhibition [99]. The good modeling of cell-intrinsic responses to the immune response caused by SARS-CoV-2 propels iPSC-derived cells as a useful model for drug screening repurposing strategy.
Taken together, primary cell models appear to be essential to validate previous obtained results and confirm potential drug candidates. Despite their more realistic host-virus interactions, primary cells are often limited by the availability of donor materials. It is further evident that primary cells have a limited proliferation capacity, which is a major obstacle to large drug screening campaigns [100]. Another important caveat regarding human lineages derived in vitro from iPSCs is their immature or fetal phenotypes, possibly confounding disease modeling [97]. However, since studies revealed that genetic defects of type I immunity drive COVID-19 severity [101,102], iPSC obtained from a COVID-19 patient tissue biopsy provide useful new tools in developing or testing specific drugs with more personalized-therapy strategies.

2.2. 3D Models

Interestingly, 3D-cell models like organoids are ex vivo tissue constructs derived from primary tissues and embryonic stem cells (ESCs) or iPSCs, and exhibit multiple cell types once differentiated [103,104]. They are self-organized assemblies that mimic complex environmental interactions, tissue structures, and functions of the investigated organs [105]. As SARS-CoV-2 infection not only targets the lungs [106], but also the kidney [107], liver [108], and the cardiovascular systems [109], with almost 25% of patients exhibiting gastrointestinal illness [59], organoid modeling may be a useful model in this context. Interestingly, RNA-seq data demonstrated spontaneous expressions of ACE2 and TMPRSS2 in lung [110], colon [111], kidney [112,113], liver ductal [114], and brain organoids [115]. Combined with these observations, bronchioalveolar [116], distal lung [60], kidney, blood vessel [61], small intestinal [62,63], and even liver ductal organoids [114] were reported to be highly permissive to SARS-CoV-2 infection. The use of this model has been of great interest in better understanding the pathophysiology of SARS-CoV-2 infection in human tissues from different locations.
  • Organoid models provided understanding of the neurological impairment associated with SARS-CoV-2 infection. Ramani et al. reported that SARS-CoV-2 is able to target human neurons [117]. They observed an altered distribution of the Tau protein from axons to soma, hyperphosphorylation, and apparent neuronal death, revealing the potential neurotoxic effect of SARS-CoV-2, including Alzheimer’s disease.
  • Makovoz et al. generated human eye organoids and found that ocular cells express both ACE2 and TMPRSS2 receptors, and are susceptible to SARS-CoV-2 infection [118], contesting previous observations [119,120]. They also pointed out viral tropism, as higher viral titers were observed in the border of the cornea compared to the central corneal zone.
  • A crucial question was raised by Triana et al., who observed no correlation between ACE2 expression level and the copy number of the SARS-CoV-2 genome in intestinal organoids [121]. They additionally revealed that ACE2 expression is downregulated upon SARS-CoV-2 infection. This consequently questions using ACE2 expression as the only key element for anticipating the infectability of cell types.
  • The organoid model may also allow partially investigating ancillary pathways of SARS-CoV-2 infection, such as inflammation and immunity. Even though the resident immune cell activity of the mucosal/epithelial tissue can be monitored, most organoid models lack relevant immune cells; e.g., macrophages and B and T cells that modulate severe disease [122,123]. Focusing on HAE and airway organoid combined models, a robust induction of chemokines similar to what is seen in COVID-19 patients was observed [36,59]. A next important step would be to complexify organoid models using lung tissues from SARS-CoV-2 infected patients and adding their own key immune cells to investigate different forms and phases of the disease.
The use of organoid models to screen drugs has been little explored. Chen et al. performed drug screening assays covering around 15,000 compounds and demonstrated that only a handful of treatments efficiently inhibited SARS-CoV-2 entry in lung organoids [59]. Among the investigated compounds, they highlighted imatinib, which was supported in a clinical trial [124], while results obtained with Caco-2 model did not promote this hypothesis [125]. To the same extent, intestinal organoid models confirmed the absence of benefit of HCQ treatment in SARS-CoV-2 infection [126]. Organoids have become a pertinent and reliable tool to invalidate/promote potential drug candidates before moving to animal models or clinical trials. Patient-derived organoids also enable screening of efficient therapeutic agents within the framework of personalized medicine, especially with cancer patient-derived organoids [127]. As genetic predisposition could be a risk factor in developing severe COVID-19 [101,102], the genesis of mutant organoids for genes previously identified as implicated in coronavirus biology could be of interest to better understand the pathophysiology of SARS-CoV-2 infection. More representative 3D-structured models and personalized medicine capability are organoid features essential to consider regarding the certification of the antiviral activity of a compound.
Organoids nevertheless lack a vasculature system [104]. Organ-on-chip (OOC) technology is a physiological organ biomimetic system built on a 3D microfluidic cell culture chip. The structure simulates tissue interfaces, mechanical stimulation, and fluid exchanges, as in living organs. Si et al. developed a model including microfluidic bronchial-airway-on-a-chip lined by highly differentiated human bronchial-airway epithelium and pulmonary endothelium [64]. The authors set up a model of SARS-CoV-2 infection with strain-dependent virulence, cytokine production, and the recruitment of immune cells. Others have reproduced key features of the alveolar-capillary barrier based on a human alveolar chip through the co-culture of human alveolar epithelium, microvascular endothelium, and immune cells under fluidic flow [128]. This model revealed that human alveolar epithelial cells were more permissive to SARS-CoV-2 infection than microvascular endothelial cells and provides a new device to screen antiviral drugs. Si et al. used these airway chips containing human lung epithelial primary cells from healthy donors that express high levels of ACE2 and TMPRSS2 to investigate the inhibitory activities of clinically approved drugs against SARS-CoV-2 [66]. They reported that when drugs are administered under flow similar to the maximal ones reported in clinical studies, few drugs significantly inhibited viral entry. However, OOC technology suffers from relatively low throughput [129,130].
3D technology originating from primary cells has emerged as a powerful device. Major benefits are the full range of differentiated cell types, as are present in the target organ, and self-organization, leading to close mimesis of functional and architectural features of corresponding tissues. Organoids also reduce experimental complexity compared to animal models and are further compliant with real-time and kinetic techniques. They could consequently become a new standard for SARS-CoV-2 investigations, as they are more informative than in vitro models and a potent alternative to animal models. However, organ 3D modeling remains complex, with a quite high failure rate.

3. SARS-CoV-2 Infection in Animals

3.1. SARS-CoV-2 Animal Reservoirs

3.1.1. Wildlife

SARS-CoV-2 isolated from horseshoe bats [30,64] and Malayan pangolins [66,128] exhibited high sequence similarities compared to the whole-SARS-CoV-2 genome. Bats and pangolins were therefore initially considered as a possible source and intermediate hosts for SARS-CoV2, respectively. Interestingly, fruit bats were found susceptible to SARS-CoV-2 infection using intranasal inoculation, presenting mild clinical signs including rhinitis with congener transmission [131]. White-tailed deer express ACE2 that shares a high degree of similarity with humans, and they are highly susceptible to SARS-CoV-2 infection [132,133]. Although they do not present overt clinical symptoms following SARS-CoV-2 infection, they can transmit the virus through direct contact or in vertical transmission from doe to fetus. Griffin et al. raised the question of a plausible risk of SARS-CoV-2 reverse zoonosis through deer mice [134]. Interestingly, they indicated that deer mouse ACE2 differed from human (h)ACE2 by four amino acid residues known to confer efficient binding to the SARS-CoV-2 S protein. These differences were unlikely to have a prejudicial effect on S binding efficiency. The viral infection was persistent for up to 21 days, and the animals exhibited asymptomatic-to-mild disease, with lesions limited to mild lung pathology with elevated levels of inflammatory cytokines. Fagre et al. further described neurological manifestations in the olfactory bulb and tongue that could affect smell and taste senses, respectively, as in humans [135]. Infected animals were also capable of transmitting SARS-CoV-2 to naïve co-housed congeners through direct contact. SARS-CoV-2 zoo animal infections were first noticed with a Malayan tiger, which spread the disease to congeners and lions in the same zoo [136,137]. Independently, puma [138] and snow leopards [139] were declared infected. Considering the susceptibility of the Felidae family, SARS-CoV-2 transmission to the wild animal population should be greater, as predicted.

3.1.2. Domestic Animals

Cats are highly susceptible to SARS-CoV-2 infection and exhibit asymptomatic-to-moderate symptoms, notably interstitial pneumonia, tracheal and nasal inflammation [106], and even severe respiratory clinical signs [140]. Viral transmission to naive congeners has also been monitored [141,142]. Interestingly, human-to-feline transmission has been reported [143], while cat-to-human transmission has not yet been observed. However, since SARS-CoV-2 infected cats shed high levels of virus and transmission to congeners via droplets is possible, the risk of cat-to-human transmission cannot be ignored. In contrast, dogs have low susceptibility to SARS-CoV-2, probably due to the relatively low ACE2 receptor expression in their respiratory tract [144]. Antibody responses have been detected in infected dogs, and virus sequencing suggested that it was the result of human-to-animal transmission [145]. Although current evidence suggests a low risk for domestic pets transmitting SARS-CoV-2 to humans, their close link to their owners encouraged us to consider them as virus carriers and potential intermediate hosts.

3.1.3. Farm Animals

Pigs, chickens, and ducks have been found to have low or even no susceptibility to SARS-CoV-2 intranasal inoculation [131,146]. While domestic pigs are not a suitable model for COVID-19, significant similarities with humans in terms of anatomy, genetics, and physiology promoted them as a useful model for assessing seroconversion and immunogenicity of the upcoming vaccine candidates. Interestingly, following parental inoculation, seroconversion and the presence of neutralizing antibodies were observed in animals, while none resulted in productive infection [147]. The safety and immunogenicity of AVX/COVID-12-HEXAPRO, a viral vector vaccine, were established using a pig model [148]. In contrast, the mink has been reported to experience the first and only known farm anthropozoonotic outbreak to date [149,150]. Among the 16 Netherlands farms investigated, 49% of workers were PCR-positive due to the breeding conditions and mink natural susceptibility to SARS-CoV-2 infection as initially introduced by humans [150]. Three-dimensional structures of the protein region involved in direct interactions with the SARS-CoV-2 S receptor-binding domain (RBD) of the mink ACE2 was found to be similar to humans [151]. Viral variants with mutations on the S protein were reported from infected mink [152]. Interestingly, SARS-CoV-2 infected minks experienced asymptomatic-to-severe symptoms and even death [153]. The viral load obtained from swabs collected post-mortem demonstrated higher rates in the throat than in the rectal swabs [13]. As minks might become a continual source of human infection, with alarming SARS-CoV-2 variants, this resulted in mass mink culling worldwide. Although almost all previously cited animals cannot be used as standard experimental models, their investigations brought new insights regarding human-animal transmission and monitoring of an unexpected potential permanent reservoir for SARS-CoV-2. Given their susceptibility and the ability of SARS-CoV-2 de novo mutation, two-way zoonotic transmission must not be neglected, as it might provoke the appearance of new or the reappearance of endemic foci due to animal migrations and human travel.

3.2. Animal Models for SARS-CoV-2 Research

In the context of SARS-CoV-2 infection, mice, hamsters, ferrets, and non-human primates (NHPs) have been prominent models (Table 2). Interspecies-related differences are essential aspects to consider, such as host specificity, clinical symptoms, or immune response divergence to evaluate viral pathophysiology.

3.2.1. Mice

Mouse models are currently used to investigate viral pathogenesis and to evaluate both drugs and vaccine responses. As observed for MERS-CoV and SARS-CoV-1 [187,188,189], viral replication was insufficient in wild-type mice using diverse viral strains [28,154,190], due to structural differences in the mouse ACE2 compared to human [191]. Generated in 2007 [191,192], hACE2 transgenic mice were rapidly used during the SARS-CoV-2 pandemic, showing replication of the virus in the lungs leading to local immune cell infiltrates, cytokine storm, and interstitial pneumonia [157]. In contrast, infected K18-hACE2 mice to present weight loss, respiratory distress, and reduced activity, similar to the symptoms observed in humans [157,158,193]. K18-hACE2 [155,157,184] and Hfh4-hACE2 [155] mice models demonstrate encephalitis-related lethality due to viral dissemination in the brain. Viral vector-mediated delivery systems responded to this problem. The AAV-hACE2 model using an adeno-associated virus (AAV) was indeed proposed as a viable model without encephalitis issues, characterized by viral replication until immune clearance, avoiding permanent genetic modification. However, AAV delivery of hACE2 is unspecific, leading to artificial ACE2 expression in non-relevant mouse cell types and generating mouse-to-mouse variation [157,193].
CRISPR/Cas9 knock-in (KI) technology has been used to engineer an hACE2 model [161] that exhibited high viral loads in the lungs, trachea, and brain with no fatalities. Tissue distribution of hACE2 matches clinical signs from COVID-19 patients, making the model highly valuable, contrary to the AAV-hACE2 model. Pathological features of SARS-CoV-2-induced ARDS in humans, including hemorrhage, thrombi, and hyaline membrane-like changes, were however rarely observed in models challenged with nasal inoculation. hACE2-KI mice with intratracheal inoculation of SARS-CoV-2 developed severe form of lung injury [194]. Double hACE2/hTMPRSS2 models were proposed to better recapitulate the precise role of the two proteins during SARS-CoV-2 infection and to better investigate antibody-based therapies, as they will closer resume human-like antibody clearance [195,196,197].
Another way to optimize the mouse model is to use a modified SARS-CoV-2 S protein to enhance binding affinity to the ACE2 mouse [198]. A recombinant strain replicates in the respiratory tract but was cleared within 4-days post-infection, causing mild-to-moderate pneumonia [190]. Leist et al. obtained the SARS-CoV-2 MA10 strain via several passages in mice lungs, in which mutations emerged concerning the S protein, as well as nonstructural proteins and an open reading frame [199]. MA10 infection led to acutely impaired lung function, high levels of proinflammatory cytokines, and non-lethal disease. A mouse-adapted strain, WBP-1, produced lethal lung infections with substitutions increasing the binding affinity to the ACE2 mouse [200]. It has also been suggested to use reverse genetics to reshape the viral RBD, sensitizing mice for infection since the ACE2 mouse protein can be correctly recognized [190]. One caveat of this method is the appearance of genetic mutations that will affect the RB, the primary target site for antibody response.

3.2.2. Hamsters

The Syrian hamster model has already been employed to understand SARS-CoV-1 infection and to test antiviral drugs [201,202,203]. With only 4 amino acids of the critical ACE2 domain differing from the human domain, no genetic manipulations are required for SARS-CoV-2 investigations. This model presents an efficient replication rate in the lungs, with mild-to-severe lung pathological lesions following intranasal virus infection [162,163]. Analysis of lung tissues demonstrated that the organs developed pulmonary inflammation, edema, and cell death [164]. Animals further suffer from weight loss, respiratory distress, and signs of morbidity, such as hunched posture, ruffled fur, and lethargy [164,204]. With a clinical picture similar to that observed in humans, this model has led to a better understanding of the pathophysiological mechanisms (cellular and molecular) of SARS-CoV-2 infection. (1) SARS-CoV-2 infected hamsters exhibited a strong neutralizing antibody protective response to consecutive virus rechallenge [162]. (2) Aged male hamsters developed more severe infection compared to young female hamsters [205]. Moreover, histopathological analysis revealed massive immune cell infiltration into the lungs of young animals compared to older ones. This exuberant innate immune response was linked to the STAT2 signaling that drives viral dissemination in the body, as STAT2-knockout individuals presented lower immune cell infiltration and decreased lung pathology [204]. (3) Airborne and fomite SARS-CoV-2 transmission was observed using the Syrian model. Viral transmission was found to be reduced by the use of surgical mask partitions between cages, limiting respiratory/airborne droplet exchanges and providing the first in vivo experimental evidence to support the potential benefit of wearing masks [206]. More recently, a study explored the different routes of exposure; i.e., contact/fomite/airborne transmission, and found that hamsters presented distinct disease manifestations according to exposure method [207]. Intranasal and aerosol inoculation led to severe respiratory pathology, with an altered shedding profile, while fomite exposure had a delayed shedding pattern associated with milder disease manifestation. Using Syrian hamsters, the Alpha variant was demonstrated to replicate and shed more efficiently in the nasal cavity than other variants, even with a low dose and short duration of exposure [208]. The model additionally showed that a synthetic nanobody targeting RBD [209] and a SARS-CoV-2 neutralizing nanobody [210] protect hamsters from SARS-CoV-2 infection, while favipiravir, a small-molecule drug, was associated with significant toxicity [211]. Recently, the orally or intranasally-delivered Adenovirus type (Ad) 5-vectored SARS-CoV-2 vaccine candidate was demonstrated to have a robust and cross-reactive antibody responses in Syrian hamster model [212]. Animals experienced less lung pathology and airborne transmission compared to control group SARS-CoV-2 challenge. Regarding Omicron variant, Zou et al. showed that prophylactic and therapeutic vaccine-induced ZCB11 administration protects from lung infection in golden Syrian hamsters. Interestingly, ZCB11 also protected animals against the circulating Omicron BA.1 and Delta variants [213].
Bertzbach et al. established Chinese hamsters as a suitable supplemental model for SARS-CoV-2 study, showing pronounced clinical symptoms. The animals indeed present robust viral replication in the upper and lower respiratory tract along with significant weight loss, bronchitis, and prolonged pneumonia associated with acute alveolar damage [214]. The model can spontaneously develop diabetes, making them attractive for investigating COVID-19 with preexisting diabetes [215].
The SARS-CoV-2 infected Roborovski dwarf hamster had a rapid and consistent onset of fulminant clinical disease, including a sudden fall in body temperature, severe acute diffuse alveolar damage, and hyaline microthrombi in the lungs [165]. The authors thus introduced the hamster as a highly susceptible COVID-19 model, having stable and fulminant clinical signs similar to changes described in patients who died from SARS-CoV-2 infection. Such a severe form of COVID-19 has not been reproduced in any experimentally infected animal, positioning this model as a valuable model for screening therapeutics and vaccine candidates in highly susceptible individuals. The therapeutic benefit of molnupiravir in dwarf hamsters infected with omicron variant was evaluated [216]. Interestingly, the authors described a significant virus load reduction in treated males but not in females, highlighting a sex effect regarding the efficacy of the antiviral.

3.2.3. Ferrets

Ferrets have been extensively used for diverse viral pathogenesis. Anatomical and physical ferret characteristics explain why they are generally ideal animals for understanding respiratory virus pathophysiology and mode of action. Ferrets possess an unusually long trachea, permitting easy compartmentation of the upper and lower respiratory tracts [217]. Thus, they are widely employed for studying the transmissibility and tropism of respiratory viruses [218,219,220]. Ferrets have also been employed to study SARS-CoV-1 permissivity and transmission [166,221]. The predominance of alpha 2,6-linked sialic acids in the upper airway epithelia [222,223,224] and the ferret ACE2 critical residues needed for binding to SARS-CoV-2 RBD [225] allow SARS virus attachment patterns to respiratory tissues in a manner similar to humans [226,227]. These characteristics favor high susceptibility to SARS-CoV-2 infection, predominantly in the upper-respiratory-tract location [146,228]. Indeed, after intratracheal virus inoculation, viral RNA was detected in the nasal turbinate, soft palate, and tonsils, but not in the lungs. SARS-CoV-2 vRNA was also detected in rectal swabs, but in lower numbers than nasal samples. Clinical manifestations in ferrets are mainly undetectable-to-mild, including wheezing, lethargy, sneezing, and elevated body temperature [122,229], with acute bronchiolitis in infected lungs for some individuals [167]. Nevertheless, no significant mortality was observed in the ferret model, nor were the severe clinical signs and symptoms observed in humans [230]. Because ferrets can cough and sneeze, they are able to efficiently transmit the virus to their naive cage-mates, indicating that this model is also appropriate for transmission studies [168]. The Kutter et al. study demonstrated efficient transmission through the air between ferrets over more than one meter distance, without discriminating between transmission via small aerosols, large droplets and fomites [228]. Ferrets are further used to assess the immune response to SARS-CoV-2 intranasal inoculation. A similar immune response was observed in lung samples from post-mortem COVID-19 patients compared to ferret samples; i.e., induced high levels of chemokines such as CCL2/8/11 and also a cellular response to IFN-γ [36]. SARS-CoV-2 specific IFN-γ responses were observed in high and medium dose ferrets using lung mononucleocyte samples [230]. Interestingly, it was found that infiltrating macrophages differentiate into M1 or M2 macrophages after SARS-CoV-2 infection [231]. As the ferret immune system reactions are similar to human reactions, they constitute a potentially relevant animal model for therapeutic and vaccine assays. Antiviral efficacies of FDA-approved drugs or under-development compounds have been tested against SARS-CoV-2 infection in ferrets [227,229], supporting the launch of clinical trials.

3.2.4. Chinese Tree Shrew

The Chinese tree shrew is a model which has been successfully employed in replacing primates in biomedical research and drug testing during hepatitis research [232]. Tree shrews exhibit short reproductive cycles and have the advantage of small size. Two studies reported alternative results regarding the use of the mammal in SARS-CoV-2 research. The first study demonstrated undetectable viral RNA in throat and nasal swabs as well in serum samples, while the lung lobes had higher numbers of viral RNA copies [233]. The animals further showed thickened alveolar septa and interstitial hemorrhage with increased levels of aspartate aminotransferase and blood urea nitrogen. The second study found detectable SARS-CoV-2 RNA in nasal, throat, and anal swabs, along with the observation of absent clinical signs except for increased body temperature in females, while lung damage was similar to the previous study [234]. Interestingly, the highest level of virus shedding was observed in young individuals, suggesting their greater susceptibility. The SARS-CoV-2 thus had constrained viral replication and shedding and disparate pathogenesis in tree shrews.

3.2.5. Non-Human Primates

NHPs are thought to be the animal gold standard for modeling diseases, having strong physical and genetic similarities to humans. They have been widely employed for studying viral infection, including the AIDS, Zika, MERS-CoV, and SARS-CoV-1/-2 [235,236,237]. To date, different models have been studied for COVID-19 research, including rhesus, cynomolgus, African green monkeys (AGMs), common marmosets, and baboons. Altogether, NHP species-based studies highlighted the heterogeneous spectrum of SARS-CoV-2 infection [238,239]. (1) Rhesus and cynomolgus macaques exhibited high levels of viral replication in the upper and lower respiratory tracts, as well as the digestive and urinary tracts [173,174]. AGMs have lower respiratory tract viral titers compared to macaques [181,240], and common marmosets exhibited only low transient levels of viral RNA in nasopharyngeal swab samples [239]. (2) NHP exhibit absent-to-moderate clinical signs characterized by inconstant and benign fever for marmosets and an altered general status for macaques and AGMs [238]. Common marmoset low viral susceptibility and almost absent clinical manifestations make this species less attractive. In contrast, macaques and AGMs experienced weight loss, fever and diffuse alveolar damage, associated with a high permissivity to SARS-CoV-2 [241]. Similarly, baboons had higher viral titers and longer viremia, as well as more pronounced lung pathology relative to SARS-CoV-2 infected macaques [242]. Because SARS-CoV-2 infected baboons experience severe COVID-19 symptoms and breed throughout the year with high fecundity, they should be examined as a supplemental model for biomedical research. As observed in the hamster model, older baboon individuals were more affected by viral infection, having higher viral loads in lung tissues for a longer period time, and an increased number of histopathological changes [3,243]. Aged monkeys could thus be suggested to mimic the most severe form of COVID-19. Interestingly, macaques and AGMs also exhibited virus-specific antibodies with neutralizing activity and T cell responses, making them remarkable models for the study of immune responses after viral challenge [181,244,245]. Rhesus macaque sera samples presented increasingly high levels of neutralizing antibodies 5- and 14-days post-rechallenge, acting as protection against re-infection [175].
NHP models have been mainly useful for therapeutic agents and vaccine candidate testing. Regarding drug assays, remdesivir has been evaluated on rhesus macaques, showing reduced pulmonary infiltrates and reduced viral titers on bronchioalveolar lavage only 12 h after the first dose [176]. Lung tissue autopsies further revealed lower lung viral loads and decreased lung damage in the remdesivir-treated group compared to the control group. HCQ antiviral activity against SARS-CoV-2 infection was also monitored in the cynomolgus model, either alone or in combination with azithromycin [93]. All treated NHP had viral RNA load kinetics similar to untreated NHP at the tracheal, nasopharyngeal, and bronchioalveolar level, and HCQ did not enhance viral clearance duration. mRNA-1273 and BNT162b2 demonstrated a protective effect for both lower and upper airways, as viral subgenomic RNA in bronchioalveolar fluids and in nasal swabs were significantly lower compared to the control group after 2 and 3 days post-inoculation, respectively [246,247]. None of the animals experienced additional weight or temperature change or altered oxygen saturation rates compared to the control group. Regarding conventional vaccines, ChAdOx1-nCoV-19 assays showed much lower levels of the virus in the bronchioalveolar lavage fluid and lower respiratory tract tissues for the vaccinated animals compared to control [248]. In addition, the adenovirus (Ad26) vaccine entered clinical trials after having demonstrated in rhesus macaques that a preventive single administration of the encoding S protein vaccine led to no detectable virus in bronchioalveolar lavage samples and nasal swabs [249].
Although inevitable during the COVID-19 pandemic, NHP represent less than 5% of all animal research [250], as working with these animals constitutes technical and financial issues as well as ethical challenges. Moreover, different challenge dosages and routes of infection in the NHP model may contribute to highly significant alterations in the level and duration of viral replication observed, requiring standardized protocols.
Nevertheless, animal models clarified the positioning of drugs/vaccines during the COVID-19 pandemic, giving precise ideas of their antiviral potency. Finally, constant evaluation and correction of the most appropriate model used during pandemic period is crucial. Selected models should indeed be utilized in the best possible manner in accordance with assumptions made and the latest scientific updates.

4. Conclusions & Discussion

The COVID-19 global health crisis rapidly reminded us how great the latency period between the development of adapted state-of-the-art models versus the need to promptly understand disease pathophysiology and to evaluate therapeutic candidates could be. Initially, the lack of consensus regarding study design and experimental readouts sometimes led to unreliable data and general confusion. To date, many different preclinical models are accessible for the investigation of SARS-CoV-2 infection. The in vitro models are mandatory for the understanding of the virus life cycle and compound high-throughput screening. They have the advantage of being quite simple and highly informative in considering virus-host interactions and compound mechanisms of action. Animal models have value in the study of virus transmission, tropism, and evaluation of systemic effects of the infection. These complex models are crucial to assess drug and vaccine efficacy before moving onto clinical trials. A lesson to be drawn from the SARS-CoV-2 outbreak is that none of the models tested so far completely reflects human COVID-19. A more precise examination of the SARS-CoV-2 life cycle and pathophysiology via the diversification of assays will improve the comprehension of virus features and save precious time. The advent of 3D cell-based assays coupled with new emergent engineered technologies is surfacing as the new potent modeling strategy for biomedical research. These human-centered approaches recapitulate cellular heterogeneity, the extracellular environment, and more importantly are exploitable over long periods. Combined-3D models represent quite well the human biology complexity and also overcome animal use limitations, being applicable on a larger scale and therefore facilitating clinical translation. Following such an emergency period, it is of critical importance to pursue modeling enhancement and to recall the relevance of each respective model, with their advantages and limitations.

Author Contributions

E.B. performed the literature review, draft the work and wrote the manuscript; S.M. and J.-L.M. contributed to the conceptualization, design of the work, and substantively revised it. Manuscript proofreading and validation were assured by S.M., P.H. and J.-L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

E.B., S.M. and P.H. are employees of Genoscience Pharma.

References

  1. Zheng, J. SARS-CoV-2: An Emerging Coronavirus That Causes a Global Threat. Int. J. Biol. Sci. 2020, 16, 1678–1685. [Google Scholar] [CrossRef] [Green Version]
  2. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 3 June 2021).
  3. Rockx, B.; Kuiken, T.; Herfst, S.; Bestebroer, T.; Lamers, M.M.; Oude Munnink, B.B.; de Meulder, D.; van Amerongen, G.; van den Brand, J.; Okba, N.M.A.; et al. Comparative Pathogenesis of COVID-19, MERS, and SARS in a Nonhuman Primate Model. Science 2020, 368, 1012–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Transmission of SARS-CoV-2: Implications for Infection Prevention Precautions. Available online: https://www.who.int/news-room/commentaries/detail/transmission-of-SARS-CoV-2-implications-for-infection-prevention-precautions (accessed on 3 June 2021).
  5. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
  6. Yuki, K.; Fujiogi, M.; Koutsogiannaki, S. COVID-19 Pathophysiology: A Review. Clin. Immunol. 2020, 215, 108427. [Google Scholar] [CrossRef] [PubMed]
  7. Zhong, P.; Xu, J.; Yang, D.; Shen, Y.; Wang, L.; Feng, Y.; Du, C.; Song, Y.; Wu, C.; Hu, X.; et al. COVID-19-Associated Gastrointestinal and Liver Injury: Clinical Features and Potential Mechanisms. Signal Transduct. Target. Ther. 2020, 5, 256. [Google Scholar] [CrossRef]
  8. Wu, Y.; Xu, X.; Chen, Z.; Duan, J.; Hashimoto, K.; Yang, L.; Liu, C.; Yang, C. Nervous System Involvement after Infection with COVID-19 and Other Coronaviruses. Brain Behav. Immun. 2020, 87, 18–22. [Google Scholar] [CrossRef] [PubMed]
  9. Gu, J.; Korteweg, C. Pathology and Pathogenesis of Severe Acute Respiratory Syndrome. Am. J. Pathol. 2007, 170, 1136–1147. [Google Scholar] [CrossRef] [Green Version]
  10. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-Acute COVID-19 Syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef]
  11. COVID-19-Search Details-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/results/details?cond=Covid19 (accessed on 21 December 2020).
  12. Hui, K.P.Y.; Cheung, M.-C.; Perera, R.A.P.M.; Ng, K.-C.; Bui, C.H.T.; Ho, J.C.W.; Ng, M.M.T.; Kuok, D.I.T.; Shih, K.C.; Tsao, S.-W.; et al. Tropism, Replication Competence, and Innate Immune Responses of the Coronavirus SARS-CoV-2 in Human Respiratory Tract and Conjunctiva: An Analysis in Ex-Vivo and in-Vitro Cultures. Lancet Respir. Med. 2020, 8, 687–695. [Google Scholar] [CrossRef]
  13. Muñoz-Fontela, C.; Dowling, W.E.; Funnell, S.G.P.; Gsell, P.-S.; Riveros-Balta, A.X.; Albrecht, R.A.; Andersen, H.; Baric, R.S.; Carroll, M.W.; Cavaleri, M.; et al. Animal Models for COVID-19. Nature 2020, 586, 509–515. [Google Scholar] [CrossRef]
  14. Cao, B.; Wang, Y.; Wen, D.; Liu, W.; Wang, J.; Fan, G.; Ruan, L.; Song, B.; Cai, Y.; Wei, M.; et al. A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19. N. Engl. J. Med. 2020, 382, 1787–1799. [Google Scholar] [CrossRef] [PubMed]
  15. Remdesivir|COVID-19|Start Date from 11/01/2019 to 02/25/2021-Search Details-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/results/details?term=remdesivir&cond=COVID-19&strd_s=11%2F01%2F2019&strd_e=02%2F25%2F2021 (accessed on 28 June 2022).
  16. Hydroxychloroquine|COVID-19|Start Date from 11/01/2019 to 02/25/2021-Search Details-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/results/details?term=Hydroxychloroquine&cond=COVID-19&strd_s=11%2F01%2F2019&strd_e=02%2F25%2F2021 (accessed on 28 June 2022).
  17. Tocilizumab|COVID-19|Start Date from 11/01/2019 to 02/25/2020-Search Details-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/results/details?term=tocilizumab&cond=COVID-19&strd_s=11%2F01%2F2019&strd_e=02%2F25%2F2020 (accessed on 28 June 2022).
  18. Borba, M.G.S.; Val, F.F.A.; Sampaio, V.S.; Alexandre, M.A.A.; Melo, G.C.; Brito, M.; Mourão, M.P.G.; Brito-Sousa, J.D.; Baía-da-Silva, D.; Guerra, M.V.F.; et al. Effect of High vs Low Doses of Chloroquine Diphosphate as Adjunctive Therapy for Patients Hospitalized With Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection: A Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e208857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Chorin, E.; Dai, M.; Shulman, E.; Wadhwani, L.; Roi-Bar-Cohen; Barbhaiya, C.; Aizer, A.; Holmes, D.; Bernstein, S.; Spinelli, M.; et al. The QT Interval in Patients with SARS-CoV-2 Infection Treated with Hydroxychloroquine/Azithromycin. medRxiv 2020. [Google Scholar] [CrossRef]
  20. Cortegiani, A.; Catalisano, G.; Ippolito, M.; Giarratano, A.; Absalom, A.R.; Einav, S. Retracted Papers on SARS-CoV-2 and COVID-19. Br. J. Anaesth. 2021, 126, e155–e156. [Google Scholar] [CrossRef] [PubMed]
  21. Dowden, H.; Munro, J. Trends in Clinical Success Rates and Therapeutic Focus. Nat. Rev. Drug Discov. 2019, 18, 495–496. [Google Scholar] [CrossRef] [PubMed]
  22. Ren, X.; Glende, J.; Al-Falah, M.; de Vries, V.; Schwegmann-Wessels, C.; Qu, X.; Tan, L.; Tschernig, T.; Deng, H.; Naim, H.Y.; et al. Analysis of ACE2 in Polarized Epithelial Cells: Surface Expression and Function as Receptor for Severe Acute Respiratory Syndrome-Associated Coronavirus. J. Gen. Virol. 2006, 87, 1691–1695. [Google Scholar] [CrossRef]
  23. Rattanapisit, K.; Shanmugaraj, B.; Manopwisedjaroen, S.; Purwono, P.B.; Siriwattananon, K.; Khorattanakulchai, N.; Hanittinan, O.; Boonyayothin, W.; Thitithanyanont, A.; Smith, D.R.; et al. Rapid Production of SARS-CoV-2 Receptor Binding Domain (RBD) and Spike Specific Monoclonal Antibody CR3022 in Nicotiana Benthamiana. Sci. Rep. 2020, 10, 17698. [Google Scholar] [CrossRef]
  24. Takayama, K. In Vitro and Animal Models for SARS-CoV-2 Research. Trends Pharmacol. Sci. 2020, 41, 513–517. [Google Scholar] [CrossRef]
  25. Ogando, N.S.; Dalebout, T.J.; Zevenhoven-Dobbe, J.C.; Limpens, R.W.A.L.; van der Meer, Y.; Caly, L.; Druce, J.; de Vries, J.J.C.; Kikkert, M.; Bárcena, M.; et al. SARS-Coronavirus-2 Replication in Vero E6 Cells: Replication Kinetics, Rapid Adaptation and Cytopathology. J. Gen. Virol. 2020, 101, 925–940. [Google Scholar] [CrossRef]
  26. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef]
  27. Essaidi-Laziosi, M.; Rodriguez, F.J.P.; Hulo, N.; Jacquerioz, F.; Kaiser, L.; Eckerle, I. Estimating Clinical SARS-CoV-2 Infectiousness in Vero E6 and Primary Airway Epithelial Cells. Lancet Microbe 2021, 2, e571. [Google Scholar] [CrossRef]
  28. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. Inhi. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; et al. Characterization of Spike Glycoprotein of SARS-CoV-2 on Virus Entry and Its Immune Cross-Reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Matsuyama, S.; Nao, N.; Shirato, K.; Kawase, M.; Saito, S.; Takayama, I.; Nagata, N.; Sekizuka, T.; Katoh, H.; Kato, F.; et al. Enhanced Isolation of SARS-CoV-2 by TMPRSS2-Expressing Cells. Proc. Natl. Acad. Sci. USA 2020, 117, 7001–7003. [Google Scholar] [CrossRef] [Green Version]
  31. Harcourt, J.; Tamin, A.; Lu, X.; Kamili, S.; Sakthivel, S.K.; Murray, J.; Queen, K.; Tao, Y.; Paden, C.R.; Zhang, J.; et al. Isolation and Characterization of SARS-CoV-2 from the First US COVID-19 Patient. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  32. Hoffmann, M.; Mösbauer, K.; Hofmann-Winkler, H.; Kaul, A.; Kleine-Weber, H.; Krüger, N.; Gassen, N.C.; Müller, M.A.; Drosten, C.; Pöhlmann, S. Chloroquine Does Not Inhibit Infection of Human Lung Cells with SARS-CoV-2. Nature 2020, 585, 588–590. [Google Scholar] [CrossRef]
  33. Harcourt, J.; Tamin, A.; Lu, X.; Kamili, S.; Sakthivel, S.K.; Murray, J.; Queen, K.; Tao, Y.; Paden, C.R.; Zhang, J.; et al. Severe Acute Respiratory Syndrome Coronavirus 2 from Patient with Coronavirus Disease, United States. Emerg. Infect. Dis. 2020, 26, 1266–1273. [Google Scholar] [CrossRef]
  34. Chen, H.; Liu, W.; Liu, D.; Zhao, L.; Yu, J. SARS-CoV-2 Activates Lung Epithelia Cell Proinflammatory Signaling and Leads to Immune Dysregulation in COVID-19 Patients by Single-Cell Sequencing. medRxiv 2020. [Google Scholar] [CrossRef]
  35. Chu, H.; Chan, J.F.-W.; Yuen, T.T.-T.; Shuai, H.; Yuan, S.; Wang, Y.; Hu, B.; Yip, C.C.-Y.; Tsang, J.O.-L.; Huang, X.; et al. Comparative Tropism, Replication Kinetics, and Cell Damage Profiling of SARS-CoV-2 and SARS-CoV with Implications for Clinical Manifestations, Transmissibility, and Laboratory Studies of COVID-19: An Observational Study. Lancet Microbe 2020, 1, e14–e23. [Google Scholar] [CrossRef]
  36. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.-C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  37. Bojkova, D.; Klann, K.; Koch, B.; Widera, M.; Krause, D.; Ciesek, S.; Cinatl, J.; Münch, C. Proteomics of SARS-CoV-2-Infected Host Cells Reveals Therapy Targets. Nature 2020, 583, 469–472. [Google Scholar] [CrossRef] [PubMed]
  38. Paolacci, S.; Ceccarini, M.R.; Codini, M.; Manara, E.; Tezzele, S.; Percio, M.; Capodicasa, N.; Kroni, D.; Dundar, M.; Ergoren, M.C.; et al. Pilot Study for the Evaluation of Safety Profile of a Potential Inhibitor of SARS-CoV-2 Endocytosis. Acta Biomed. 2020, 91, e2020009. [Google Scholar] [CrossRef] [PubMed]
  39. Identification of Inhibitors of SARS-CoV-2 in-Vitro Cellular Toxicity in Human (Caco-2) Cells Using a Large Scale Drug Repurposing Collection. Available online: https://www.researchsquare.com (accessed on 1 June 2021).
  40. Cagno, V. SARS-CoV-2 Cellular Tropism. Lancet Microbe 2020, 1, e2–e3. [Google Scholar] [CrossRef]
  41. Banerjee, A.; Nasir, J.A.; Budylowski, P.; Yip, L.; Aftanas, P.; Christie, N.; Ghalami, A.; Baid, K.; Raphenya, A.R.; Hirota, J.A.; et al. Isolation, Sequence, Infectivity, and Replication Kinetics of Severe Acute Respiratory Syndrome Coronavirus 2. Emerg. Infect. Dis. 2020, 26, 2054–2063. [Google Scholar] [CrossRef] [PubMed]
  42. Li, C.; Chu, H.; Liu, X.; Chiu, M.C.; Zhao, X.; Wang, D.; Wei, Y.; Hou, Y.; Shuai, H.; Cai, J.; et al. Human Coronavirus Dependency on Host Heat Shock Protein 90 Reveals an Antiviral Target. Emerg. Microbes Infect. 2020, 9, 2663–2672. [Google Scholar] [CrossRef] [PubMed]
  43. Lapaillerie, D.; Charlier, C.; Fernandes, H.S.; Sousa, S.F.; Lesbats, P.; Weigel, P.; Favereaux, A.; Guyonnet-Duperat, V.; Parissi, V. In Silico, In Vitro and In Cellulo Models for Monitoring SARS-CoV-2 Spike/Human ACE2 Complex, Viral Entry and Cell Fusion. Viruses 2021, 13, 365. [Google Scholar] [CrossRef]
  44. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell Entry Mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  45. Bayati, A.; Kumar, R.; Francis, V.; McPherson, P.S. SARS-CoV-2 Infects Cells after Viral Entry via Clathrin-Mediated Endocytosis. J. Biol. Chem. 2021, 296. [Google Scholar] [CrossRef]
  46. Xia, S.; Liu, M.; Wang, C.; Xu, W.; Lan, Q.; Feng, S.; Qi, F.; Bao, L.; Du, L.; Liu, S.; et al. Inhibition of SARS-CoV-2 (Previously 2019-NCoV) Infection by a Highly Potent Pan-Coronavirus Fusion Inhibitor Targeting Its Spike Protein That Harbors a High Capacity to Mediate Membrane Fusion. Cell Res. 2020, 30, 343–355. [Google Scholar] [CrossRef] [Green Version]
  47. Buchrieser, J.; Dufloo, J.; Hubert, M.; Monel, B.; Planas, D.; Rajah, M.M.; Planchais, C.; Porrot, F.; Guivel-Benhassine, F.; Van der Werf, S.; et al. Syncytia Formation by SARS-CoV-2-infected Cells. EMBO J. 2020, 39, e106267. [Google Scholar] [CrossRef]
  48. Wang, C.; Li, W.; Drabek, D.; Okba, N.M.A.; van Haperen, R.; Osterhaus, A.D.M.E.; van Kuppeveld, F.J.M.; Haagmans, B.L.; Grosveld, F.; Bosch, B.-J. A Human Monoclonal Antibody Blocking SARS-CoV-2 Infection. Nat. Commun. 2020, 11, 2251. [Google Scholar] [CrossRef] [PubMed]
  49. Saccon, E.; Chen, X.; Mikaeloff, F.; Rodriguez, J.E.; Szekely, L.; Vinhas, B.S.; Krishnan, S.; Byrareddy, S.N.; Frisan, T.; Végvári, Á.; et al. Cell-Type-Resolved Quantitative Proteomics Map of Interferon Response against SARS-CoV-2. iScience 2021, 24, 102420. [Google Scholar] [CrossRef] [PubMed]
  50. Mirabelli, C.; Wotring, J.W.; Zhang, C.J.; McCarty, S.M.; Fursmidt, R.; Pretto, C.D.; Qiao, Y.; Zhang, Y.; Frum, T.; Kadambi, N.S.; et al. Morphological Cell Profiling of SARS-CoV-2 Infection Identifies Drug Repurposing Candidates for COVID-19. Proc. Natl. Acad. Sci. USA 2021, 118, e2105815118. [Google Scholar] [CrossRef] [PubMed]
  51. Appelberg, S.; Gupta, S.; Svensson Akusjärvi, S.; Ambikan, A.T.; Mikaeloff, F.; Saccon, E.; Végvári, Á.; Benfeitas, R.; Sperk, M.; Ståhlberg, M.; et al. Dysregulation in Akt/MTOR/HIF-1 Signaling Identified by Proteo-Transcriptomics of SARS-CoV-2 Infected Cells. Emerg. Microbes Infect. 2020, 9, 1748–1760. [Google Scholar] [CrossRef]
  52. Bartolomeo, C.; Alves, T.; Lemes, R.; Ivanov, G.; Morais, R.; Costa, A.; Nishino, M.; Bassani, T.; Pereira, G.; Smaili, S.; et al. ACE2 overexpression changes the SARS-CoV-2 infection profile in BEAS-2b cells. Cytotherapy 2021, 23, 1. [Google Scholar] [CrossRef]
  53. Wang, S.; Qiu, Z.; Hou, Y.; Deng, X.; Xu, W.; Zheng, T.; Wu, P.; Xie, S.; Bian, W.; Zhang, C.; et al. AXL Is a Candidate Receptor for SARS-CoV-2 That Promotes Infection of Pulmonary and Bronchial Epithelial Cells. Cell Res. 2021, 31, 126–140. [Google Scholar] [CrossRef]
  54. Hao, S.; Ning, K.; Kuz, C.A.; Vorhies, K.; Yan, Z.; Qiu, J. Long-Term Modeling of SARS-CoV-2 Infection of In Vitro Cultured Polarized Human Airway Epithelium. mBio 2020, 11, e02852-20. [Google Scholar] [CrossRef]
  55. Sheahan, T.P.; Sims, A.C.; Zhou, S.; Graham, R.L.; Pruijssers, A.J.; Agostini, M.L.; Leist, S.R.; Schäfer, A.; Dinnon, K.H.; Stevens, L.J.; et al. An Orally Bioavailable Broad-Spectrum Antiviral Inhibits SARS-CoV-2 in Human Airway Epithelial Cell Cultures and Multiple Coronaviruses in Mice. Sci. Transl. Med. 2020, 12, eabb5883. [Google Scholar] [CrossRef] [Green Version]
  56. Mulay, A.; Konda, B.; Garcia, G.; Yao, C.; Beil, S.; Villalba, J.M.; Koziol, C.; Sen, C.; Purkayastha, A.; Kolls, J.K.; et al. K.; et al. SARS-CoV-2 Infection of Primary Human Lung Epithelium for COVID-19 Modeling and Drug Discovery. Cell Rep. 2021, 35, 109055. [Google Scholar] [CrossRef]
  57. Pizzorno, A.; Padey, B.; Julien, T.; Trouillet-Assant, S.; Traversier, A.; Errazuriz-Cerda, E.; Fouret, J.; Dubois, J.; Gaymard, A.; Lescure, F.-X.; et al. Characterization and Treatment of SARS-CoV-2 in Nasal and Bronchial Human Airway Epithelia. Cell Rep. Med. 2020, 1, 100059. [Google Scholar] [CrossRef]
  58. Lukassen, S.; Chua, R.L.; Trefzer, T.; Kahn, N.C.; Schneider, M.A.; Muley, T.; Winter, H.; Meister, M.; Veith, C.; Boots, A.W.; et al. SARS-CoV-2 Receptor ACE2 and TMPRSS2 Are Primarily Expressed in Bronchial Transient Secretory Cells. EMBO J. 2020, 39, e105114. [Google Scholar] [CrossRef] [PubMed]
  59. Han, Y.; Duan, X.; Yang, L.; Nilsson-Payant, B.E.; Wang, P.; Duan, F.; Tang, X.; Yaron, T.M.; Zhang, T.; Uhl, S.; et al. Identification of SARS-CoV-2 Inhibitors Using Lung and Colonic Organoids. Nature 2021, 589, 270–275. [Google Scholar] [CrossRef]
  60. Salahudeen, A.A.; Choi, S.S.; Rustagi, A.; Zhu, J.; van Unen, V.; de la O, S.M.; Flynn, R.A.; Margalef-Català, M.; Santos, A.J.M.; Ju, J.; et al. Progenitor Identification and SARS-CoV-2 Infection in Human Distal Lung Organoids. Nature 2020, 588, 670–675. [Google Scholar] [CrossRef] [PubMed]
  61. Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Hurtado Del Pozo, C.; Prosper, F.; et al. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell 2020, 181, 905–913.e7. [Google Scholar] [CrossRef] [PubMed]
  62. Lamers, M.M.; Beumer, J.; van der Vaart, J.; Knoops, K.; Puschhof, J.; Breugem, T.I.; Ravelli, R.B.G.; Paul van Schayck, J.; Mykytyn, A.Z.; Duimel, H.Q.; et al. SARS-CoV-2 Productively Infects Human Gut Enterocytes. Science 2020, 369, 50–54. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, J.; Li, C.; Liu, X.; Chiu, M.C.; Zhao, X.; Wang, D.; Wei, Y.; Lee, A.; Zhang, A.J.; Chu, H.; et al. Infection of Bat and Human Intestinal Organoids by SARS-CoV-2. Nat. Med. 2020, 26, 1077–1083. [Google Scholar] [CrossRef]
  64. Si, L.; Bai, H.; Rodas, M.; Cao, W.; Oh, C.Y.; Jiang, A.; Moller, R.; Hoagland, D.; Oishi, K.; Horiuchi, S.; et al. A Human-Airway-on-a-Chip for the Rapid Identification of Candidate Antiviral Therapeutics and Prophylactics. Nat. Biomed. Eng 2021, 5, 815–829. [Google Scholar] [CrossRef]
  65. Identification of Potential Key Genes for SARS-CoV-2 Infected Human Bronchial Organoids Based on Bioinformatics Analysis|BioRxiv. Available online: https://www.biorxiv.org/content/10.1101/2020.08.18.256735v1 (accessed on 1 June 2021).
  66. Si, L.; Bai, H.; Rodas, M.; Cao, W.; Oh, C.Y.; Jiang, A.; Nurani, A.; Zhu, D.Y.; Goyal, G.; Gilpin, S.E.; et al. Human Organs-on-Chips as Tools for Repurposing Approved Drugs as Potential Influenza and COVID19 Therapeutics in Viral Pandemics. bioRxiv 2020. [Google Scholar] [CrossRef]
  67. Human Organ Chip-Enabled Pipeline to Rapidly Repurpose Therapeutics during Viral Pandemics|BioRxiv. Available online: https://www.biorxiv.org/content/10.1101/2020.04.13.039917v3 (accessed on 1 June 2021).
  68. Guo, Y.; Luo, R.; Wang, Y.; Deng, P.; Song, T.; Zhang, M.; Wang, P.; Zhang, X.; Cui, K.; Tao, T.; et al. SARS-CoV-2 Induced Intestinal Responses with a Biomimetic Human Gut-on-Chip. Sci. Bull. 2021, 66, 783–793. [Google Scholar] [CrossRef]
  69. Bertram, S.; Glowacka, I.; Blazejewska, P.; Soilleux, E.; Allen, P.; Danisch, S.; Steffen, I.; Choi, S.-Y.; Park, Y.; Schneider, H.; et al. TMPRSS2 and TMPRSS4 Facilitate Trypsin-Independent Spread of Influenza Virus in Caco-2 Cells. J. Virol. 2010, 84, 10016–10025. [Google Scholar] [CrossRef] [Green Version]
  70. Mossel, E.C.; Huang, C.; Narayanan, K.; Makino, S.; Tesh, R.B.; Peters, C.J. Exogenous ACE2 Expression Allows Refractory Cell Lines to Support Severe Acute Respiratory Syndrome Coronavirus Replication. J. Virol. 2005, 79, 3846–3850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Sherman, E.J.; Emmer, B.T. ACE2 Protein Expression within Isogenic Cell Lines Is Heterogeneous and Associated with Distinct Transcriptomes. Sci. Rep. 2021, 11, 15900. [Google Scholar] [CrossRef] [PubMed]
  72. Ma, D.; Chen, C.-B.; Jhanji, V.; Xu, C.; Yuan, X.-L.; Liang, J.-J.; Huang, Y.; Cen, L.-P.; Ng, T.K. Expression of SARS-CoV-2 Receptor ACE2 and TMPRSS2 in Human Primary Conjunctival and Pterygium Cell Lines and in Mouse Cornea. Eye 2020, 34, 1212–1219. [Google Scholar] [CrossRef] [PubMed]
  73. Sims, A.C.; Burkett, S.E.; Yount, B.; Pickles, R.J. SARS-CoV Replication and Pathogenesis in an in Vitro Model of the Human Conducting Airway Epithelium. Virus Res. 2008, 133, 33–44. [Google Scholar] [CrossRef]
  74. Tseng, C.-T.K.; Tseng, J.; Perrone, L.; Worthy, M.; Popov, V.; Peters, C.J. Apical Entry and Release of Severe Acute Respiratory Syndrome-Associated Coronavirus in Polarized Calu-3 Lung Epithelial Cells. J. Virol. 2005, 79, 9470–9479. [Google Scholar] [CrossRef] [Green Version]
  75. Liu, J.; Cao, R.; Xu, M.; Wang, X.; Zhang, H.; Hu, H.; Li, Y.; Hu, Z.; Zhong, W.; Wang, M. Hydroxychloroquine, a Less Toxic Derivative of Chloroquine, Is Effective in Inhibiting SARS-CoV-2 Infection in Vitro. Cell Discov. 2020, 6, 16. [Google Scholar] [CrossRef] [Green Version]
  76. Yao, X.; Ye, F.; Zhang, M.; Cui, C.; Huang, B.; Niu, P.; Liu, X.; Zhao, L.; Dong, E.; Song, C.; et al. In Vitro Antiviral Activity and Projection of Optimized Dosing Design of Hydroxychloroquine for the Treatment of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Clin. Infect. Dis. 2020, 71, 732–739. [Google Scholar] [CrossRef] [Green Version]
  77. Chloroquine|Covid19-Search Details-ClinicalTrials.Gov. Available online: https://www.clinicaltrials.gov/ct2/results/details?term=Chloroquine&cond=Covid19 (accessed on 29 May 2021).
  78. Touret, F.; de Lamballerie, X. Of Chloroquine and COVID-19. Antiviral. Res. 2020, 177, 104762. [Google Scholar] [CrossRef]
  79. Ferner, R.E.; Aronson, J.K. Chloroquine and Hydroxychloroquine in COVID-19. BMJ 2020, 369, m1432. [Google Scholar] [CrossRef] [Green Version]
  80. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and Chloroquine Effectively Inhibit the Recently Emerged Novel Coronavirus (2019-NCoV) in Vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
  81. Colson, P.; Rolain, J.-M.; Lagier, J.-C.; Brouqui, P.; Raoult, D. Chloroquine and Hydroxychloroquine as Available Weapons to Fight COVID-19. Int. J. Antimicrob. Agents 2020, 55, 105932. [Google Scholar] [CrossRef] [PubMed]
  82. Funnell, S.G.P.; Dowling, W.E.; Muñoz-Fontela, C.; Gsell, P.-S.; Ingber, D.E.; Hamilton, G.A.; Delang, L.; Rocha-Pereira, J.; Kaptein, S.; Dallmeier, K.H.; et al. Emerging Preclinical Evidence Does Not Support Broad Use of Hydroxychloroquine in COVID-19 Patients. Nat. Commun. 2020, 11, 4253. [Google Scholar] [CrossRef] [PubMed]
  83. Younis, N.K.; Zareef, R.O.; Al Hassan, S.N.; Bitar, F.; Eid, A.H.; Arabi, M. Hydroxychloroquine in COVID-19 Patients: Pros and Cons. Front. Pharmacol. 2020, 11, 597985. [Google Scholar] [CrossRef] [PubMed]
  84. Pandamooz, S.; Jurek, B.; Meinung, C.-P.; Baharvand, Z.; Sahebi Shahem-Abadi, A.; Haerteis, S.; Miyan, J.A.; Downing, J.; Dianatpour, M.; Borhani-Haghighi, A.; et al. Experimental Models of SARS-CoV-2 Infection: Possible Platforms to Study COVID-19 Pathogenesis and Potential Treatments. Annu. Rev. Pharmacol. Toxicol. 2022, 62, 25–53. [Google Scholar] [CrossRef] [PubMed]
  85. Sadhukhan, P.; Ugurlu, M.T.; Hoque, M.O. Effect of COVID-19 on Lungs: Focusing on Prospective Malignant Phenotypes. Cancers 2020, 12, 3822. [Google Scholar] [CrossRef] [PubMed]
  86. Ghandi, M.; Huang, F.W.; Jané-Valbuena, J.; Kryukov, G.V.; Lo, C.C.; McDonald, E.R.; Barretina, J.; Gelfand, E.T.; Bielski, C.M.; Li, H.; et al. Next-Generation Characterization of the Cancer Cell Line Encyclopedia. Nature 2019, 569, 503–508. [Google Scholar] [CrossRef]
  87. Banach, B.S.; Orenstein, J.M.; Fox, L.M.; Randell, S.H.; Rowley, A.H.; Baker, S.C. Human Airway Epithelial Cell Culture to Identify New Respiratory Viruses: Coronavirus NL63 as a Model. J. Virol. Methods 2009, 156, 19–26. [Google Scholar] [CrossRef]
  88. Farsani, S.M.J.; Deijs, M.; Dijkman, R.; Molenkamp, R.; Jeeninga, R.E.; Ieven, M.; Goossens, H.; van der Hoek, L. Culturing of Respiratory Viruses in Well-Differentiated Pseudostratified Human Airway Epithelium as a Tool to Detect Unknown Viruses. Influenza Other Respir Viruses 2015, 9, 51–57. [Google Scholar] [CrossRef]
  89. Jonsdottir, H.R.; Dijkman, R. Coronaviruses and the Human Airway: A Universal System for Virus-Host Interaction Studies. Virol. J. 2016, 13, 24. [Google Scholar] [CrossRef] [Green Version]
  90. Hou, Y.J.; Okuda, K.; Edwards, C.E.; Martinez, D.R.; Asakura, T.; Dinnon, K.H.; Kato, T.; Lee, R.E.; Yount, B.L.; Mascenik, T.M.; et al. SARS-CoV-2 Reverse Genetics Reveals a Variable Infection Gradient in the Respiratory Tract. Cell 2020, 182, 429–446.e14. [Google Scholar] [CrossRef]
  91. Zhu, N.; Wang, W.; Liu, Z.; Liang, C.; Wang, W.; Ye, F.; Huang, B.; Zhao, L.; Wang, H.; Zhou, W.; et al. Morphogenesis and Cytopathic Effect of SARS-CoV-2 Infection in Human Airway Epithelial Cells. Nat. Commun. 2020, 11, 3910. [Google Scholar] [CrossRef] [PubMed]
  92. Pruijssers, A.J.; George, A.S.; Schäfer, A.; Leist, S.R.; Gralinksi, L.E.; Dinnon, K.H.; Yount, B.L.; Agostini, M.L.; Stevens, L.J.; Chappell, J.D.; et al. Remdesivir Inhibits SARS-CoV-2 in Human Lung Cells and Chimeric SARS-CoV Expressing the SARS-CoV-2 RNA Polymerase in Mice. Cell Rep. 2020, 32, 107940. [Google Scholar] [CrossRef] [PubMed]
  93. Maisonnasse, P.; Guedj, J.; Contreras, V.; Behillil, S.; Solas, C.; Marlin, R.; Naninck, T.; Pizzorno, A.; Lemaitre, J.; Gonçalves, A.; et al. Hydroxychloroquine Use against SARS-CoV-2 Infection in Non-Human Primates. Nature 2020, 585, 584–587. [Google Scholar] [CrossRef]
  94. Jacob, F.; Pather, S.R.; Huang, W.-K.; Zhang, F.; Wong, S.Z.H.; Zhou, H.; Cubitt, B.; Fan, W.; Chen, C.Z.; Xu, M.; et al. Human Pluripotent Stem Cell-Derived Neural Cells and Brain Organoids Reveal SARS-CoV-2 Neurotropism Predominates in Choroid Plexus Epithelium. Cell Stem Cell 2020, 27, 937–950.e9. [Google Scholar] [CrossRef] [PubMed]
  95. Yu, F.; Jia, R.; Tang, Y.; Liu, J.; Wei, B. SARS-CoV-2 Infection and Stem Cells: Interaction and Intervention. Stem Cell Res. 2020, 46, 101859. [Google Scholar] [CrossRef]
  96. Surendran, H.; Nandakumar, S.; Pal, R. Human Induced Pluripotent Stem Cell-Derived Lung Epithelial System for SARS-CoV-2 Infection Modeling and Its Potential in Drug Repurposing. Stem Cells Dev. 2020, 29, 1365–1369. [Google Scholar] [CrossRef]
  97. Huang, J.; Hume, A.J.; Abo, K.M.; Werder, R.B.; Villacorta-Martin, C.; Alysandratos, K.-D.; Beermann, M.L.; Simone-Roach, C.; Lindstrom-Vautrin, J.; Olejnik, J.; et al. SARS-CoV-2 Infection of Pluripotent Stem Cell-Derived Human Lung Alveolar Type 2 Cells Elicits a Rapid Epithelial-Intrinsic Inflammatory Response. Cell Stem Cell 2020, 27, 962–973.e7. [Google Scholar] [CrossRef]
  98. Marshall, M. The Lasting Misery of Coronavirus Long-Haulers. Nature 2020, 585, 339–341. [Google Scholar] [CrossRef]
  99. Duan, F.; Guo, L.; Yang, L.; Han, Y.; Thakur, A.; Nilsson-Payant, B.E.; Wang, P.; Zhang, Z.; Ma, C.Y.; Zhou, X.; et al. Modeling COVID-19 with Human Pluripotent Stem Cell-Derived Cells Reveals Synergistic Effects of Anti-Inflammatory Macrophages with ACE2 Inhibition Against SARS-CoV-2. Res. Sq. 2022. preprint. [Google Scholar] [CrossRef]
  100. Riva, L.; Yuan, S.; Yin, X.; Martin-Sancho, L.; Matsunaga, N.; Pache, L.; Burgstaller-Muehlbacher, S.; De Jesus, P.D.; Teriete, P.; Hull, M.V.; et al. Discovery of SARS-CoV-2 Antiviral Drugs through Large-Scale Compound Repurposing. Nature 2020, 586, 113–119. [Google Scholar] [CrossRef]
  101. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.-H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Autoantibodies against Type I IFNs in Patients with Life-Threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn Errors of Type I IFN Immunity in Patients with Life-Threatening COVID-19. Science 2020, 370, eabd4570. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, J.; Koo, B.-K.; Knoblich, J.A. Human Organoids: Model Systems for Human Biology and Medicine. Nat. Rev. Mol. Cell Biol. 2020, 21, 571–584. [Google Scholar] [CrossRef] [PubMed]
  104. de Souza, N. Organoids. Nat. Methods 2018, 15, 23. [Google Scholar] [CrossRef]
  105. Yu, J. Organoids: A New Model for SARS-CoV-2 Translational Research. Int. J. Stem Cells 2021, 14, 138–149. [Google Scholar] [CrossRef]
  106. Han, Y.; Yang, L.; Duan, X.; Duan, F.; Nilsson-Payant, B.E.; Yaron, T.M.; Wang, P.; Tang, X.; Zhang, T.; Zhao, Z.; et al. Identification of Candidate COVID-19 Therapeutics Using HPSC-Derived Lung Organoids. bioRxiv 2020. [Google Scholar] [CrossRef]
  107. Caution on Kidney Dysfunctions of COVID-19 Patients|MedRxiv. Available online: https://www.medrxiv.org/content/10.1101/2020.02.08.20021212v2 (accessed on 29 May 2021).
  108. Fan, Z.; Chen, L.; Li, J.; Cheng, X.; Yang, J.; Tian, C.; Zhang, Y.; Huang, S.; Liu, Z.; Cheng, J. Clinical Features of COVID-19-Related Liver Functional Abnormality. Clin. Gastroenterol. Hepatol. 2020, 18, 1561–1566. [Google Scholar] [CrossRef]
  109. Zheng, Y.-Y.; Ma, Y.-T.; Zhang, J.-Y.; Xie, X. COVID-19 and the Cardiovascular System. Nat. Rev. Cardiol. 2020, 17, 259–260. [Google Scholar] [CrossRef] [Green Version]
  110. Tiwari, S.K.; Wang, S.; Smith, D.; Carlin, A.F.; Rana, T.M. Revealing Tissue-Specific SARS-CoV-2 Infection and Host Responses Using Human Stem Cell-Derived Lung and Cerebral Organoids. Stem Cell Rep. 2021, 16, 437–445. [Google Scholar] [CrossRef]
  111. Burgueño, J.F.; Reich, A.; Hazime, H.; Quintero, M.A.; Fernandez, I.; Fritsch, J.; Santander, A.M.; Brito, N.; Damas, O.M.; Deshpande, A.; et al. Expression of SARS-CoV-2 Entry Molecules ACE2 and TMPRSS2 in the Gut of Patients With IBD. Inflamm. Bowel Dis. 2020, 26, 797–808. [Google Scholar] [CrossRef] [Green Version]
  112. Allison, S.J. SARS-CoV-2 Infection of Kidney Organoids Prevented with Soluble Human ACE2. Nat. Rev. Nephrol. 2020, 16, 316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wysocki, J.; Ye, M.; Hassler, L.; Gupta, A.K.; Wang, Y.; Nicoleascu, V.; Randall, G.; Wertheim, J.A.; Batlle, D. A Novel Soluble ACE2 Variant with Prolonged Duration of Action Neutralizes SARS-CoV-2 Infection in Human Kidney Organoids. J. Am. Soc. Nephrol. 2021, 32, 795–803. [Google Scholar] [CrossRef] [PubMed]
  114. Zhao, B.; Ni, C.; Gao, R.; Wang, Y.; Yang, L.; Wei, J.; Lv, T.; Liang, J.; Zhang, Q.; Xu, W.; et al. Recapitulation of SARS-CoV-2 Infection and Cholangiocyte Damage with Human Liver Ductal Organoids. Protein Cell 2020, 11, 771–775. [Google Scholar] [CrossRef] [Green Version]
  115. Pellegrini, L.; Albecka, A.; Mallery, D.L.; Kellner, M.J.; Paul, D.; Carter, A.P.; James, L.C.; Lancaster, M.A. SARS-CoV-2 Infects the Brain Choroid Plexus and Disrupts the Blood-CSF Barrier in Human Brain Organoids. Cell Stem Cell 2020, 27, 951–961.e5. [Google Scholar] [CrossRef] [PubMed]
  116. Lamers, M.M.; van der Vaart, J.; Knoops, K.; Riesebosch, S.; Breugem, T.I.; Mykytyn, A.Z.; Beumer, J.; Schipper, D.; Bezstarosti, K.; Koopman, C.D.; et al. An Organoid-Derived Bronchioalveolar Model for SARS-CoV-2 Infection of Human Alveolar Type II-like Cells. EMBO J. 2021, 40, e105912. [Google Scholar] [CrossRef] [PubMed]
  117. Ramani, A.; Müller, L.; Ostermann, P.N.; Gabriel, E.; Abida-Islam, P.; Müller-Schiffmann, A.; Mariappan, A.; Goureau, O.; Gruell, H.; Walker, A.; et al. SARS-CoV-2 Targets Neurons of 3D Human Brain Organoids. EMBO J. 2020, 39, e106230. [Google Scholar] [CrossRef]
  118. Makovoz, B.; Møller, R.; Eriksen, A.Z.; tenOever, B.R.; Blenkinsop, T.A. SARS-CoV-2 Infection of Ocular Cells from Human Adult Donor Eyes and HESC-Derived Eye Organoids. Soc. Sci. Res. Netw. 2020. preprint. [Google Scholar] [CrossRef] [PubMed]
  119. Wu, P.; Duan, F.; Luo, C.; Liu, Q.; Qu, X.; Liang, L.; Wu, K. Characteristics of Ocular Findings of Patients With Coronavirus Disease 2019 (COVID-19) in Hubei Province, China. JAMA Ophthalmol. 2020, 138, 575–578. [Google Scholar] [CrossRef]
  120. Leonardi, A.; Rosani, U.; Brun, P. Ocular Surface Expression of SARS-CoV-2 Receptors. Ocul. Immunol. Inflamm. 2020, 28, 735–738. [Google Scholar] [CrossRef]
  121. Triana, S.; Metz-Zumaran, C.; Ramirez, C.; Kee, C.; Doldan, P.; Shahraz, M.; Schraivogel, D.; Gschwind, A.R.; Sharma, A.K.; Steinmetz, L.M.; et al. Single-Cell Analyses Reveal SARS-CoV-2 Interference with Intrinsic Immune Response in the Human Gut. Mol. Syst. Biol. 2021, 17, e10232. [Google Scholar] [CrossRef]
  122. Bar-Ephraim, Y.E.; Kretzschmar, K.; Clevers, H. Organoids in Immunological Research. Nat. Rev. Immunol. 2020, 20, 279–293. [Google Scholar] [CrossRef] [PubMed]
  123. Rogoz, A.; Reis, B.S.; Karssemeijer, R.A.; Mucida, D. A 3-D Enteroid-Based Model to Study T-Cell and Epithelial Cell Interaction. J. Immunol. Methods 2015, 421, 89–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Imatinib|Sars-Cov-2-Search Details-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/results/details?term=imatinib&cond=sars-cov-2 (accessed on 29 May 2021).
  125. Zhao, H.; Mendenhall, M.; Deininger, M.W. Imatinib Is Not a Potent Anti-SARS-CoV-2 Drug. Leukemia 2020, 34, 3085–3087. [Google Scholar] [CrossRef]
  126. Beumer, J.; Geurts, M.H.; Lamers, M.M.; Puschhof, J.; Zhang, J.; van der Vaart, J.; Mykytyn, A.Z.; Breugem, T.I.; Riesebosch, S.; Schipper, D.; et al. A CRISPR/Cas9 Genetically Engineered Organoid Biobank Reveals Essential Host Factors for Coronaviruses. Nat. Commun. 2021, 12, 5498. [Google Scholar] [CrossRef] [PubMed]
  127. Driehuis, E.; Kretzschmar, K.; Clevers, H. Establishment of Patient-Derived Cancer Organoids for Drug-Screening Applications. Nat. Protoc. 2020, 15, 3380–3409. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, M.; Wang, P.; Luo, R.; Wang, Y.; Li, Z.; Guo, Y.; Yao, Y.; Li, M.; Tao, T.; Chen, W.; et al. Biomimetic Human Disease Model of SARS-CoV-2 Induced Lung Injury and Immune Responses on Organ Chip System. Adv. Sci. 2020, 8, 2002928. [Google Scholar] [CrossRef]
  129. Koyilot, M.C.; Natarajan, P.; Hunt, C.R.; Sivarajkumar, S.; Roy, R.; Joglekar, S.; Pandita, S.; Tong, C.W.; Marakkar, S.; Subramanian, L.; et al. Breakthroughs and Applications of Organ-on-a-Chip Technology. Cells 2022, 11, 1828. [Google Scholar] [CrossRef]
  130. Leung, C.M.; de Haan, P.; Ronaldson-Bouchard, K.; Kim, G.-A.; Ko, J.; Rho, H.S.; Chen, Z.; Habibovic, P.; Jeon, N.L.; Takayama, S.; et al. A Guide to the Organ-on-a-Chip. Nat. Rev. Methods Primers 2022, 2, 1–29. [Google Scholar] [CrossRef]
  131. Schlottau, K.; Rissmann, M.; Graaf, A.; Schön, J.; Sehl, J.; Wylezich, C.; Höper, D.; Mettenleiter, T.C.; Balkema-Buschmann, A.; Harder, T.; et al. SARS-CoV-2 in Fruit Bats, Ferrets, Pigs, and Chickens: An Experimental Transmission Study. Lancet Microbe 2020, 1, e218–e225. [Google Scholar] [CrossRef]
  132. Palmer, M.V.; Martins, M.; Falkenberg, S.; Buckley, A.; Caserta, L.C.; Mitchell, P.K.; Cassmann, E.D.; Rollins, A.; Zylich, N.C.; Renshaw, R.W.; et al. Susceptibility of White-Tailed Deer (Odocoileus virginianus) to SARS-CoV-2. J. Virol. 2021, 95, e00083-21. [Google Scholar] [CrossRef]
  133. Cool, K.; Gaudreault, N.N.; Morozov, I.; Trujillo, J.D.; Meekins, D.A.; McDowell, C.; Carossino, M.; Bold, D.; Mitzel, D.; Kwon, T.; et al. Infection and Transmission of Ancestral SARS-CoV-2 and Its Alpha Variant in Pregnant White-Tailed Deer. Emerg. Microbes Infect. 2022, 11, 95–112. [Google Scholar] [CrossRef] [PubMed]
  134. Griffin, B.D.; Chan, M.; Tailor, N.; Mendoza, E.J.; Leung, A.; Warner, B.M.; Duggan, A.T.; Moffat, E.; He, S.; Garnett, L.; et al. SARS-CoV-2 Infection and Transmission in the North American Deer Mouse. Nat. Commun. 2021, 12, 3612. [Google Scholar] [CrossRef] [PubMed]
  135. Fagre, A.; Lewis, J.; Eckley, M.; Zhan, S.; Rocha, S.M.; Sexton, N.R.; Burke, B.; Geiss, B.; Peersen, O.; Bass, T.; et al. SARS-CoV-2 Infection, Neuropathogenesis and Transmission among Deer Mice: Implications for Spillback to New World Rodents. PLOS Pathog. 2021, 17, e1009585. [Google Scholar] [CrossRef] [PubMed]
  136. Bartlett, S.L.; Diel, D.G.; Wang, L.; Zec, S.; Laverack, M.; Martins, M.; Caserta, L.C.; Killian, M.L.; Terio, K.; Olmstead, C.; et al. SARS-COV-2 infection and longitudinal fecal screening in malayan tigers (Panthera Tigris Jacksoni), amur tigers (Panthera Tigris Altaica), and african lions (Panthera Leo Krugeri) at the bronx zoo, New York, USA. J. Zoo Wildl. Med. 2021, 51, 733–744. [Google Scholar] [CrossRef] [PubMed]
  137. McAloose, D.; Laverack, M.; Wang, L.; Killian, M.L.; Caserta, L.C.; Yuan, F.; Mitchell, P.K.; Queen, K.; Mauldin, M.R.; Cronk, B.D.; et al. From People to Panthera: Natural SARS-CoV-2 Infection in Tigers and Lions at the Bronx Zoo. mBio 2020, 11, e02220-20. [Google Scholar] [CrossRef]
  138. COVID-19. OIE-World Organisation for Animal Health. Available online: https://www.woah.org/ (accessed on 23 May 2021).
  139. Louisville Zoo Female Snow Leopard Tests Positive for SARS-CoV-2 (MEDIA RELEASE). Available online: https://louisvillezoo.org/louisville-zoo-female-snow-leopard-tests-positive-for-sars-cov-2-media-release/ (accessed on 21 October 2021).
  140. Decaro, N.; Grassi, A.; Lorusso, E.; Patterson, E.I.; Lorusso, A.; Desario, C.; Anderson, E.R.; Vasinioti, V.; Wastika, C.E.; Hughes, G.L.; et al. Long-Term Persistence of Neutralizing SARS-CoV-2 Antibodies in Pets. Transbound. Emerg. Dis. 2021. [Google Scholar] [CrossRef]
  141. Bosco-Lauth, A.M.; Hartwig, A.E.; Porter, S.M.; Gordy, P.W.; Nehring, M.; Byas, A.D.; VandeWoude, S.; Ragan, I.K.; Maison, R.M.; Bowen, R.A. Experimental Infection of Domestic Dogs and Cats with SARS-CoV-2: Pathogenesis, Transmission, and Response to Reexposure in Cats. Proc. Natl. Acad. Sci. USA 2020, 117, 26382–26388. [Google Scholar] [CrossRef]
  142. Jara, L.M.; Ferradas, C.; Schiaffino, F.; Sánchez-Carrión, C.; Martínez-Vela, A.; Ulloa, A.; Isasi-Rivas, G.; Montalván, A.; Sarmiento, L.G.; Fernández, M.; et al. Evidence of Neutralizing Antibodies against SARS-CoV-2 in Domestic Cats Living with Owners with a History of COVID-19 in Lima–Peru. One Health 2021, 13, 100318. [Google Scholar] [CrossRef]
  143. Halfmann, P.J.; Hatta, M.; Chiba, S.; Maemura, T.; Fan, S.; Takeda, M.; Kinoshita, N.; Hattori, S.-I.; Sakai-Tagawa, Y.; Iwatsuki-Horimoto, K.; et al. Transmission of SARS-CoV-2 in Domestic Cats. N. Engl. J. Med. 2020, 383, 592–594. [Google Scholar] [CrossRef]
  144. Zhai, X.; Sun, J.; Yan, Z.; Zhang, J.; Zhao, J.; Zhao, Z.; Gao, Q.; He, W.-T.; Veit, M.; Su, S. Comparison of Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein Binding to ACE2 Receptors from Human, Pets, Farm Animals, and Putative Intermediate Hosts. J. Virol. 2020, 94, e00831-20. [Google Scholar] [CrossRef]
  145. Sit, T.H.C.; Brackman, C.J.; Ip, S.M.; Tam, K.W.S.; Law, P.Y.T.; To, E.M.W.; Yu, V.Y.T.; Sims, L.D.; Tsang, D.N.C.; Chu, D.K.W.; et al. Infection of Dogs with SARS-CoV-2. Nature 2020, 586, 776–778. [Google Scholar] [CrossRef] [PubMed]
  146. Shi, J.; Wen, Z.; Zhong, G.; Yang, H.; Wang, C.; Huang, B.; Liu, R.; He, X.; Shuai, L.; Sun, Z.; et al. Susceptibility of Ferrets, Cats, Dogs, and Other Domesticated Animals to SARS-Coronavirus 2. Science 2020, 368, 1016–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Vergara-Alert, J.; Rodon, J.; Carrillo, J.; Te, N.; Izquierdo-Useros, N.; Rodríguez de la Concepción, M.L.; Ávila-Nieto, C.; Guallar, V.; Valencia, A.; Cantero, G.; et al. Pigs Are Not Susceptible to SARS-CoV-2 Infection but Are a Model for Viral Immunogenicity Studies. Transbound. Emerg. Dis. 2021, 68, 1721–1725. [Google Scholar] [CrossRef] [PubMed]
  148. Lara-Puente, J.H.; Carreño, J.M.; Sun, W.; Suárez-Martínez, A.; Ramírez-Martínez, L.; Quezada-Monroy, F.; Paz-De la Rosa, G.; Vigueras-Moreno, R.; Singh, G.; Rojas-Martínez, O.; et al. Safety and Immunogenicity of a Newcastle Disease Virus Vector-Based SARS-CoV-2 Vaccine Candidate, AVX/COVID-12-HEXAPRO (Patria), in Pigs. mBio 2021, 12, e01908-21. [Google Scholar] [CrossRef]
  149. Oreshkova, N.; Molenaar, R.J.; Vreman, S.; Harders, F.; Oude Munnink, B.B.; Hakze-van der Honing, R.W.; Gerhards, N.; Tolsma, P.; Bouwstra, R.; Sikkema, R.S.; et al. SARS-CoV-2 Infection in Farmed Minks, the Netherlands, April and May 2020. Euro. Surveill. 2020, 25, 2001005. [Google Scholar] [CrossRef]
  150. Oude Munnink, B.B.; Sikkema, R.S.; Nieuwenhuijse, D.F.; Molenaar, R.J.; Munger, E.; Molenkamp, R.; van der Spek, A.; Tolsma, P.; Rietveld, A.; Brouwer, M.; et al. Transmission of SARS-CoV-2 on Mink Farms between Humans and Mink and Back to Humans. Science 2021, 371, 172–177. [Google Scholar] [CrossRef]
  151. Hayashi, T.; Abiko, K.; Mandai, M.; Yaegashi, N.; Konishi, I. Highly Conserved Binding Region of ACE2 as a Receptor for SARS-CoV-2 between Humans and Mammals. Vet. Q. 2020, 40, 243–249. [Google Scholar] [CrossRef]
  152. Hoffmann, M.; Zhang, L.; Krüger, N.; Graichen, L.; Kleine-Weber, H.; Hofmann-Winkler, H.; Kempf, A.; Nessler, S.; Riggert, J.; Winkler, M.S.; et al. SARS-CoV-2 Mutations Acquired in Mink Reduce Antibody-Mediated Neutralization. Cell Rep. 2021, 35, 109017. [Google Scholar] [CrossRef]
  153. Molenaar, R.J.; Vreman, S.; Hakze-van der Honing, R.W.; Zwart, R.; de Rond, J.; Weesendorp, E.; Smit, L.A.M.; Koopmans, M.; Bouwstra, R.; Stegeman, A.; et al. Clinical and Pathological Findings in SARS-CoV-2 Disease Outbreaks in Farmed Mink (Neovison Vison). Vet. Pathol. 2020, 57, 653–657. [Google Scholar] [CrossRef]
  154. Bao, L.; Deng, W.; Huang, B.; Gao, H.; Liu, J.; Ren, L.; Wei, Q.; Yu, P.; Xu, Y.; Qi, F.; et al. The Pathogenicity of SARS-CoV-2 in HACE2 Transgenic Mice. Nature 2020, 583, 830–833. [Google Scholar] [CrossRef]
  155. Jiang, R.-D.; Liu, M.-Q.; Chen, Y.; Shan, C.; Zhou, Y.-W.; Shen, X.-R.; Li, Q.; Zhang, L.; Zhu, Y.; Si, H.-R.; et al. Pathogenesis of SARS-CoV-2 in Transgenic Mice Expressing Human Angiotensin-Converting Enzyme 2. Cell 2020, 182, 50–58.e8. [Google Scholar] [CrossRef] [PubMed]
  156. Khoury, D.S.; Wheatley, A.K.; Ramuta, M.D.; Reynaldi, A.; Cromer, D.; Subbarao, K.; O’Connor, D.H.; Kent, S.J.; Davenport, M.P. Measuring Immunity to SARS-CoV-2 Infection: Comparing Assays and Animal Models. Nat. Rev. Immunol. 2020, 20, 727–738. [Google Scholar] [CrossRef] [PubMed]
  157. Winkler, E.S.; Bailey, A.L.; Kafai, N.M.; Nair, S.; McCune, B.T.; Yu, J.; Fox, J.M.; Chen, R.E.; Earnest, J.T.; Keeler, S.P.; et al. SARS-CoV-2 Infection of Human ACE2-Transgenic Mice Causes Severe Lung Inflammation and Impaired Function. Nat. Immunol. 2020, 21, 1327–1335. [Google Scholar] [CrossRef] [PubMed]
  158. Oladunni, F.S.; Park, J.-G.; Pino, P.A.; Gonzalez, O.; Akhter, A.; Allué-Guardia, A.; Olmo-Fontánez, A.; Gautam, S.; Garcia-Vilanova, A.; Ye, C.; et al. Lethality of SARS-CoV-2 Infection in K18 Human Angiotensin-Converting Enzyme 2 Transgenic Mice. Nat. Commun. 2020, 11, 6122. [Google Scholar] [CrossRef]
  159. Hassan, A.O.; Case, J.B.; Winkler, E.S.; Thackray, L.B.; Kafai, N.M.; Bailey, A.L.; McCune, B.T.; Fox, J.M.; Chen, R.E.; Alsoussi, W.B.; et al. A SARS-CoV-2 Infection Model in Mice Demonstrates Protection by Neutralizing Antibodies. Cell 2020, 182, 744–753.e4. [Google Scholar] [CrossRef]
  160. Sun, J.; Zhuang, Z.; Zheng, J.; Li, K.; Wong, R.L.-Y.; Liu, D.; Huang, J.; He, J.; Zhu, A.; Zhao, J.; et al. Generation of a Broadly Useful Model for COVID-19 Pathogenesis, Vaccination, and Treatment. Cell 2020, 182, 734–743.e5. [Google Scholar] [CrossRef] [PubMed]
  161. Sun, S.-H.; Chen, Q.; Gu, H.-J.; Yang, G.; Wang, Y.-X.; Huang, X.-Y.; Liu, S.-S.; Zhang, N.-N.; Li, X.-F.; Xiong, R.; et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe 2020, 28, 124–133.e4. [Google Scholar] [CrossRef]
  162. Imai, M.; Iwatsuki-Horimoto, K.; Hatta, M.; Loeber, S.; Halfmann, P.J.; Nakajima, N.; Watanabe, T.; Ujie, M.; Takahashi, K.; Ito, M.; et al. Syrian Hamsters as a Small Animal Model for SARS-CoV-2 Infection and Countermeasure Development. Proc. Natl. Acad. Sci. USA 2020, 117, 16587–16595. [Google Scholar] [CrossRef]
  163. Sia, S.F.; Yan, L.-M.; Chin, A.W.H.; Fung, K.; Choy, K.-T.; Wong, A.Y.L.; Kaewpreedee, P.; Perera, R.A.P.M.; Poon, L.L.M.; Nicholls, J.M.; et al. Pathogenesis and Transmission of SARS-CoV-2 in Golden Hamsters. Nature 2020, 583, 834–838. [Google Scholar] [CrossRef]
  164. Chan, J.F.-W.; Zhang, A.J.; Yuan, S.; Poon, V.K.-M.; Chan, C.C.-S.; Lee, A.C.-Y.; Chan, W.-M.; Fan, Z.; Tsoi, H.-W.; Wen, L.; et al. Simulation of the Clinical and Pathological Manifestations of Coronavirus Disease 2019 (COVID-19) in Golden Syrian Hamster Model: Implications for Disease Pathogenesis and Transmissibility. Clin. Infect. Dis. 2020, 71, 2428–2446. [Google Scholar] [CrossRef]
  165. Trimpert, J.; Vladimirova, D.; Dietert, K.; Abdelgawad, A.; Kunec, D.; Dökel, S.; Voss, A.; Gruber, A.D.; Bertzbach, L.D.; Osterrieder, N. The Roborovski Dwarf Hamster Is A Highly Susceptible Model for a Rapid and Fatal Course of SARS-CoV-2 Infection. Cell Rep. 2020, 33, 108488. [Google Scholar] [CrossRef] [PubMed]
  166. Chu, Y.-K.; Ali, G.D.; Jia, F.; Li, Q.; Kelvin, D.; Couch, R.C.; Harrod, K.S.; Hutt, J.A.; Cameron, C.; Weiss, S.R.; et al. The SARS-CoV Ferret Model in an Infection-Challenge Study. Virology 2008, 374, 151–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Kim, Y.-I.; Kim, S.-G.; Kim, S.-M.; Kim, E.-H.; Park, S.-J.; Yu, K.-M.; Chang, J.-H.; Kim, E.J.; Lee, S.; Casel, M.A.B.; et al. Infection and Rapid Transmission of SARS-CoV-2 in Ferrets. Cell Host Microbe 2020, 27, 704–709.e2. [Google Scholar] [CrossRef] [PubMed]
  168. Richard, M.; Kok, A.; de Meulder, D.; Bestebroer, T.M.; Lamers, M.M.; Okba, N.M.A.; Fentener van Vlissingen, M.; Rockx, B.; Haagmans, B.L.; Koopmans, M.P.G.; et al. SARS-CoV-2 Is Transmitted via Contact and via the Air between Ferrets. Nat. Commun. 2020, 11, 3496. [Google Scholar] [CrossRef]
  169. Pandey, K.; Acharya, A.; Mohan, M.; Ng, C.L.; Reid, S.P.; Byrareddy, S.N. Animal Models for SARS-CoV-2 Research: A Comprehensive Literature Review. Transbound Emerg. Dis. 2020, 68, 1868–1885. [Google Scholar] [CrossRef]
  170. Salguero, F.J.; White, A.D.; Slack, G.S.; Fotheringham, S.A.; Bewley, K.R.; Gooch, K.E.; Longet, S.; Humphries, H.E.; Watson, R.J.; Hunter, L.; et al. Comparison of Rhesus and Cynomolgus Macaques as an Infection Model for COVID-19. Nat. Commun. 2021, 12, 1260. [Google Scholar] [CrossRef]
  171. Guebre-Xabier, M.; Patel, N.; Tian, J.-H.; Zhou, B.; Maciejewski, S.; Lam, K.; Portnoff, A.D.; Massare, M.J.; Frieman, M.B.; Piedra, P.A.; et al. NVX-CoV2373 Vaccine Protects Cynomolgus Macaque Upper and Lower Airways against SARS-CoV-2 Challenge. Vaccine 2020, 38, 7892–7896. [Google Scholar] [CrossRef]
  172. Characteristic and Quantifiable COVID-19-Like Abnormalities in CT- and PET/CT-Imaged Lungs of SARS-CoV-2-Infected Crab-Eating Macaques (Macaca Fascicularis)|BioRxiv. Available online: https://www.biorxiv.org/content/10.1101/2020.05.14.096727v1 (accessed on 1 June 2021).
  173. Munster, V.J.; Feldmann, F.; Williamson, B.N.; van Doremalen, N.; Pérez-Pérez, L.; Schulz, J.; Meade-White, K.; Okumura, A.; Callison, J.; Brumbaugh, B.; et al. Respiratory Disease in Rhesus Macaques Inoculated with SARS-CoV-2. Nature 2020, 585, 268–272. [Google Scholar] [CrossRef]
  174. Chandrashekar, A.; Liu, J.; Martinot, A.J.; McMahan, K.; Mercado, N.B.; Peter, L.; Tostanoski, L.H.; Yu, J.; Maliga, Z.; Nekorchuk, M.; et al. SARS-CoV-2 Infection Protects against Rechallenge in Rhesus Macaques. Science 2020, 369, 812–817. [Google Scholar] [CrossRef]
  175. Deng, W.; Bao, L.; Liu, J.; Xiao, C.; Liu, J.; Xue, J.; Lv, Q.; Qi, F.; Gao, H.; Yu, P.; et al. Primary Exposure to SARS-CoV-2 Protects against Reinfection in Rhesus Macaques. Science 2020, 369, 818–823. [Google Scholar] [CrossRef]
  176. Williamson, B.N.; Feldmann, F.; Schwarz, B.; Meade-White, K.; Porter, D.P.; Schulz, J.; van Doremalen, N.; Leighton, I.; Yinda, C.K.; Pérez-Pérez, L.; et al. Clinical Benefit of Remdesivir in Rhesus Macaques Infected with SARS-CoV-2. Nature 2020, 585, 273–276. [Google Scholar] [CrossRef] [PubMed]
  177. Bao, L.; Deng, W.; Gao, H.; Xiao, C.; Liu, J.; Xue, J.; Lv, Q.; Liu, J.; Yu, P.; Xu, Y.; et al. Lack of Reinfection in Rhesus Macaques Infected with SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  178. Shan, C.; Yao, Y.-F.; Yang, X.-L.; Zhou, Y.-W.; Gao, G.; Peng, Y.; Yang, L.; Hu, X.; Xiong, J.; Jiang, R.-D.; et al. Infection with Novel Coronavirus (SARS-CoV-2) Causes Pneumonia in Rhesus Macaques. Cell Res. 2020, 30, 670–677. [Google Scholar] [CrossRef] [PubMed]
  179. Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z.; et al. Development of an Inactivated Vaccine Candidate for SARS-CoV-2. Science 2020, 369, 77–81. [Google Scholar] [CrossRef] [PubMed]
  180. Deng, W.; Bao, L.; Gao, H.; Xiang, Z.; Qu, Y.; Song, Z.; Gong, S.; Liu, J.; Liu, J.; Yu, P.; et al. Ocular Conjunctival Inoculation of SARS-CoV-2 Can Cause Mild COVID-19 in Rhesus Macaques. Nat. Commun. 2020, 11, 4400. [Google Scholar] [CrossRef]
  181. Woolsey, C.; Borisevich, V.; Prasad, A.N.; Agans, K.N.; Deer, D.J.; Dobias, N.S.; Heymann, J.C.; Foster, S.L.; Levine, C.B.; Medina, L.; et al. Establishment of an African Green Monkey Model for COVID-19 and Protection against Re-Infection. Nat. Immunol. 2021, 22, 86–98. [Google Scholar] [CrossRef]
  182. Blair, R.V.; Vaccari, M.; Doyle-Meyers, L.A.; Roy, C.J.; Russell-Lodrigue, K.; Fahlberg, M.; Monjure, C.J.; Beddingfield, B.; Plante, K.S.; Plante, J.A.; et al. Acute Respiratory Distress and Cytokine Storm in Aged, SARS-CoV-2 Infected African Green Monkeys, but Not in Rhesus Macaques. bioRxiv 2020. [Google Scholar] [CrossRef]
  183. Cross, R.W.; Agans, K.N.; Prasad, A.N.; Borisevich, V.; Woolsey, C.; Deer, D.J.; Dobias, N.S.; Geisbert, J.B.; Fenton, K.A.; Geisbert, T.W. Intranasal Exposure of African Green Monkeys to SARS-CoV-2 Results in Acute Phase Pneumonia with Shedding and Lung Injury Still Present in the Early Convalescence Phase. Virol. J. 2020, 17, 125. [Google Scholar] [CrossRef]
  184. Rathnasinghe, R.; Strohmeier, S.; Amanat, F.; Gillespie, V.L.; Krammer, F.; García-Sastre, A.; Coughlan, L.; Schotsaert, M.; Uccellini, M.B. Comparison of Transgenic and Adenovirus HACE2 Mouse Models for SARS-CoV-2 Infection. Emerg. Microbes Infect. 2020, 9, 2433–2445. [Google Scholar] [CrossRef]
  185. Singh, D.K.; Ganatra, S.R.; Singh, B.; Cole, J.; Alfson, K.J.; Clemmons, E.; Gazi, M.; Gonzalez, O.; Escobedo, R.; Lee, T.-H.; et al. SARS-CoV-2 Infection Leads to Acute Infection with Dynamic Cellular and Inflammatory Flux in the Lung That Varies across Nonhuman Primate Species. bioRxiv 2020. [Google Scholar] [CrossRef]
  186. Greenough, T.C.; Carville, A.; Coderre, J.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Mansfield, K. Pneumonitis and Multi-Organ System Disease in Common Marmosets (Callithrix jacchus) Infected with the Severe Acute Respiratory Syndrome-Associated Coronavirus. Am. J. Pathol. 2005, 167, 455–463. [Google Scholar] [CrossRef] [Green Version]
  187. Menachery, V.D.; Yount, B.L.; Sims, A.C.; Debbink, K.; Agnihothram, S.S.; Gralinski, L.E.; Graham, R.L.; Scobey, T.; Plante, J.A.; Royal, S.R.; et al. SARS-like WIV1-CoV Poised for Human Emergence. Proc. Natl. Acad. Sci. USA 2016, 113, 3048–3053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Coleman, C.M.; Matthews, K.L.; Goicochea, L.; Frieman, M.B. Wild-Type and Innate Immune-Deficient Mice Are Not Susceptible to the Middle East Respiratory Syndrome Coronavirus. J. Gen. Virol. 2014, 95, 408–412. [Google Scholar] [CrossRef]
  189. Frieman, M.; Yount, B.; Agnihothram, S.; Page, C.; Donaldson, E.; Roberts, A.; Vogel, L.; Woodruff, B.; Scorpio, D.; Subbarao, K.; et al. Molecular Determinants of Severe Acute Respiratory Syndrome Coronavirus Pathogenesis and Virulence in Young and Aged Mouse Models of Human Disease. J. Virol. 2012, 86, 884–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Dinnon, K.H.; Leist, S.R.; Schäfer, A.; Edwards, C.E.; Martinez, D.R.; Montgomery, S.A.; West, A.; Yount, B.L.; Hou, Y.J.; Adams, L.E.; et al. A Mouse-Adapted Model of SARS-CoV-2 to Test COVID-19 Countermeasures. Nature 2020, 586, 560–566. [Google Scholar] [CrossRef] [PubMed]
  191. Wang, Q. HACE2 Transgenic Mouse Model for Coronavirus (COVID-19) Research. Available online: https://www.jax.org/news-and-insights/2020/february/introducing-mouse-model-for-corona-virus (accessed on 20 July 2021).
  192. McCray, P.B.; Pewe, L.; Wohlford-Lenane, C.; Hickey, M.; Manzel, L.; Shi, L.; Netland, J.; Jia, H.P.; Halabi, C.; Sigmund, C.D.; et al. Lethal Infection of K18-HACE2 Mice Infected with Severe Acute Respiratory Syndrome Coronavirus. J. Virol. 2007, 81, 813–821. [Google Scholar] [CrossRef] [Green Version]
  193. Moreau, G.B.; Burgess, S.L.; Sturek, J.M.; Donlan, A.N.; Petri, W.A.; Mann, B.J. Evaluation of K18-HACE2 Mice as a Model of SARS-CoV-2 Infection. Am. J. Trop. Med. Hyg. 2020, 103, 1215–1219. [Google Scholar] [CrossRef]
  194. Hong, W.; Yang, J.; Bi, Z.; He, C.; Lei, H.; Yu, W.; Yang, Y.; Fan, C.; Lu, S.; Peng, X.; et al. A Mouse Model for SARS-CoV-2-Induced Acute Respiratory Distress Syndrome. Signal. Transduct. Target. Ther. 2021, 6, 1. [Google Scholar] [CrossRef]
  195. Jarnagin, K.; Alvarez, O.; Shresta, S.; Webb, D.R. Animal Models for SARS-Cov2/Covid19 Research-A Commentary. Biochem. Pharmacol. 2021, 188, 114543. [Google Scholar] [CrossRef]
  196. Soldatov, V.O.; Kubekina, M.V.; Silaeva, Y.Y.; Bruter, A.V.; Deykin, A.V. On the Way from SARS-CoV-Sensitive Mice to Murine COVID-19 Model. Res. Results Pharmacol. 2020, 6, 1–7. [Google Scholar] [CrossRef]
  197. Gurumurthy, C.B.; Quadros, R.M.; Richardson, G.P.; Poluektova, L.Y.; Mansour, S.L.; Ohtsuka, M. Genetically Modified Mouse Models to Help Fight COVID-19. Nat. Protoc. 2020, 15, 3777–3787. [Google Scholar] [CrossRef] [PubMed]
  198. Gu, H.; Chen, Q.; Yang, G.; He, L.; Fan, H.; Deng, Y.-Q.; Wang, Y.; Teng, Y.; Zhao, Z.; Cui, Y.; et al. Adaptation of SARS-CoV-2 in BALB/c Mice for Testing Vaccine Efficacy. Science 2020, 369, 1603–1607. [Google Scholar] [CrossRef] [PubMed]
  199. Leist, S.R.; Dinnon, K.H.; Schäfer, A.; Tse, L.V.; Okuda, K.; Hou, Y.J.; West, A.; Edwards, C.E.; Sanders, W.; Fritch, E.J.; et al. A Mouse-Adapted SARS-CoV-2 Induces Acute Lung Injury and Mortality in Standard Laboratory Mice. Cell 2020, 183, 1070–1085.e12. [Google Scholar] [CrossRef] [PubMed]
  200. Huang, K.; Zhang, Y.; Hui, X.; Zhao, Y.; Gong, W.; Wang, T.; Zhang, S.; Yang, Y.; Deng, F.; Zhang, Q.; et al. Q493K and Q498H Substitutions in Spike Promote Adaptation of SARS-CoV-2 in Mice. EBioMedicine 2021, 67, 103381. [Google Scholar] [CrossRef]
  201. Roberts, A.; Thomas, W.D.; Guarner, J.; Lamirande, E.W.; Babcock, G.J.; Greenough, T.C.; Vogel, L.; Hayes, N.; Sullivan, J.L.; Zaki, S.; et al. Therapy with a Severe Acute Respiratory Syndrome-Associated Coronavirus-Neutralizing Human Monoclonal Antibody Reduces Disease Severity and Viral Burden in Golden Syrian Hamsters. J. Infect. Dis. 2006, 193, 685–692. [Google Scholar] [CrossRef]
  202. Roberts, A.; Vogel, L.; Guarner, J.; Hayes, N.; Murphy, B.; Zaki, S.; Subbarao, K. Severe Acute Respiratory Syndrome Coronavirus Infection of Golden Syrian Hamsters. J. Virol. 2005, 79, 503–511. [Google Scholar] [CrossRef] [Green Version]
  203. Schaecher, S.R.; Stabenow, J.; Oberle, C.; Schriewer, J.; Buller, R.M.; Sagartz, J.E.; Pekosz, A. An Immunosuppressed Syrian Golden Hamster Model for SARS-CoV Infection. Virology 2008, 380, 312–321. [Google Scholar] [CrossRef] [Green Version]
  204. Boudewijns, R.; Thibaut, H.J.; Kaptein, S.J.F.; Li, R.; Vergote, V.; Seldeslachts, L.; Van Weyenbergh, J.; De Keyzer, C.; Bervoets, L.; Sharma, S.; et al. STAT2 Signaling Restricts Viral Dissemination but Drives Severe Pneumonia in SARS-CoV-2 Infected Hamsters. Nat. Commun. 2020, 11, 5838. [Google Scholar] [CrossRef]
  205. Osterrieder, N.; Bertzbach, L.D.; Dietert, K.; Abdelgawad, A.; Vladimirova, D.; Kunec, D.; Hoffmann, D.; Beer, M.; Gruber, A.D.; Trimpert, J. Age-Dependent Progression of SARS-CoV-2 Infection in Syrian Hamsters. Viruses 2020, 12, 779. [Google Scholar] [CrossRef]
  206. Chan, J.F.-W.; Yuan, S.; Zhang, A.J.; Poon, V.K.-M.; Chan, C.C.-S.; Lee, A.C.-Y.; Fan, Z.; Li, C.; Liang, R.; Cao, J.; et al. Surgical Mask Partition Reduces the Risk of Noncontact Transmission in a Golden Syrian Hamster Model for Coronavirus Disease 2019 (COVID-19). Clin. Infect. Dis. 2020, 71, 2139–2149. [Google Scholar] [CrossRef]
  207. Port, J.R.; Yinda, C.K.; Owusu, I.O.; Holbrook, M.; Fischer, R.; Bushmaker, T.; Avanzato, V.A.; Schulz, J.E.; Martens, C.; van Doremalen, N.; et al. SARS-CoV-2 Disease Severity and Transmission Efficiency Is Increased for Airborne Compared to Fomite Exposure in Syrian Hamsters. Nat. Commun. 2021, 12, 4985. [Google Scholar] [CrossRef] [PubMed]
  208. Mok, B.W.-Y.; Liu, H.; Deng, S.; Liu, J.; Zhang, A.J.; Lau, S.-Y.; Liu, S.; Tam, R.C.-Y.; Cremin, C.J.; Ng, T.T.-L.; et al. Low Dose Inocula of SARS-CoV-2 Alpha Variant Transmits More Efficiently than Earlier Variants in Hamsters. Commun. Biol. 2021, 4, 1102. [Google Scholar] [CrossRef] [PubMed]
  209. Li, T.; Cai, H.; Yao, H.; Zhou, B.; Zhang, N.; van Vlissingen, M.F.; Kuiken, T.; Han, W.; GeurtsvanKessel, C.H.; Gong, Y.; et al. A Synthetic Nanobody Targeting RBD Protects Hamsters from SARS-CoV-2 Infection. Nat. Commun. 2021, 12, 4635. [Google Scholar] [CrossRef] [PubMed]
  210. Huo, J.; Mikolajek, H.; Le Bas, A.; Clark, J.J.; Sharma, P.; Kipar, A.; Dormon, J.; Norman, C.; Weckener, M.; Clare, D.K.; et al. A Potent SARS-CoV-2 Neutralising Nanobody Shows Therapeutic Efficacy in the Syrian Golden Hamster Model of COVID-19. Nat. Commun. 2021, 12, 5469. [Google Scholar] [CrossRef]
  211. Driouich, J.-S.; Cochin, M.; Lingas, G.; Moureau, G.; Touret, F.; Petit, P.-R.; Piorkowski, G.; Barthélémy, K.; Laprie, C.; Coutard, B.; et al. Favipiravir Antiviral Efficacy against SARS-CoV-2 in a Hamster Model. Nat. Commun. 2021, 12, 1735. [Google Scholar] [CrossRef]
  212. Langel, S.N.; Johnson, S.; Martinez, C.I.; Tedjakusuma, S.N.; Peinovich, N.; Dora, E.G.; Kuehl, P.J.; Irshad, H.; Barrett, E.G.; Werts, A.; et al. Adenovirus Type 5 SARS-CoV-2 Vaccines Delivered Orally or Intranasally Reduced Disease Severity and Transmission in a Hamster Model. Sci. Transl. Med. 2022, eabn6868. [Google Scholar] [CrossRef]
  213. Zhou, B.; Zhou, R.; Tang, B.; Chan, J.F.-W.; Luo, M.; Peng, Q.; Yuan, S.; Liu, H.; Mok, B.W.-Y.; Chen, B.; et al. A Broadly Neutralizing Antibody Protects Syrian Hamsters against SARS-CoV-2 Omicron Challenge. Nat. Commun. 2022, 13, 3589. [Google Scholar] [CrossRef]
  214. Bertzbach, L.D.; Vladimirova, D.; Dietert, K.; Abdelgawad, A.; Gruber, A.D.; Osterrieder, N.; Trimpert, J. SARS-CoV-2 Infection of Chinese Hamsters (Cricetulus griseus) Reproduces COVID-19 Pneumonia in a Well-Established Small Animal Model. Transbound. Emerg. Dis. 2021, 68, 1075–1079. [Google Scholar] [CrossRef]
  215. Wang, L.; Wang, C.; Zhang, R.; Liu, Y.; Wang, C.; Song, G.; Yu, J.; Chen, Z. Phenotypic Characterization of a Novel Type 2 Diabetes Animal Model in a SHANXI MU Colony of Chinese Hamsters. Endocrine 2019, 65, 61–72. [Google Scholar] [CrossRef]
  216. Lieber, C.M.; Cox, R.M.; Sourimant, J.; Wolf, J.D.; Juergens, K.; Phung, Q.; Saindane, M.T.; Natchus, M.G.; Painter, G.R.; Sakamoto, K.; et al. SARS-CoV-2 Variant of Concern Type and Biological Sex Affect Efficacy of Molnupiravir in Dwarf Hamster Model of Severe COVID-19. bioRxiv 2022. [Google Scholar] [CrossRef]
  217. Renegar, K.B. Influenza Virus Infections and Immunity: A Review of Human and Animal Models. Lab. Anim. Sci. 1992, 42, 222–232. [Google Scholar] [PubMed]
  218. Belser, J.A.; Eckert, A.M.; Tumpey, T.M.; Maines, T.R. Complexities in Ferret Influenza Virus Pathogenesis and Transmission Models. Microbiol. Mol. Biol. Rev. 2016, 80, 733–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Maher, J.A.; DeStefano, J. The Ferret: An Animal Model to Study Influenza Virus. Lab. Anim. 2004, 33, 50–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Smith, W.; Andrewes, C.H.; Laidlaw, P.P. A virus obtained from influenza patients. Lancet 1933, 222, 66–68. [Google Scholar] [CrossRef] [Green Version]
  221. Martina, B.E.E.; Haagmans, B.L.; Kuiken, T.; Fouchier, R.A.M.; Rimmelzwaan, G.F.; van Amerongen, G.; Peiris, J.S.M.; Lim, W.; Osterhaus, A.D.M.E. SARS Virus Infection of Cats and Ferrets. Nature 2003, 425, 915. [Google Scholar] [CrossRef]
  222. Baker, A.N.; Richards, S.-J.; Guy, C.S.; Congdon, T.R.; Hasan, M.; Zwetsloot, A.J.; Gallo, A.; Lewandowski, J.R.; Stansfeld, P.J.; Straube, A.; et al. The SARS-COV-2 Spike Protein Binds Sialic Acids and Enables Rapid Detection in a Lateral Flow Point of Care Diagnostic Device. ACS Cent. Sci. 2020, 6, 2046–2052. [Google Scholar] [CrossRef]
  223. Tortorici, M.A.; Walls, A.C.; Lang, Y.; Wang, C.; Li, Z.; Koerhuis, D.; Boons, G.-J.; Bosch, B.-J.; Rey, F.A.; de Groot, R.J.; et al. Structural Basis for Human Coronavirus Attachment to Sialic Acid Receptors. Nat. Struct. Mol. Biol. 2019, 26, 481–489. [Google Scholar] [CrossRef] [Green Version]
  224. Leung, H.S.Y.; Li, O.T.W.; Chan, R.W.Y.; Chan, M.C.W.; Nicholls, J.M.; Poon, L.L.M. Entry of Influenza A Virus with a A2,6-Linked Sialic Acid Binding Preference Requires Host Fibronectin. J. Virol. 2012, 86, 10704–10713. [Google Scholar] [CrossRef] [Green Version]
  225. Wan, Y.; Shang, J.; Graham, R.; Baric, R.S.; Li, F. Receptor Recognition by the Novel Coronavirus from Wuhan: An Analysis Based on Decade-Long Structural Studies of SARS Coronavirus. J. Virol. 2020, 94, e00127-20. [Google Scholar] [CrossRef] [Green Version]
  226. van Riel, D.; Munster, V.J.; de Wit, E.; Rimmelzwaan, G.F.; Fouchier, R.A.M.; Osterhaus, A.D.M.E.; Kuiken, T. Human and Avian Influenza Viruses Target Different Cells in the Lower Respiratory Tract of Humans and Other Mammals. Am. J. Pathol. 2007, 171, 1215–1223. [Google Scholar] [CrossRef] [Green Version]
  227. Cox, R.M.; Wolf, J.D.; Plemper, R.K. Therapeutically Administered Ribonucleoside Analogue MK-4482/EIDD-2801 Blocks SARS-CoV-2 Transmission in Ferrets. Nat. Microbiol. 2021, 6, 11–18. [Google Scholar] [CrossRef] [PubMed]
  228. Kutter, J.S.; de Meulder, D.; Bestebroer, T.M.; Lexmond, P.; Mulders, A.; Richard, M.; Fouchier, R.A.M.; Herfst, S. SARS-CoV and SARS-CoV-2 Are Transmitted through the Air between Ferrets over More than One Meter Distance. Nat. Commun. 2021, 12, 1653. [Google Scholar] [CrossRef] [PubMed]
  229. Park, S.-J.; Yu, K.-M.; Kim, Y.-I.; Kim, S.-M.; Kim, E.-H.; Kim, S.-G.; Kim, E.J.; Casel, M.A.B.; Rollon, R.; Jang, S.-G.; et al. Antiviral Efficacies of FDA-Approved Drugs against SARS-CoV-2 Infection in Ferrets. mBio 2020, 11, e01114-20. [Google Scholar] [CrossRef] [PubMed]
  230. Ryan, K.A.; Bewley, K.R.; Fotheringham, S.A.; Slack, G.S.; Brown, P.; Hall, Y.; Wand, N.I.; Marriott, A.C.; Cavell, B.E.; Tree, J.A.; et al. Dose-Dependent Response to Infection with SARS-CoV-2 in the Ferret Model and Evidence of Protective Immunity. Nat. Commun. 2021, 12, 81. [Google Scholar] [CrossRef] [PubMed]
  231. Lee, J.S.; Koh, J.-Y.; Yi, K.; Kim, Y.-I.; Park, S.-J.; Kim, E.-H.; Kim, S.-M.; Park, S.H.; Ju, Y.S.; Choi, Y.K.; et al. Single-Cell Transcriptome of Bronchoalveolar Lavage Fluid Reveals Sequential Change of Macrophages during SARS-CoV-2 Infection in Ferrets. Nat. Commun. 2021, 12, 4567. [Google Scholar] [CrossRef]
  232. Feng, Y.; Feng, Y.-M.; Lu, C.; Han, Y.; Liu, L.; Sun, X.; Dai, J.; Xia, X. Tree Shrew, a Potential Animal Model for Hepatitis C, Supports the Infection and Replication of HCV in Vitro and in Vivo. J. Gen. Virol. 2017, 98, 2069–2078. [Google Scholar] [CrossRef]
  233. Xu, L.; Yu, D.-D.; Ma, Y.-H.; Yao, Y.-L.; Luo, R.-H.; Feng, X.-L.; Cai, H.-R.; Han, J.-B.; Wang, X.-H.; Li, M.-H.; et al. COVID-19-like Symptoms Observed in Chinese Tree Shrews Infected with SARS-CoV-2. Zool. Res. 2020, 41, 517–526. [Google Scholar] [CrossRef]
  234. Zhao, Y.; Wang, J.; Kuang, D.; Xu, J.; Yang, M.; Ma, C.; Zhao, S.; Li, J.; Long, H.; Ding, K.; et al. Susceptibility of Tree Shrew to SARS-CoV-2 Infection. Sci. Rep. 2020, 10, 16007. [Google Scholar] [CrossRef]
  235. Muthumani, K.; Falzarano, D.; Reuschel, E.L.; Tingey, C.; Flingai, S.; Villarreal, D.O.; Wise, M.; Patel, A.; Izmirly, A.; Aljuaid, A.; et al. A Synthetic Consensus Anti-Spike Protein DNA Vaccine Induces Protective Immunity against Middle East Respiratory Syndrome Coronavirus in Nonhuman Primates. Sci. Transl. Med. 2015, 7, 301ra132. [Google Scholar] [CrossRef] [Green Version]
  236. Wang, L.; Shi, W.; Joyce, M.G.; Modjarrad, K.; Zhang, Y.; Leung, K.; Lees, C.R.; Zhou, T.; Yassine, H.M.; Kanekiyo, M.; et al. Evaluation of Candidate Vaccine Approaches for MERS-CoV. Nat. Commun. 2015, 6, 7712. [Google Scholar] [CrossRef]
  237. McDonald, I.; Murray, S.M.; Reynolds, C.J.; Altmann, D.M.; Boyton, R.J. Comparative Systematic Review and Meta-Analysis of Reactogenicity, Immunogenicity and Efficacy of Vaccines against SARS-CoV-2. NPJ Vaccines 2021, 6, 74. [Google Scholar] [CrossRef] [PubMed]
  238. Lu, S.; Zhao, Y.; Yu, W.; Yang, Y.; Gao, J.; Wang, J.; Kuang, D.; Yang, M.; Yang, J.; Ma, C.; et al. Comparison of Nonhuman Primates Identified the Suitable Model for COVID-19. Signal Transduct. Target. Ther. 2020, 5, 157. [Google Scholar] [CrossRef] [PubMed]
  239. Singh, D.K.; Singh, B.; Ganatra, S.R.; Gazi, M.; Cole, J.; Thippeshappa, R.; Alfson, K.J.; Clemmons, E.; Gonzalez, O.; Escobedo, R.; et al. Responses to Acute Infection with SARS-CoV-2 in the Lungs of Rhesus Macaques, Baboons and Marmosets. Nat. Microbiol. 2021, 6, 73–86. [Google Scholar] [CrossRef] [PubMed]
  240. Hartman, A.L.; Nambulli, S.; McMillen, C.M.; White, A.G.; Tilston-Lunel, N.L.; Albe, J.R.; Cottle, E.; Dunn, M.D.; Frye, L.J.; Gilliland, T.H.; et al. SARS-CoV-2 Infection of African Green Monkeys Results in Mild Respiratory Disease Discernible by PET/CT Imaging and Shedding of Infectious Virus from Both Respiratory and Gastrointestinal Tracts. PLoS Pathog. 2020, 16, e1008903. [Google Scholar] [CrossRef] [PubMed]
  241. Johnston, S.C.; Ricks, K.M.; Jay, A.; Raymond, J.L.; Rossi, F.; Zeng, X.; Scruggs, J.; Dyer, D.; Frick, O.; Koehler, J.W.; et al. Development of a Coronavirus Disease 2019 Nonhuman Primate Model Using Airborne Exposure. PLoS ONE 2021, 16, e0246366. [Google Scholar] [CrossRef]
  242. Chang, M.C.; Hild, S.; Grieder, F. Nonhuman Primate Models for SARS-CoV-2 Research: Consider Alternatives to Macaques. Lab. Anim. 2021, 50, 113–114. [Google Scholar] [CrossRef]
  243. Che, Y.; Liu, X.; Pu, Y.; Zhou, M.; Zhao, Z.; Jiang, R.; Yin, Z.; Xu, M.; Yin, Q.; Wang, J.; et al. Randomized, Double-Blinded, Placebo-Controlled Phase 2 Trial of an Inactivated Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine in Healthy Adults. Clin. Infect. Dis. 2020, 73, e3949–e3955. [Google Scholar] [CrossRef]
  244. Song, T.-Z.; Zheng, H.-Y.; Han, J.-B.; Jin, L.; Yang, X.; Liu, F.-L.; Luo, R.-H.; Tian, R.-R.; Cai, H.-R.; Feng, X.-L.; et al. Delayed Severe Cytokine Storm and Immune Cell Infiltration in SARS-CoV-2-Infected Aged Chinese Rhesus Macaques. Zool. Res. 2020, 41, 503–516. [Google Scholar] [CrossRef]
  245. Ishigaki, H.; Nakayama, M.; Kitagawa, Y.; Nguyen, C.T.; Hayashi, K.; Shiohara, M.; Gotoh, B.; Itoh, Y. Neutralizing Antibody-Dependent and -Independent Immune Responses against SARS-CoV-2 in Cynomolgus Macaques. Virology 2021, 554, 97–105. [Google Scholar] [CrossRef]
  246. Corbett, K.S.; Flynn, B.; Foulds, K.E.; Francica, J.R.; Boyoglu-Barnum, S.; Werner, A.P.; Flach, B.; O’Connell, S.; Bock, K.W.; Minai, M.; et al. Evaluation of the MRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates. N. Engl. J. Med. 2020, 383, 1544–1555. [Google Scholar] [CrossRef]
  247. Vogel, A.B.; Kanevsky, I.; Che, Y.; Swanson, K.A.; Muik, A.; Vormehr, M.; Kranz, L.M.; Walzer, K.C.; Hein, S.; Güler, A.; et al. BNT162b Vaccines Protect Rhesus Macaques from SARS-CoV-2. Nature 2021, 592, 283–289. [Google Scholar] [CrossRef] [PubMed]
  248. van Doremalen, N.; Lambe, T.; Spencer, A.; Belij-Rammerstorfer, S.; Purushotham, J.N.; Port, J.R.; Avanzato, V.A.; Bushmaker, T.; Flaxman, A.; Ulaszewska, M.; et al. ChAdOx1 NCoV-19 Vaccine Prevents SARS-CoV-2 Pneumonia in Rhesus Macaques. Nature 2020, 586, 578–582. [Google Scholar] [CrossRef] [PubMed]
  249. Mercado, N.B.; Zahn, R.; Wegmann, F.; Loos, C.; Chandrashekar, A.; Yu, J.; Liu, J.; Peter, L.; McMahan, K.; Tostanoski, L.H.; et al. Single-Shot Ad26 Vaccine Protects against SARS-CoV-2 in Rhesus Macaques. Nature 2020, 586, 583–588. [Google Scholar] [CrossRef]
  250. Jones, K.E.; Patel, N.G.; Levy, M.A.; Storeygard, A.; Balk, D.; Gittleman, J.L.; Daszak, P. Global Trends in Emerging Infectious Diseases. Nature 2008, 451, 990–993. [Google Scholar] [CrossRef] [PubMed]
Table 1. Cell-based approaches: key points, benefits, and limitations.
Table 1. Cell-based approaches: key points, benefits, and limitations.
TypeName (Origin)InvestigationBenefitsDrawbacksReferences
Immortalized cellsVero E6
(African green monkey kidney
epithelial cells)
WT
  • Study of infection mechanism and viral isolation
  • In vitro replication and amplification of viral particles
  • Pharmacological screening
  • Easy to cultivate
  • High replication rate
  • ACE2 expression
  • CPE
  • Clinical strain isolation and production
  • Viral vectors and vaccines production
  • No TMPRSS2 receptor expression
  • Non-human cells
  • No direct target cell (kidney cells)
[4,25,32,33]
TMPRSS2-cells
  • Easy to cultivate
  • Higher replication rate compared to WT
  • ACE2 and TMPRSS2 expression
  • CPE
  • Clinical strain isolation and production
  • Non-human cells
  • Not direct virus target (kidney cells)
[30,32,34]
A549
(Human lung carcinoma)
  • Study of infection mechanism
  • Comparing tropism, replication kinetics, and cell damage profiling of SARS-CoV-2
  • Easy to cultivate
  • Moderate replication rate
  • Basal expression level of ACE2 and no TMPRSS2 expression
  • hACE2 and hACE2-TMPRSS2 cells commercially available
  • Main target cell (lung cells)
  • No TMPRSS2 receptor expression
  • No CPE
  • Minimal viral replication in WT cells without the addition of exogenous proteases
[5,30,33,34,35,36]
Caco-2
(Human colorectal adenocarcinoma)
  • Study of infection mechanism
  • Pharmacological screening
  • Easy to cultivate
  • High replication rate
  • ACE2 and TMPRSS2 expression
  • Main target cell (colon cells)
  • No CPE
  • Minimal viral replication
[37,38,39]
Calu-3
(Human non-small lung adenocarcinoma)
  • Study of infection mechanism
  • Pharmacological screening
  • Easy to cultivate
  • High replication rate
  • ACE2 and TMPRSS2 expression
  • Main target cell (lung cells)
  • No CPE
  • Laborious cell culture (time-consuming, low replicative rate)
  • Minimal viral replication
  • Differentiation stage
[30,31,32,35,40,41]
HEK293T
(Human embryonic kidney cells)
  • Study of infection mechanism
  • Pharmacological screening
  • Easy to cultivate
  • Modest replication rate
  • ACE2 and TMPRSS2 expression
  • No CPE
  • Minimal viral replication
  • Not direct virus target (kidney cells)
[29,42,43,44,45,46,47,48]
Huh7
(Human hepatocellular carcinoma)
  • Study of infection mechanism and viral isolation
  • Pharmacological screening
  • Study of tropism, replication kinetics and cell damage profiling of SARS-CoV-2
  • Easy to cultivate
  • High replication rate
  • ACE2 and TMPRSS2 expression
  • No CPE
  • Low titer of infectious SARS-CoV-2
  • Not direct virus target (liver cells)
  • No ACE2 expression
[35,42,49,50,51]
Beas-2B
(Human lung, bronchus)
  • Study of infection mechanism
  • Easy to cultivate
  • Moderate replication rate
  • CPE
  • hACE2 cells production possible
  • Controversial ACE2 receptor expression
[52,53]
Primary cellsHAE/HAE-ALI
(Human airway epithelium)
  • Study of infection mechanism
  • Model closest to human lungs
  • Diagnosis of SARS-CoV-2 infection and viral isolation
  • Cytokine response profiling and sensitivity to interferons
  • Pharmacological screening
  • More complex culture and short lifetime
  • ACE2 and TMPRSS2 expression
  • CPE
  • Understanding pro-inflammatory responses of proximal and distal lung epithelium
  • Study of immune signature
  • Uncontrolled region-to-region and donor-to-donor variation
  • Limited availability of resources and expensive
  • Not possible to study viral evolution
[54,55,56,57,58]
OrganoidsHuman bronchial/lung organoid
  • Study of infection mechanism
  • Pharmacological screening
  • Mapping of genes associated with SARS-CoV-2 infection
  • Understanding the tissue tropism of SARS-CoV-2
  • More complex culture
  • High titers of infectious viral particles depending on target organ
  • ACE2 and TMPRSS2 expression depending on studied organ
  • Permissive to SARS-CoV-2 infection
  • Contain a full range of differentiated cell types
  • Lack of air-liquid interface, vasculature, and immune cells
  • Absence of relevant mechanical cues (air flow, vascular flow)
  • Thick ECM gel complicates permeability and drug studies
  • Not fully recapitulate immune cell population
[59,60,61,62,63,64,65]
Human colonic/intestinal organoid
Organ-on-a-chipPrimary lung/intestinal epithelium-on-a-chip
  • Study of infection mechanism
  • Pharmacological screening
  • Mapping of genes associated with SARS-CoV-2 infection
  • Understanding the tissue tropism of SARS-CoV-2
  • Immune response
  • Complex culture
  • High titers of infectious viral particles
  • ACE2 and TMPRSS2 expression depending on studied organ
  • Tissue-tissue interfaces
  • Organ-level physical microenvironments
  • Relevant mechanical cues, and fluid flow
  • Closest model to mimic human physiopathology
  • Relatively low throughput
  • Reproducibility issues
[64,66,67,68]
ACE2: angiotensin-converting enzyme 2, ALI: airway-liquid interface, CPE: cytopathic effect, ECM: extracellular matrix, HAE: human airway epithelium, TMPRSS2: transmembrane protease serine 2, WT: wild type.
Table 2. Animal models: key points, benefits, and limitations in COVID-19 investigation.
Table 2. Animal models: key points, benefits, and limitations in COVID-19 investigation.
TypeModelKey PointsBenefitsDrawbacksReferences
Small animalsMice h ACE2Mouse Ace2 promoter
High susceptibility to SARS-CoV-2 infection
Experience mild symptoms of COVID-19 patients
COVID-19:
Absence-to-lethal symptoms: pneumonia, weight loss, reduce activity, increased respiration, cytokine storm
Screening of antiviral drugs and vaccines
Different susceptibility based on gender
Model:
Rapid metabolism
COVID-19:
Do not develop severe disease
Limited availability during pandemic
S protein do not favorably interact with mice ACE2 receptor, must develop transgenic models
Potential mouse-to-mouse variation in hACE2 expression and tissue distribution
Lethality after brain dissemination
[154]
Mouse Hfh4 promoter
Valuable model for vaccines and drugs assays
[155,156]
Mouse K18 promoter
Investigation of lung
Immune and antiviral-based countermeasures
[156,157,158]
Adenoviral
vector
Viable model (no brain migration)
Allow not permanent genetic mutation
Artificial ACE2 expression in non-relevant cell types
[156,159,160]
CRISPR–Cas9 knock-in
Artificial ACE2 expression in relevant cell types
High levels of hACE2 expression in lung
[156,161]
Golden Syrian
hamsters
Develop severe pneumonia similar to COVID-19 patients
Efficiently transmitted from inoculated hamsters to naive hamsters by direct contact and via aerosols
SARS-CoV-2-infected hamsters can develop neutralizing antibodies, protecting them from reinfection
Develop immunity against reinfection
COVID-19:
ACE2 expression similar to human
Susceptible to SARS-CoV-2 infection
Mild-to-moderate symptoms: weight loss, hunched posture, lethargy, respiratory distress
Sex and age susceptibility
Viral transmission, antiviral drugs and vaccines exploration
Model:
Do not reflect human pharmacology
Fast metabolism (no metabolized drugs test)
Cannot be used for long-term pathogenesis due to rapid metabolism
COVID-19:
No severe symptoms
Mild weight loss only making hard to test therapies
[162,163,164]
Roborovski dwarf hamster
Develop rapid and consistent on-set of fulminant clinical disease ().
Highly susceptible COVID-19 model, having stable and fulminant clinical signs similar to changes described in patients who died from SARS-CoV-2 infection. Such a severe form of COVID-19 has not been reproduced in any experimentally infected animal, positioning this model as a valuable model for screening therapeutics and vaccine candidates in highly susceptible individuals.
COVID-19:
ACE2 expression similar to human
Susceptible to SARS-CoV-2 infection
Absence-to-lethal symptoms: weight loss, fall in body temperature, severe acute diffuse alveolar damage, hyaline microthrombi, severe fulminant pneumonia
Viral transmission, antiviral drugs and vaccines exploration
Relevant central nervous system infection is not observed
Model:
Do not reflect human pharmacology
Fast metabolism (no metabolized drugs test)
COVID-19:
Lethality due to fatal lung pathology
[165]
Ferrets
Effectively transmit the virus by direct or indirect contact
Experience mild symptoms of COVID-19 patients
Model:
Commonly used small animal model
Similarities to human respiratory tract
Comparable immune system to human
COVID-19:
High susceptibility to SARS-CoV-2 infection
ACE2 sequence similar to human
Absence-to-moderate symptoms: wheezing, lethargy, sneezing, high temperature
Disease transmission, antiviral drugs, immunotherapies, and vaccines exploration
Model:
Expensive small model due to handling procedures and needs
Long-living model
May not reflect human pharmacokinetics
COVID-19:
Low viral titer in lungs
Not the same severity compared to human
[146,166,167,168,169]
Large animalsCynomolgus macaques
Effective virus transmission to other animals
Development of lung disease
Early peak of virus resembles asymptomatic patients
Useful for the evaluation of vaccines, immunotherapies, and antiviral drugs
Model:
Appreciable size
Good longevity
Similar pharmacokinetics to humans
Innate and adaptive immunity, and physiology exploration
COVID-19:
Similar infection to human
ACE2 sequence similar to human
Vaccines, immunotherapies, and antiviral drugs exploration
Model:
Difficult to handle
Ethical considerations
Small population size
COVID-19:
Limited clinical signs developed
[3,170,171,172]
Rhesus macaques
Exhibits high ACE2-spike activity and is susceptible to SARS-CoV-2 infection
SARS-CoV-2 infection induced protective immunity against subsequent reinfection
DNA vaccine encoding full-length S protein protected them from SARS-CoV-2 infection ChAdOx1 nCoV-19 vaccine prevented SARS-CoV-2 pneumonia
Inactivated SARS-CoV-2 virus vaccine (PiCoVacc) induced SARS-CoV-2-specific neutralizing antibodies
Single dose of Ad26 vector-based vaccine protected against SARS-CoV-2 infection
[173,174,175,176,177,178,179,180]
African green
monkeys
High level of SARS-CoV-2 replication and pronounced respiratory tract infection
Heterologous response along with the ability to collect tissues and longitudinal samples permits a detailed study of pathogenesis and immunity to COVID-19
Developed more severe lung pathology
Young healthy AGMs may represent subclinical or mild human disease
Tissue tropism and transmission studies
[181,182,183]
Marmosets
Quite new model in research field
Not much investigated yet
Relatively resistant to SARS-CoV-2 infection
[184,185,186]
WT: wild type, ACE2: angiotensin-converting enzyme 2.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bestion, E.; Halfon, P.; Mezouar, S.; Mège, J.-L. Cell and Animal Models for SARS-CoV-2 Research. Viruses 2022, 14, 1507. https://doi.org/10.3390/v14071507

AMA Style

Bestion E, Halfon P, Mezouar S, Mège J-L. Cell and Animal Models for SARS-CoV-2 Research. Viruses. 2022; 14(7):1507. https://doi.org/10.3390/v14071507

Chicago/Turabian Style

Bestion, Eloïne, Philippe Halfon, Soraya Mezouar, and Jean-Louis Mège. 2022. "Cell and Animal Models for SARS-CoV-2 Research" Viruses 14, no. 7: 1507. https://doi.org/10.3390/v14071507

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop