Skip to main content
Erschienen in: Critical Care 1/2020

Open Access 01.12.2020 | COVID-19 | Research

Evidence of a wide gap between COVID-19 in humans and animal models: a systematic review

verfasst von: Salleh N. Ehaideb, Mashan L. Abdullah, Bisher Abuyassin, Abderrezak Bouchama

Erschienen in: Critical Care | Ausgabe 1/2020

Abstract

Background

Animal models of COVID-19 have been rapidly reported after the start of the pandemic. We aimed to assess whether the newly created models reproduce the full spectrum of human COVID-19.

Methods

We searched the MEDLINE, as well as BioRxiv and MedRxiv preprint servers for original research published in English from January 1 to May 20, 2020. We used the search terms (COVID-19) OR (SARS-CoV-2) AND (animal models), (hamsters), (nonhuman primates), (macaques), (rodent), (mice), (rats), (ferrets), (rabbits), (cats), and (dogs). Inclusion criteria were the establishment of animal models of COVID-19 as an endpoint. Other inclusion criteria were assessment of prophylaxis, therapies, or vaccines, using animal models of COVID-19.

Result

Thirteen peer-reviewed studies and 14 preprints met the inclusion criteria. The animals used were nonhuman primates (n = 13), mice (n = 7), ferrets (n = 4), hamsters (n = 4), and cats (n = 1). All animals supported high viral replication in the upper and lower respiratory tract associated with mild clinical manifestations, lung pathology, and full recovery. Older animals displayed relatively more severe illness than the younger ones. No animal models developed hypoxemic respiratory failure, multiple organ dysfunction, culminating in death. All species elicited a specific IgG antibodies response to the spike proteins, which were protective against a second exposure. Transient systemic inflammation was observed occasionally in nonhuman primates, hamsters, and mice. Notably, none of the animals unveiled a cytokine storm or coagulopathy.

Conclusions

Most of the animal models of COVID-19 recapitulated mild pattern of human COVID-19 with full recovery phenotype. No severe illness associated with mortality was observed, suggesting a wide gap between COVID-19 in humans and animal models.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13054-020-03304-8.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
COVID-19
Coronavirus disease 2019
SARS-CoV-2
Severe acute respiratory syndrome–coronavirus 2
ARDS
Acute respiratory distress syndrome
MOSD
Multiple organ dysfunction/failure
ACE
Angiotensin-converting enzyme
TMPRSS
Transmembrane serine protease
PRISMA
Preferred Reporting Items for Systematic Reviews and Meta-analysis
hACE2
Chimeric mouse expressing human angiotensin-converting enzyme 2
IFN
Interferon
STAT2
Signal transducer and activator of transcription 2
Ces1c
Serum esterase
RBD
Receptor-binding domain
MERS-CoV
Middle East Respiratory Syndrome

Background

Coronavirus disease 2019 (COVID-19) is a febrile respiratory illness due to a novel viral pathogen severe acute respiratory syndrome–coronavirus 2 (SARS-CoV-2) [1, 2]. COVID-19 can progress to acute respiratory distress syndrome (ARDS), multiple organ dysfunction/failure (MOSD) including central nervous system alteration, acute kidney injury, cardiovascular failure, liver injury, and coagulopathy culminating in death [29].
SARS-CoV-2 is a beta coronavirus that binds with a high affinity to angiotensin-converting enzyme (ACE) 2 receptor and uses the transmembrane serine protease (TMPRSS) 2 as co-receptor to gain entry to cells [1012]. ACE2 and TMPRSS2 are co-expressed in many tissues and organs, particularly the nasal epithelial cells and alveolar type II cells of the lungs, which may explain in part the easy transmission from person-to-person, and its dissemination within the body in severe and fatal cases [1118]. Accordingly, SARS-CoV-2-induced COVID-19 has led to a pandemic that overwhelmed the capacity of most national health systems, resulting in a global health crisis [19]. So far, an estimated 11,280 million persons in 188 countries were infected, of which 531,000 died [20].
The clinical spectrum of COVID-19 is complex and has been categorized as mild, severe, and critical, representing 81, 14, and 5% [2, 3]. The mild pattern comprises patients with either no signs and symptoms or fever and radiological evidence of pneumonia [3]. The severe pattern manifests as rapidly progressive hypoxemic pneumonia involving more than half of the lung with a full recovery phenotype [2, 3]. The critical pattern consists of ARDS requiring respiratory assistance and MOSD that result in death in approximately half of the patients [2, 3, 7, 21]. Mortality was associated with host factors such as old age, comorbidities, and immune response [4].
Viral and immunopathological studies revealed distinct patterns between mild and severe or critical forms of COVID-19 [4, 5, 9, 2127]. Both severe and critically ill patients displayed higher viral load in the upper respiratory tract than mild cases, together with delayed clearance overtime [21, 22]. Likewise, they presented with lymphopenia due to a decrease in CD4+ and CD8+ T cells, as well as T cell exhaustion accompanied by a marked inflammatory response [5, 9, 2427]. Pro- and anti-inflammatory cytokines and chemokine concentrations were increased systemically and locally in the lung and correlated with severity [5, 9, 24]. In contrast, in the mild illness, the lymphocyte count was normal, with no or minimal inflammatory response [5, 23]. Together, these suggest that the viral load and dynamic together with the host inflammatory response may play a pathogenic role.
Clinical and post-mortem studies of fatal cases of COVID-19 demonstrated major alteration of coagulation and fibrinolysis [17, 18]. This was associated with widespread thrombosis of small and large vessels, particularly of the pulmonary circulation contributing to death in a third of patients [8, 2833]. These observations suggest that dysregulated coagulation may be an important mechanism of COVID-19 morbidity and mortality [34].
In this context, animal models appear crucial to a better understanding of the complex biology of COVID-19. Animal models of SARS-CoV-2-induced COVID-19 have been rapidly reported since the start of the pandemic [35]. However, whether they express the full phenotype of COVID-19, particularly the severe and critical patterns associated with lethality, remains to be determined. In this systematic review, we examined whether the newly created animal models reproduce the phenotype of human COVID-19. Moreover, we examined the knowledge generated by these models of COVID-19 including viral dynamic and transmission, pathogenesis, and testing of therapy and vaccines.

Methods

Search strategy and selection criteria

We conducted a systematic review according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) statement [36] to identify studies describing the creation of an animal model of COVID-19 as an endpoint (Table 1 and Additional file 1). Additional file 1 shows the data extraction and appraisal approach as well as the selected outcome.
Table 1
Search strategy and selection criteria
We searched the MEDLINE, as well as BioRxiv and MedRxiv preprint servers for original research describing or using an animal model of SARS-CoV-2 induced COVID published in English from January 1, 2020, to May 20, 2020. We used the search terms (COVID-19) OR (SARS-CoV-2) AND, (animal models), (hamsters), (nonhuman primates), (macaques), (rodent), (mice), (rats), (ferrets), (rabbits), (cats), and (dogs). The preprint servers were included in the search as the field of COVID-19 is developing quickly. Inclusion criteria were the establishment of animal models of COVID-19 as an endpoint. Other inclusion criteria were assessment of prophylaxis, therapies, or vaccines, using animal models of COVID-19. Exclusion criteria consisted of reviews, non-original articles, and unrelated to the COVID-19 infection or experimental animals that do not support SARS-CoV-2 replication. 101 studies and 326 preprints were screened of which 13 peer-reviewed studies and 14 preprints were included in the final analysis (Fig. 1). The variables extracted were the population type, study aim, the virus strain used, clinical response, pathology, viral replication, and host response as well as the effects of prophylaxis, drugs, or vaccines. The outcomes were organized according to species and categorized into phenotype (signs or symptoms; histopathology, time-course of the illness and outcome), viral (titer in each tissue organ, detection methods, duration of positivity), and host response (dynamic of seroconversion, inflammatory, and hemostatic markers), therapy, and vaccine (efficacy and safety)

Results

The systematic search identified 101 studies and 326 preprints, of which 400 articles were excluded because they were reviews, non-original articles, unrelated to the COVID-19 infection, or experimental animals that do not support SARS-CoV-2 replication such as pigs, ducks, and chickens (Fig. 1 and Additional file 2). Additional file 2 displays all the excluded studies and the rationale for their exclusion. Thirteen peer-reviewed studies and 14 preprints were included in the analysis.
The studies used nonhuman primates (n = 13) [3749], mice (n = 7) [5056], hamsters (n = 4) [5659], ferrets (n = 4) [6063], cats, and dogs (n = 1) [63] (Tables 234, and 5). Male and female, as well as young and old, were included but with no associated comorbidities. The aims were to investigate the pathogenesis of COVID-19 (n = 15), testing drugs and vaccines (n = 14), the host immune response (n = 6), and the virus dynamic and transmission (n = 4) (Tables 234, and 5).
Table 2
Summary of studies using nonhuman primate models of COVID-19
Species (ref)
Number age (gender)
Virus strain dose* (inoculation route)†
Clinical signs & observation duration (DPI) §
Viral replication‡ (DPI)
Pathology & sacrificing date (DPI)
Immune response
Seroconversion (DPI)
Outcome measures
Rhesus macaques
n = 8
SARS-CoV-2 nCoV-WA1–2020
Fever
Nose, oropharynx, lung
Anemia
At 1 dpi only, significant increases in IL1ra, IL6, IL10, IL15, MCP-1, MIP-1b
IgG antibody anti-spike protein (10)
Pathogenesis of COVID-19
Adults
Weight loss
Rectum (1)
Mild to moderate, interstitial pneumonia,
Munster et al. (2020) [37]
(M/F)
4 ×  105 TCID50 (IT, IN, PO)
Dyspnea Tachypnea
Edema
Piloerection
Hyaline membranes formation
At 3 dpi decrease in TGFα
Reduced appetite
Hyperplasia type II pneumocytes
Hunched posture
Swollen mediastinal lymph nodes (3, 4, 21)
Pale appearance
Dehydration (21)
Rhesus macaques
n = 3
BetaCoV/Wuhan/IVDC-HB-01/2020
Weight loss
Nose, oropharynx, lung
Interstitial pneumonia
Decreased CD4+ T and CD8+ T cells in young and old.
IgG antibody anti- SARS-CoV-2 (14)
Pathogenesis of COVID-19 in aging animals
3–5 years
 
Asthenia
Rectum, alveolar epithelia
Inflammation
Yu et al. (2020) [38]
n = 2
1 ×  106 TCID50 (IT)
More severe in old than young (14)
Macrophages (3)
Edema
15 years (NA)‖
  
Higher replication in old than young
More severe in old than young (7)
Rhesus macaques
n = 4 per group (6 vaccinated groups)
DNA vaccine**
NA‖ (14)
Lowest BAL levels of viral RNA with full-length S protein encoding vaccine
NA
Upregulation IFN-γ antipeptide spike proteins.
IgG antibody anti- SARS-CoV-2 (day14 post-vaccination)
Evaluation of candidate DNA vaccine
IM at week 0 and week 3
Yu et al. (2020) [39]
6–12 years (M/F)
1.1 × 104 PFU (IN and IT) (day 21 post-vaccine)
   
S1 and RBD lower response than other variant Spike proteins**
n = 10 sham control
1.1 × 104 PFU (IN and IT)
 
High BAL levels of viral RNA
NA
Anamnestic humoral and cellular immune responses including IFN-γ ELISPOT responses
NA
6–12 years (M/F)
Rhesus macaques
n = 6 vaccine
2.5 × 1010 ChAdOx1 nCoV-19 (IM)
Tachypnea (3/6), dyspnea (2/6),
Nose, BAL (2/6)
NO
Upregulation of IFN-γ (1)
IgG antibody anti-SARS-CoV-2 spike protein (day 14 post-vaccination)
Evaluation of DNA vaccine
M/F
SARS-CoV-2 nCoV-WA1-2020
Ruffled fur (1/6) (7)
Lung (very low), oropharynx, mediastinal, duodenum (3)
  
2.6 × 106 TCID50 (IT, IN, PO, CJ) (day 28 post-vaccine)
 
No BAL subgenomic viral RNA
 
No difference in TNF-α, IL-2, IL-4, IL-6, and IL-10 vaccine vs. control
Van Doremalen et al. (2020) [40]
n = 3 control`
Vaccinated with 2.5 × 1010 ChAdOx1 GFP (IM)
Tachypnea (3/3) Ruffled fur (2/3) Diarrhea (1/3) Pale appearance (1/3)
BAL, nasal swabs, lung, cervical, mediastinal lymph nodes, duodenum, urinary bladder
Interstitial pneumonia (2 of 3)
TNF-α, IL-2, IL-4, IL-6, and IL-10
NA
Thickening of alveolar septae
M/F`
 
Red nose (1/3)
 
Edema
 
SARS-CoV-2 nCoV-WA1-2020
 
BAL subgenomic viral RNA (3, 5)
Hyperplasia type I & II pneumocytes syncytial cells
2.6 × 106 TCID50 (IT, IN, PO, CJ) (day 28 post-vaccine)
No extra pulmonary injury
Rhesus macaques
n = 4 per vaccine group
PiCoVacc 6 μg/dose (high) or 3 μg/dose (low) at 0, 7, and 14 days (IM)
NA
Pharyngeal, anal, and pulmonary (3)
Mild and focal histopathological changes both lower lobes
No differences CD3+, CD4+, CD8+, TNF-α, IFN-γ, IL-2, IL-4, IL-5, IL-6 vaccine vs. control
IgG antibody anti-SARS-CoV-2 (day 14 post-vaccination)
Evaluation of an inactivated vaccine
3–4 years (M/F)
SARS-CoV-2-2/human/CHN/CN1/2020
1 × 106 TCID50 (IT) (day 22 post-vaccine)
Gao et al. (2020) [41]
n = 4 control
Vaccinated with Al(OH)3 adjuvant (sham) or physiological saline (control) at 0, 7, and 14 days
NA (7)
Oropharynx, crissum, lung, rectum (3)
Severe interstitial pneumonia
CD3+, CD4+, CD8+, TNF-α, IFN-γ, IL-2, IL-4, IL-5, IL-6
 
3–4 years (M/F)
IM
SARS-CoV-2-2/human/CHN/CN1/2020
1 × 106 TCID50 (IT)
(22 days post-vaccine)
Rhesus macaques
n = 6
SARS-CoV-2 nCoV-WA1-2020
Dyspnea (1/6) (7)
Nose, oropharynx, lung (1)
Minimal interstitial pneumonia subpleural spaces (3/6) (7) (7)
NA
NA
Testing of antiviral therapy
Remdesivir
Low BAL titers (1)
(M/F)
2.6 × 106 TCID50 (IT, IN, OC, PO)
 
No virus in BAL (3)
No extra pulmonary injury
Williamson, B.N. et al. (2020) [42]
n = 6 control
Vehicle solution
Tachypnea, dyspnea
Nose, oropharynx, lung, and BAL (1)
Multifocal, mild to moderate, interstitial pneumonia (7)
NA
NA
(M/F)
SARS-CoV-2 CoV-WA1–2020
  
No extra pulmonary injury
2.6 × 106 TCID50 (IT, IN, OC, PO)
Rhesus macaques
n = 9
SARS-CoV-2 USA-WA1/2020
Reduced appetite
(35)
Nose, pharynx, trachea, lung, gastrointestinal tract, liver, kidney, pneumocytes I & II, ciliated bronchial epithelial cells (1)
Acute interstitial pneumonia
Neutropenia
IgG anti- SARS-CoV-2 Spike protein (35)
Immune protection after a second exposure
Consolidation
Lymphopenia (mild and transitory in high dose group)
6–12 years
initial inoculation
  
Edema
IFN-γ upregulation
1.1 × 106, n = 3
Multiple Inflammatory foci
(M/F)
1.1 × 105, n = 3
Hyaline membranes
1.1 × 104 PFU, n = 3
Damage to type I and type II pneumocytes
(IN, IT)
Necrotic bronchiolar epithelium
Bronchiolar epithelial syncytial cells
No extra pulmonary injury
Chandrashekar et al. (2020) [43]c
n = 9
SARS-CoV-2 USA-WA1/2020
No (14)
5 log10 reduction BAL & nasal viral loads (1)
NA
Increased virus-specific Nab titers
 
 
Second inoculation
 
day 35 post-initial infection
6–12 years
1.1 × 106
1.1 × 105
(M/F)
1.1 × 104 PFU (IN, IT)
Rhesus macaques
n = 7
SARS-CoV-2
Fever
Nose, oropharynx, lung, gut, spinal cord, bladder, rectum (3)
Thickened alveolar septa
Increase CD4+ T cells
IgG antibody anti-SARS-CoV-2 (14)
Immune protection after a second exposure
WH-09/hum/2020
Weight loss
Macrophages accumulation in alveoli Degeneration alveolar epithelia
3–5 years
 
Posture change
 
Inflammatory infiltrates (5, 7)
 
Initial inoculation
Rapid breathing
(NA)
1 × 106 TCID50 (IT)
Reduced appetite (28)
Bao et al. (2020) [44]
(n = 4)
SARS-CoV-2
Transient temperature increase
(14)
Negative
No pathology (5)
CD4+ T higher at 7 day post-exposure vs. post-initial exposure
Higher IgG antibody anti-SARS-CoV-2 (14) vs. initial exposure
WH-09/hum/2020
3–5 years
 
(NA)
second inoculation day 28 post-initial infection
1 × 106 TCID50 (IT)
Rhesus macaques
(n = 5)
SARS-CoV-2 WH-09/hum/2020
weight loss (IT route) (21)
Nasal, oropharynx, rectum (IG route)
Interstitial pneumonia (IT route)
NA
IgG anti-SARS-CoV-2 on 21 dpi (CJ route)
Viral infection routes
Conjunctival (CJ route)
Mild interstitial pneumonia (CJ route)
Deng, W. et al. (2020) [45]
3–5 years (M)
1 × 106 TCID50 (IT, CJ, IG)
Lung, ileum, caecum (IT) (1)
No pneumonia (IG route) (7)
Rhesus macaques
n = 4 young
SARS-CoV-2 CDC, Guangdong, China
Fever
Nose, oropharynx, trachea
Inflammatory cell infiltrates
Peak CD4+ T cells, CD8+ T cells, and monocytes (2)
IgG antibody anti-SARS-CoV-2 (4)
Pathogenesis of COVID-19 in different species of nonhuman primates
n = 6 adult
Weight loss (21)
Bronchus, lung, rectum
Diffuse hemorrhage and necrosis
n = 4 old
Blood, spleen (2)
Swollen lymph nodes (hilar, mediastinal, mesenteric)
 
4.75 × 106 PFU (IT, IN, CJ)
  
Pericardial effusion
Young stronger B cell responses vs. adults vs. old
IgG levels lower in young vs. adult vs. old
(NA)
(50% given to young)
  
Mild hepatic steatosis
Increased G-CSF, IL-1A, IL-8, IL-15, IL-18, MCP-1, MIP-1B, sCD40-L
 
splenic hemorrhage (4, 7, 12, 13, 15)
Common Marmoset
n = 6
SARS-CoV-2
None
Nose, oropharynx, rectum
Broken pulmonary septum
NA
No
CDC, Guangdong
 
Blood (2)
Inflammatory infiltrates
Age = NA (M/F)
1 × 106 PFU (IN)
  
Splenic hemorrhage
Swollen hepatocytes
Renal inflammatory infiltrate
Cynomolgus macaques
n = 6
SARS-CoV-2 CDC, Guangdong
Fever
Nose, oropharynx, trachea
Inflammatory cell infiltrates
CD4+ T cells, CD8+ T cells, and monocytes (2)
IgG antibody anti-SARS-CoV-2 (4)
Weight loss
Bronchus, lung, rectum
Diffuse hemorrhage and necrosis
Adult
4.75 × 106 PFU (IT, IN, PO)
 
Blood, spleen (2)
Swollen lymph nodes (hilar, mediastinal)
Young stronger B cell responses vs. adults vs. old
Hepatic steatosis
Lu et al. (2020) [46]
(M/F)
   
Splenic hemorrhage
Increased G-CSF, IL-1A, IL-8, IL-15, IL-18, MCP-1, MIP-1B, sCD40-L
Cynomolgus macaques
n = 4
SARS-Cov-2 BetaCoV/Munich/BavPat1/2020
serous nasal discharge (1/4 old monkey) (21)
Nose, oropharynx, lung
Foci pulmonary consolidation
NA
IgG antibody anti-SARS-CoV-2 (14)
Comparisons of pathogenesis between COVID-19, SARS-CoV and MERS-CoV
Pneumocytes I & II
Diffuse alveolar damage
4–5 y (F)
  
Ciliated nasal, bronchial & bronchiolar epithelial cells
Hyaline membrane
15–20 years (F)
2 × 105 TCID50 (IT, IN)
  
Multinucleated giant cells
 
Type I & II pneumocytes hyperplasia
Earlier detection in young (2) vs. old (4).
Alveolar edema
 
Leukocyte infiltration
Higher nasal replication in old vs. young
(4)
Rockx et al. (2020) [48]
n = 10
MERS-CoV
No
Nose, oropharynx, lung
Foci pulmonary consolidation
 
IgG antibody anti-MERS-CoV (21)
EMC strain, accession no. NC_019843
 
Pneumocytes II
Alveolar edema
3–5 years
106 TCID50
 
& rectal swabs (2)
Leukocyte infiltration
Type II pneumocytes hyperplasia
F
(IT, IN)
   
NA
NA
No
Nose, oropharynx, lung
Type I & II pneumocytes hyperplasia
NA
NA
Pneumocytes I & II
Alveolar edema (aged only)
Leukocyte infiltration
Hyaline membrane (aged only)
Cynomolgus macaques
n = 6
2019-nCoV/USA-WA1-A12/2020
None (30)
Nose, eye, oropharynx, rectum (2)
CT scan: Ground glass appearance
Increased CXCL8, IL6, IL13, IL15, IL1RN, and TNF (6) in one macaque.
IgG antibody anti-SARS-CoV-2 spike S1 subunit (10)
Evaluation of medical interventions
Reticulonodular opacities
Finch et al. (2020) [47]
4–4.5 years (M/F)
3.65 × 106 PFU (IT, IN)
  
Peri-bronchial thickening
Subpleural nodules
Alveolar dense consolidation (n = 1)
PET scan: FDG uptake lung and regional lymph nodes (2), mediastinal lymph nodes and spleen (6)
African green monkey
n = 6
SARS-CoV-2-2/INMI1-/2020/Italy
Reduced appetite
Nasal, oropharynx, lung, rectum, pneumocytes I & II, alveolar macrophages (2)
Interstitial pneumonia
Increased CRP ¶ (n = 2)
IgG antibody against SARS-CoV-2 b (5)
Pathogenesis of COVID-19
Bronchiolitis
Fever (31)
Woolsey et al. (2020) [49]
NA
5 × 105 PFU (IT, IN)
  
Edema
IL-8, IP-10, IL-12, IL-6, IFN-beta, IL10, and MCP-1 (2)
Hemorrhage
Hyaline membrane
Hyperplasia type II pneumocytes
Distention and flaccidity small intestines segments
(5)
*TCID50 Median Tissue Culture Infectious Dose at which 50% of the cells are infected, PFU plaque-forming unit, †IT intratracheal, IN intranasal, CJ intraconjunctival, OC ocular, IG intragastric, PO per oral. ‡ Viral replication: RNA copies (PCR), viral antigen (immunostaining), viral particles (electron microscopy). § dpi day post-inoculation, ¶ CRP C-reactive protein, || NA Not available. **Vaccine encoding spike protein variants: Full-length SARS-CoV-2 S protein, S.dCT Deletion of the cytoplasmic tail of SARS-CoV-2 S protein, S.dTM deletion of the transmembrane domain and cytoplasmic tail reflecting the soluble ectodomain, S1 S1 domain with a fold on trimerization tag, RBD Receptor-binding domain with a fold on trimerization tag, S.dTM.PP a prefusion stabilized soluble ectodomain with deletion of the furin cleavage site, two proline mutations, and a fold on trimerization tag, IM Intramuscular
Table 3
Summary of studies using mice models of COVID-19
Species (ref)
Number age (gender)
Virus strain dose* (inoculation route)†
Clinical signs & observation duration (DPI) §
Viral replication‡ (DPI)
Pathology & sacrificing date (DPI)
Immune response
Seroconversion
(DPI)
Outcome Measures
Mice
WT-BALB/c, n = 3
2 × 105 TCID50 of P 4†† or 2 × 106 of P 6†† (IN)
NA
Lung (3)
Mild lung pathology (2)
Mild inflammatory response
NA
Interferon response to SARS-CoV-2 infection
BALB/c:
SCID, n = 3
 
(14)
No difference in viral load WT vs. SCID
No difference in lung pathology WT vs. SCID
(2, 4, 7, 14)
WT¶
 
SCID||
6–8 weeks (F)
C57BL/6:
C57BL/6 n = 5
2 × 105 TCID50 of P 4 or 2 × 106 of P 6 (IN)
NA
(14)
Lung (3)
Greater intra-alveolar hemorrhage and peribronchiolar inflammation in IFNar1−/− mice than WT and IL28r−/− mice (3)
Higher inflammatory response in IFNar1−/− vs. WT and IL28r−/− mice
NA
WT
IFNar1−/−¶¶ n = 14
Ifnar1−/−
 
Higher viral replication in IFNar1−/− mice vs. WT and IL28r−/− mice
(2, 4, 7, 14)
Il28r−/−
6–8 weeks (F)
Boudewijns et al. (2020) [56]
C57BL/6, n = 5
2 × 105 TCID50 of P 4 or 2 × 106 of P 6 (IN)
NA
(14)
Lung (3)
Mild lung pathology (3)
Mild inflammatory response
NA
IL28r−/−, n = 5
No difference in viral load between WT and IL28r−/−
(2, 4, 7, 14)
6–8 weeks (F)
Mice
hACE2 mice
SARS-CoV-2 (BetaCoV/Wuhan/IVDC-HB-01/2020|EPI_ISL_402119)
Slight
Highest viral load
Moderate interstitial pneumonia
MAC2, CD3+ T and CD19+ B cells in alveolar septum
IgG antibody response against SARS-CoV-2 (21)
Pathogenesis of COVID-19
hACE2‖‖ transgenic mice
(ACE2-HB-01)
Bristles
In lung (3)
Thickened alveolar septa
Weight loss
Intestine (1)
n = 19
105 TCID50 (IN)
Arched back
(14)
Alveolar macrophage, and alveolar epithelia (3)
Lymphocytes, macrophages, and monocytes infiltrates in the interstitial and alveolar space
6–11 months (M/F)
Bronchioles degeneration (3)
No pathology in intestine, spleen, heart, liver, kidney, brain, and testis
(1, 3, 5, 7)
Bao et al. (2020) [50]
WT-HB-01 (n = 15)
SARS-CoV-2 (BetaCoV/Wuhan/IVDC-HB-01/2020|EPI_ISL_402119)
105 TCID50 (IN)
No
(14)
No viral RNA detectable in lung or intestine (1)
No
(1, 3, 5, 7)
No
No
6–11 months (M/F)
Mock-treated hACE2 mice (n = 15)
PBS 50 μl
(IN)
No
(14)
No viral RNA detectable in lung or intestine
No
(1, 3, 5, 7)
No
No
6–11 months (M/F)
Mice
n = 5
6–8 weeks (M)
SARS-CoV-2 (BetaCoV/Hong Kong/VM20001061/2020 [KH1])
NA
NA
NA
NA
IgG antibody response against SARS-CoV and SARS-CoV-2 spike protein and RBD
Cross-reactivity of antibodies against SARS-CoV and SARS-CoV-2
Infection
SARS-CoV (HK39849, SCoV)
105 PFU (IN)
BALB/c:
n = 5
Immunization with heat-inactivated plasma from SARS-CoV and SARS-CoV-2 (IP)
NA
NA
NA
NA
Cross-reactive antibody binding responses SARS-CoV-2 and SARS-CoV
No cross-neutralization SARS-CoV-2 and SARS-CoV
WT
6–8 weeks (M)
Lv et al. (2020) [53]
Immunization
 
n = 6
Vehicle (IN)
NA
NA
NA
NA
 
6–8 weeks (M)
control
Transgenic mice
n = 7
SARS1/SARS2-RdRp §§
Improvement of pulmonary function
reduced lung viral load 102 PFU/lobe (5)
Decreased lung hemorrhage (5)
(5)
NA
NA
Antiviral therapy testing
C57BL/6***:
17 weeks (F)
103 PFU (IN)
(5)
Ces1c−/−
Remdesivir given at 1dpi
Pruijssers et al. (2020) [55]
Remdesivir
n = 7
SARS1/SARS2-RdRp
Reduced pulmonary function by WPH†††
(5)
Lung viral load
Lung hemorrhage (5)
(5)
NA
NA
17 weeks (F)
103 PFU (IN)
105 PFU/lobe (5)
Control
Vehicle
 
n = 3
Mouse-adapted SARS-CoV-2 (BetaCoV/Wuhan/AMMS01/2020)
Weight loss old mice (5)
(7)
Trachea, lung, heart, liver, and intestine, pneumocytes Type II
Thickened alveolar septa
Increased TNF-α, IL-1β, IL-6, and IL-5, MCP-1, G-CSF, and GM-CSF (3)
NA
Establishment of mouse-adapted SARSCoV-2 model of COVID19
Young, 6 weeks (F)
Alveolar damage and focal exudation
Hemorrhage,
n = 3
7.2 × 105 PFU (IN)
 
Viral replication similar in old vs. young cells (3)
Inflammatory cell infiltration
Higher and sustained cytokines levels in aged mice vs. young
Denaturation of endothelial tissues (3)
Mice
Old, 9 months (F)
 
Lung pathology similar in old vs. young mice
(3, 5, 7)
BALB/c:
WT
Control mice
NA
No weight loss
No viral protein
NO
NO
 
Evaluation of candidates vaccine
Gu et al. (2020) [54]
NA
n = 10
Immunization day 1, 14
NA
No viral replication detectable in lungs (5)
No
NA
Higher IgG antibody response against SARS-CoV-2 (14)
6–8 weeks
Challenged with mouse adapted SARS-CoV-2
Immunized with SARS-CoV-2 RBD-Fc protein
(IN), 4 weeks after second immunization
PBS control with aluminum adjuvant
  
High viral load in the trachea and lungs (5)
Focal perivascular and peribronchiolar inflammation
Thickened alveolar septa
NA
NA
Mice
n = 5
SARS-CoV-2
No
(2)
No viral replication detectable in lung (2)
NA
NA
IgG1 ab1 protects hACE2 transgenic mice from SARS-CoV-2 infection.
(2)
Evaluation of prophylaxis with monoclonal antibody
6–9 weeks (F)
105 PFU (IN)
C3B6: hACE2 mice
 
Immunization
Human monoclonal IgG1 antibody (12 h)
C3B6:
 
Prior the virus challenges (IP)
hACE2 transgenic mice
n = 6
SARS-CoV-2
No
(2)
Viral replication 103 PFU per lung (2)
NA
NA
No
6–9 weeks (F)
105 PFU (IN)
C3B6: hACE2 mice
 
BALB/c mice
Control
IgG1 m336 (no activity in vitro)
Li et al. (2020) [51]
Balb/c, n = 5
Mouse ACE2 adapted SARS-CoV-2‡‡
No
(2)
No viral replication detectable in lung lobe at different dosages (2)
NA
NA
IgG1 ab1 protected mice SARS-CoV-2 challenge
(2)
10–12 months (F)
105 PFU (IN)
Human monoclonal IgG1 ab1 antibody (12 h)
Prior the virus challenges (IP)
Mice
hACE2 mice
SARS-CoV-2
Weight loss
Lung (2), brain (5)
NA
(2, 5)
NA
NA
Evaluation of vaccine and therapy in mouse-adapted SARS-CoV-2 model
BALB/c:
And
hACE2
NA
105 PFU (IN)
Mortality 40% (5)
(5)
Transgenic mice
BALB/c mice
SARS-CoV-2MA§
Pulmonary obstruction (WBP)†††
Upper airway
 
NA
NA
Dinnon et al. (2020) [52]
n = 33
105 PFU (IN)
 
Lung (2,4)
Greater lung inflammation and hemorrhage in old vs. young mice (2,4)
Young 12 weeks
BALB/c mice
 
Greater
Higher replication in old vs. young mice
n = 34
 
Weight and pulmonary
12 months
 
Function loss in old vs. young mice
Vaccination
SARS-CoV-2 spike (S) or nucleocapsid (N)
NA
Vaccine with spike S reduced lung and nasal turbinate titer (2)
NA
NA
NA
n = 8–10
10 weeks
Challenged 4 weeks post-inoculation with SARS-CoV-2 MA
BALB/c
105 PFU (IN)
Prophylaxis
Subcutaneous administration interferon (IFN) lambda-1a 2 μg
 
Reduced SARS-CoV-2 MA replication in the lung (2)
NA
NA
NA
Therapy
18 h prior or 12 h after
BALB/c
SARS-CoV-2 MA
12 weeks
105 PFU (IN)
n = NA
 
*TCID50 Median Tissue Culture Infectious Dose at which 50% of the cells are infected, PFU plaque-forming unit, † IN intranasal, IP intraperitoneal. ‡ Viral replication: RNA copies (PCR), viral antigen (immunostaining), viral particles (electron microscopy). § dpi day post-inoculation. ¶ WT wild type, || SCID severe combined immunodeficiency (lacking functional T and B cells). ** SARS-CoV-2MA A recombinant mouse ACE2 adapted SARS-CoV-2 variant remodeled by introduction of two amino acid changes at the ACE2 binding pocket in the receptor-binding domain to facilitate efficient binding to mouse ACE2. †† P4 and P6: Number of serial passaging of patient SARS-CoV-2 on HuH7 and Vero-E6 cells. ‡‡ Remodeling of the SARS-CoV-2 spike protein in the receptor-binding domain to facilitate efficient binding to mouse ACE2. §§ Chimeric mouse-adapted SARS-CoV1 MA15 variant encoding the SARS-CoV2 RNA-dependent RNA polymerase (“SARS1/SARS2-RdRp”). ¶¶ Genetic ablation of type I (Ifnar1−/−), III interferon (IFN) receptors (Il28r−/−), and Signal transducer and activator of transcription 2 (STAT2−/−). ‖‖ hACE2 chimera expressing human ACE2 receptor. *** C57BL/6 Mice Ces1c−/−: lack a serum esterase, an enzyme that is not present in humans, that reduces markedly the Remdesivir half-life. ††† WPH whole-body plethysmography
Table 4
Summary of studies using hamsters models of COVID-19
Species
(ref)
Number
age (gender)
Virus strain
dose*
(inoculation route)†
Clinical signs & observation duration
(DPI) §
Viral replication‡
(DPI)
Pathology & sacrificing date
(DPI)
Immune response
Seroconversion
(DPI)
Outcome measures
Syrian hamsters
Chan et al. (2020) [57]
n = 8
SARS-CoV-2 Hong Kong
Tachypnea
Nose, trachea, lung
Diffuse alveolar damage (exudative)
Upregulation of Interferon-γ and proinflammatory chemokine, cytokine genes expression
IgG antibody response against SARS-CoV-2
(7)
Viral transmission and immunoprophylaxis
Weight loss
Intestine (high viral load¶) (2–7)
6–10 weeks (M/F)
(Donor)
105 PFU (IN)
Lethargy
 
Apoptosis
  
Ruffled furs
Blood (low viral load)
Diffuse alveolar damage (proliferative)
Early convalescent serum Immunoprophylaxis decreased nasal and lung viral load but not lung pathology or clinical signs
Hunched back posture
(14)
Tissue repair
Intestinal villi damage and necrosis
Reduced spleen size (2–14)
(2, 4, 7, 14)
n = 8
Direct contact with donor
Less weight loss than inoculated animals
(14)
No difference in viral load inoculated animals vs. infected animals via contact (4)
No difference inoculated vs. infected by contact
(2, 4, 7, 14)
NA
IgG antibody response against SARS-CoV-2
(7)
6–10 weeks (M/F)
Inoculated with 100 ul of PBS
Syrian hamsters
n = 6 (mAb CC12.1 or CC12.23)
SARS-CoV-2 (USAWA1/2020)
Weight loss dose-dependent
(5)
Reduced lung viral load
NA
(5)
NA
Neutralizing antibody
(5)
Immunoprophylaxis and therapy
1X106 PFU (IN)
12 h post-Ab infusion
Rogers et al. (2020) [58]
n = 6
Control IgG1 (Den3)
SARS-CoV-2 (USAWA1/2020)
Weight loss
(5)
No difference in lung viral loads control vs. low dose groups
NA
(5)
NA
NA
1 × 106 PFU (IN)
12 h post-Ab infusion
Golden Syrian hamsters
(n = 9))
4–5 weeks (M)
(Donor)
SARS-CoV-2 BetaCoV/Hong Kong/VM20001061/2020
Weight loss (6)
Upper respiratory tract, nose, olfactory
Inflammatory infiltrates nasal turbinate Progressive lung consolidation (5 to 60%) Mononuclear cell infiltration.
CD3 positive T lymphocytes in peribronchial region (5)
IgG antibody response against SARS-CoV-2 (14)
Viral transmission
Ruffled hair coat (5)
(14)
Neurons, bronchus, lung
No extrapulmonary pathology
Kidney, duodenum
8 × 104 TCID50 (IN)
  
No pathology in the intestine, spleen, heart, and brain (2, 5, 7)
(2, 5, 7)
Sia et al. (2020) [59]
(n = 9)
Infection via contact with donor hamster
Weight loss (6)
Detectable infectious viruses (9/9)
NA
NA
IgG antibody response against SARS-CoV-2 (14)
4–5 weeks (M)
 
Ruffled hair coat day (4)
(14)
Day 1 post-contact
(Contact)
No difference in viral shedding contact vs. donor
Syrian hamster
(n = 7)
SARS-CoV-2 (BetaCoV/Belgium/GHB03021/2020)
NA
(4)
Lungs, blood, spleen, liver, upper & lower gastrointestinal tract
Multifocal necrotizing bronchiolitis,
Increased inflammation-related gene expression
NA
Host interferon response to SARS-CoV-2
Age: NA (F)
2 × 105 TCID50 (P4 virus) or 2 × 106 TCID50 (P6 virus) (IN)
 
Leukocyte infiltration
 
Wild type
Edema (4)
(2, 3, 4)
No increase in serum levels of IL-6, IL-10, and IFN-γ (4)
Hamster (STAT2−/− and IL28R-a −/−) strains
(n = 7)
Same as wild type
NA
(4)
Greater levels of viral RNA in the lung, spleen, liver, blood, and upper and lower gastrointestinal tract in STAT2−/− hamster vs. WT and IL28ra−/−
Lung pathology and inflammation decreased in (STAT2−/−) but not in IL28R-a−/− hamsters (2,3,4)
(2, 3, 4)
Increased IL-6 and IL-10 expression in lungs
NA
7–12 weeks (F)
No increase in serum levels of IL-6, IL-10, and IFN-γ (4)
STAT2−/−
Boudewijns et al. (2020) [56]
(n = 7)
Same as wild type
NA
(4)
Lungs, blood, spleen, liver, upper, & lower gastrointestinal tract
Bronchopneumonia and peribronchiolar inflammation (2,3,4)
(2, 3, 4)
High (MMP)-9 levels in lung homogenates compare to WT
NA
5–7 weeks (F)
No differences in lung viral RNA levels in WT, vs. STAT2−/− vs. IL28R-a−/− hamsters
Increased IL-6 and IL-
IL28R-a−/−
10 expression in lungs
No increase in serum levels of IL-6, IL-10 and IFNγ (4)
*TCID50 Median Tissue Culture Infectious Dose at which 50% of the cells are infected, PFU plaque-forming unit, † IN intranasal, ‡ viral replication: RNA copies (PCR), and or viral antigen (immunostaining), viral particles (electron microscopy). § dpi day post-inoculation, ¶ mAb CC12.1 IP SARS-CoV-2-2-specific human neutralizing monoclonal antibodies, ‖ IgG1 (Den3) 2 mg of a dengue specific human IgG1
Table 5
Summary of studies using ferrets, cat, and dog models of SARS-CoV-2 infection
Species (ref)
Number age (gender)
Virus strain dose* (inoculation route)†
Clinical signs & observation duration (DPI)§
Viral replication‡ (DPI)
Pathology & sacrificing date (DPI)
Immune response
Seroconversion (DPI)
Outcome measures
Ferrets
n = 12
NMC-nCoV02/Korea
Increased body temperature
Nose, saliva, urine, and feces
Acute bronchiolitis
NA‡
IgG and serum-neutralizing antibody response against SARS-CoV-2 (12)
Pathogenesis of COVID-19
Infiltrates of immune cells and debris (4)
12–20 months
105.5 TCID50
Reduced activity `
 
(4, 8, 12)
Assess viral transmission
M/F
IN
occasional coughs
(12)
Trachea, lung, kidney, serum, and intestine (2)
n = 12
Control
NO
(12)
NO
NO
(4, 8, 12)
NO
NO
12–20 months
 
M/F
 
Kim et al. (2020) [60]
n = 6
(naïve direct contact)
Direct contact
Increased body temperature
(12)
All animals were infected (2)
(12)
 
IgG and serum-neutralizing antibody response against SARS-CoV-2 (12)
n = 6
(naïve indirect contact)
Indirect contacts
No increased body temperature
(12)
Nose, feces (2 out of 6 animals)
(4)
(12)
 
IgG antibody response against SARS-CoV-2 (12)
Serum-neutralizing antibody response in 1 out of 6
Ferrets
Blanco-Melo et al. (2020) [61]
n = 6
USA-WA1/2020
NA
(14)
Nose, trachea
NA
(3, 14)
Reduced interferon type I and III response
NA
Host interferon response
4 months
5 × 104 PFU
(3,7)
 
Castrated male
IN
 
Increase proinflammatory chemokines and cytokines response
Ferrets
Richard et al. (2020) [62]
n = 4
BetaCoV/Munich/BavPat1/2020
NA
(21)
Nose, throat, rectum
NA
(21)
NA
IgG antibody response against SARS-CoV-2 (21)
Viral transmission
6 months (F)
6.105 TCID50
Donor
IN
n = 4
6 h post-inoculation co-housed with donor
NA
(21)
Nose, throat, rectum (1)
NA
(21)
NA
IgG antibody response against SARS-CoV-2 (21)
6 months (F)
direct contact
n = 4
1 dpi placed in an opposite cage (10 cm) of donor
NA
(21)
Nose, throat, rectum (3)
NA
(21)
NA
IgG antibody response against SARS-CoV-2 (21)
6 months (F)
Indirect contact
Ferrets
n = 10
SARS-CoV-2 /F13/environment/2020/Wuhan
Fever
Nose, throat, and rectum (low titer) (4–8)
Severe lymphoplasmacytic, perivasculitis
 
IgG antibody response against SARS-CoV-2 (13)
Pathogenesis of COVID-19
3–4 months
F
 
Loss of appetite
(20)
 
Vasculitis
NA
SARS-CoV2/CTan/human/2020/Wuhan]
  
Increased type II pneumocytes, macrophages, and neutrophils in the alveolar septa and alveolar lumen.
105 PFU
  
Mild peribronchitis (13)
IN
  
(4, 20)
n = 8
SARS-CoV2/CTan/human/2020/Wuhan]
NA
(14)
Nose, throat
(2–8)
NA
(2, 4, 8, 14)
NA
NA
3–4 months
105 PFU
F
IT
Cats
n = 10
SARS-CoV-2 /CTan/human/2020/Wuhan
NA
(20)
Nose, soft palate, tonsil, trachea, lungs, and small intestines.
NA
(3, 6, 10, 20)
NA
IgG antibody response against SARS-CoV-2 (10)
6–9 months
105 PFU
M\F
IN
Sub-adult
n = 10
SARS-CoV-2 /CTan/human/2020/Wuhan
One cat died (3)
(12)
Nose, soft palate, tonsil, trachea, lungs, and small intestines.
Extensive nasal, tonsil, tracheal, lung, and small intestine epithelial mucosal lesions (3)
(3, 6, 11,12)
NA
IgG antibody response against SARS-CoV-2 (10)
70–100 days
105 PFU
M/F
IN
Juvenile
Dogs
n = 5
SARS-CoV-2-2/CTan/human/2020/Wuhan
NA
Rectum (2/5)
NA
NA
IgG antibody response against SARS-CoV-2 (14)
3-month beagles
105 PFU
IN,
Shi et al. (2020) [63]
n = 2
Exposed to donor
NA
No
NA
NA
No
3 month
Beagles
*TCID50 Median Tissue Culture Infectious Dose at which 50% of the cells are infected, PFU plaque-forming unit, †IT intratracheal, IN intranasal, CJ intraconjunctival, OC ocular, IG intragastric, PO per oral, IP intraperitoneal, ‡ viral replication RNA copies (PCR), viral antigen (immunostaining), viral particles (electron microscopy), § dpi day post-inoculation
All the experimental animals were inoculated with SARS-CoV-2 with various strains, doses, and route of administration that differed across studies (Tables 234, and 5). Likewise, the time-point for tissue collection and pathological assessment were variables. These together precluded any comparisons between the animal models either intra-species or inter-species.

Nonhuman primate models

Viral model

Rhesus macaques (n = 10) [3746], cynomolgus (n = 3) [4648], and African Green model (n = 1) [49] and common marmoset (n = 1) [46] were assessed as models for COVID-19 (Table 2). SARS-CoV-2 strains, dose, and route of inoculation were different across studies. Different doses of virus inoculum were compared in a single study and showed that viral load in the upper and lower respiratory tract, fever, weight loss, respiratory distress, and mortality were comparable regardless of the doses except for mild transient neutropenia and lymphopenia in the high dose group [43]. In contrast, the route of administration resulted in different pathological response as the intratracheal route elicited severe interstitial pneumonia, as compared with mild interstitial pneumonia and no pneumonia from the intraconjunctival and intragastric routes, respectively [45]. The animals were euthanized at different time-points post-inoculation ranging from 3 to 33 days.

Phenotype

The animals displayed variable clinical manifestations from none to fever, altered respiratory patterns, and other general signs (Table 2). The clinical manifestations were not different between old and young macaques [4648]. Structural and ultrastructural examination of the respiratory tract were also variables including mild to moderate interstitial pneumonitis, edema, foci of diffuse alveolar damage with occasional hyaline membrane formation, and pneumocytes type II hyperplasia (Table 2). Old rhesus macaques exhibited more diffuse and severe interstitial pneumonia than young ones [47]. The extrapulmonary injury was investigated in five studies [40, 42, 43, 46, 49]. These revealed pathological changes in two studies [46, 49] including distention and flaccidity of the intestine, inflammatory cells infiltrating the jejunum, and colon, steatosis of the liver, and alteration of myocardial fiber architecture with increased mitochondrial density [46, 49]. No mortality was observed in any of the nonhuman primate models.
Comparisons between species of nonhuman primates were not possible except in one study, which suggested that rhesus macaques were superior to cynomolgus and common marmoset as models of human COVID-19 [46]. Other comparisons suggested that SARS-CoV elicited more severe lung pathology than SARS-CoV-2 and Middle East Respiratory Syndrome (MERS-CoV) [48] (Table 2).

Viral and host interaction

The virus replicated rapidly and at higher titers in the upper airway and lung in all four species [3749]. The virus was detected in pneumocytes type I and II and ciliated epithelial cells of nasal, bronchial, and bronchiolar mucosa [3749]. This differs from MERS-CoV where the virus was mainly present in type II pneumocytes [46] (Table 2). Replication of the virus was also demonstrated in jejunum, duodenum, colon, and rectum [37, 38, 4049]. Viral genome was detected in the blood of rhesus macaques, cynomolgus, and marmoset in one study [46]. Viral replication of nasopharyngeal as well as anal swabs, and the lung in old macaques was higher than in young ones [47, 48].
SARS-CoV-2 infection-induced IgG antibodies response against the SARS-CoV-2 spike was noted in all species [37, 46, 48, 49] except in marmoset [46]. The antibodies were protective against a second exposure to the virus [43, 44]. There was no difference between males and females [37, 3943, 46, 47]; however, young rhesus macaques had lower antibody titers than the old macaques [46]. The innate immune response to SARS-CoV-2 infection was variable with normal, high, or low leucocytes and lymphocyte counts [37, 46]. Occasional reduction of CD4+ and CD8+ T cell concentrations was documented [37] as well as the transitory release of various cytokines and chemokines at different days post-inoculation [37, 46, 49].

Drugs and vaccines

DNA and inactivated virus-based vaccines were evaluated and showed protection in these nonhuman primates. However, the DNA vaccine did not reduce the virus presence in the upper airway, while there was a residual small interstitial pneumonitis in the macaques that received the inactivated virus [40, 41]. This suggests that none of the virus tested so far displayed a comprehensive protection against SARS-CoV-2 infection. Several candidate DNA vaccines based in various forms of the SARS-CoV-2 Spike (S) protein were also tested in rhesus macaques [39]. The findings revealed that only the vaccine encoding the full-length (S) offered optimal protection against SARS-CoV-2 [64]. Nonhuman primates served also for the evaluation of antiviral therapies and medical interventions such as CT- and PET-scanners [47].

Mouse models

Viral model

Wild type mice (BALB/c, C57BL/6), immunodeficient mice (SCID), chimeric mouse expressing human angiotensin-converting enzyme 2 (hACE2), and the RNA-dependent RNA polymerase (SARS1/SARS2-RdRp) were evaluated as models of COVID-19 (Table 3). Moreover, knockout (KO) mice were generated to test specific immunological pathways or therapy, including ablation of type I (IFNar1−/−), III interferon (IFN) receptors, (IL28r−/−), signal transducer and activator of transcription 2 (STAT2−/−), and serum esterase (Ces1c−/−).
Patient isolates of SARS-CoV-2 from different sources and variable times of passaging on various cell cultures or BALB/c mice were employed (Table 3). Mouse-adapted SARS-CoV-2 was developed using two methods. The first by serial passaging (up to 6) through the lungs of BALB/c mice until the virus spike receptor-binding domain (RBD) adapted to the murine ACE-2 [54]. In the second, using genetic engineering, the SARS-CoV-2 RBD was remodeled to enhance its binding efficiency to murine ACE2 [52].

Phenotype

The clinical signs and symptoms varied from none to mild weight loss, arched back, and slight bristles. Whole-body plethysmography was used to measure the respiratory function of the animals and showed a mild to moderate reduction in old more than in young (Table 3). Likewise, the pathological changes varied according to the experimental models and included peribronchiolar inflammation, lung edema, moderate multifocal interstitial pneumonia, lymphocyte infiltration, and intra-alveolar hemorrhage. Survival of hACE2 mice was decreased at 5-day post-inoculation and was attributed to high viral replication in the brain, while it was minimal in the lung, suggesting a different pathogenic mechanism of death from human COVID-19 [52]. Wild type mice showed no pathology as compared to hACE2 mice, indicating that the lack of human ACE2 receptor cannot be infected or inefficiently with SARS-CoV-2 [50, 56]. On the other hand, mouse-adapted SARS-CoV-2 exhibited more severe pathology, particularly in the aged mouse than hACE2 transgenic mouse, suggesting that these models may be more relevant for the study of human COVID-19 [52, 54]. However, whether the pathogenesis induced by the mouse-adapted SARS-CoV-2 is translatable to humans warrants further studies [52, 54].

Viral and host interaction

The virus replicated to high titers in the upper and lower respiratory tract in most of the genetically modified mice models but not in wild type. Viral replication was detected outside the respiratory tract in the intestine of hACE2 mice [50] as well as in the liver, and heart in mouse modified SARS-CoV-2 RBD [52]. Increased viral replication in KO mice IFNar1−/− suggested that interferon limited the viral replication [56].
Specific IgG antibodies against SARS-CoV-2 were documented in two studies (Table 3). The IgG antibodies were found to cross-react in their binding to the spike protein of SARS-CoV, however, with no cross-neutralization, hence suggesting the conservation of the same spike protein epitopes among coronaviruses [53]. Proinflammatory cytokines and chemokines were demonstrated in mouse-adapted SARS-CoV-2 and KO mouse (Table 3). The inflammatory response was significantly higher in the old than young mice.

Drugs and vaccines

Antiviral therapies, including remdesivir [55], IFN lambda [52], and human monoclonal IgG1 antibody against RBD [50], were tested in these mouse models and produced a protective effect. Likewise, vaccines using viral particles expressing SARS-2-S protein [52] or an RBD-based vaccine were tested and showed protection [55].

Hamster

Viral model

Wild type Syrian hamsters and knockout hamsters for signal transducer and activator of transcription 2 (STAT2−/− lacking type I and III interferon signaling) and interleukin 28 receptors (IL28r−/− lacking IFN type III signaling) were reported as models for COVID-19. Patient isolate of SARS-CoV-2 from different sources and different passages on various cell cultures was used (Table 4). SARS-CoV-2 was administered intranasally at different titers to anesthetized hamsters. Viral transmission between hamsters was demonstrated either through direct contact or indirectly via airborne transmission.

Phenotype

The clinical manifestations included weight loss, which was consistently observed. Other clinical signs and symptoms such as rapid breathing, lethargy, ruffled furs, and hunched back posture were reported in one study [57]. The histopathological findings were variables according to the experimental models and ranged from lung consolidation to multifocal necrotizing bronchiolitis, leukocyte infiltration, and edema. STAT2−/− hamsters exhibited attenuated lung pathology as compared with IL28R-a−/− hamsters [56].

Viral and host interaction

The virus replicated to high titer in the upper and lower respiratory tract in most of the hamsters’ models. Viral replication was detected in the blood and kidney with a low concentration (Table 4). STAT2−/− hamsters had higher titers of infectious virus in the lung, viremia, and high levels of viral RNA in the spleen, liver, and upper and lower gastrointestinal tract in comparison with wild type and IL28R-a−/− hamsters. Specific IgG antibodies against SARS-CoV-2 were documented in the sera of hamsters at different time-points from virus inoculation ranging from 7 to 21 days. Increased expression of proinflammatory and chemokine genes was demonstrated in the lungs of the SARS-CoV-2 infected animals, however with no increase in circulating levels of proteins such as TNF, interferon-γ, and IL-6.

Drugs and vaccines

Immunoprophylaxis with early convalescent serum achieved a significant decrease in viral lung load but not in lung pathology [57].

Ferrets, cat, and dog

Viral model and phenotype

Ferrets, cats, and dogs were administered intranasally or intratracheally with various doses and strains of SARS-CoV-2 (Table 5). Ferrets displayed elevated body temperature for several days associated with signs that differed according to the studies. These include decreased activity and appetite, sporadic cough, and no body weight loss [6063]. No clinical signs were reported either in cats or in dogs.

Histopathological changes

Ferrets exhibited acute bronchiolitis [61, 63], with perivasculitis and vasculitis [63], but with no discernible pneumonia. Cats disclosed lesions in epithelial nasal, tracheal, and lung mucosa (Table 5).

Viral and host interaction

The virus replication and shedding were demonstrated in the upper airways and rectal swabs in ferrets and cats, but the extent to other tissues varied in ferrets from none to multiple organs, including the lung, blood, and urine. No viral RNA was detected in cats’ lungs. Dogs showed RNA-positive rectal swab but none in the upper or lower airways. Viral transmission between ferrets and cats was demonstrated either through direct contact [55] or indirectly via airborne route [62].
Ferrets, cats, and dogs exhibited specific antibody response against SARS-CoV-2 [60, 62, 63]. A study of the ferret immune response to SARS-CoV-2 revealed a subdued low interferon type I and type III response that contrasts with increased chemokines and proinflammatory cytokine IL6, which is reminiscent of the human response [61].

Discussion

This systematic review of experimental animal models of SARS-CoV-2 induced- COVID-19 identified 13 peer-reviewed studies and 14 preprints that reported data on nonhuman primates [3749], mice [5056], hamsters [5659], ferrets [6063], cats, and dog [63] models of COVID-19. The main findings indicate that most of the animal models could mimic many features of mild human COVID-19 with a full recovery phenotype [3]. They also revealed that older animals display relatively more severe illness than the younger ones [38, 46, 48, 52, 54], which evokes human COVID-19 [3, 6]. However, none of the animal models replicated the severe or critical patterns associated with mortality as observed in humans with COVID-19 [3].
The results of this systematic review are consistent with studies of animal models of SARS-CoV and MERS-CoV, which failed to replicate the full spectrum of humans’ illness [65, 66]. Nonetheless, several features of mild COVID-19 in humans could be mirrored. High viral titers in the upper and lower respiratory tract and lung pathology were demonstrated in both large and small animal models. The pathology encompassed mild interstitial pneumonia, consolidation, and diffuse alveolar damage, albeit localized to a small lung area, edema, hyaline membrane formation, and inflammation. SARS-CoV-2 elicited specific antibody response against various viral proteins in the sera of most of the animal models.
This systematic review revealed that none of these newly established animal models replicated the common complications of human COVID-19 such as ARDS and coagulopathy [6, 8, 2833, 67, 68]. ARDS can be particularly severe and results in refractory hypoxemia requiring maximum respiratory supportive measures in the intensive care unit [6, 67, 68]. The coagulopathy can lead to severe complications such as massive pulmonary embolism, cerebrovascular stroke, and mesenteric infarction, including in younger people [8, 28, 32, 33]. The pathology underlying these two complications were recently revealed by post-mortem studies disclosing diffuse alveolar damage involving the whole lung, hyaline membrane formation, and infiltration with inflammatory cells, thus leaving no air space open for ventilation [17, 18, 64, 69, 70]. It also detected the presence of diffuse and widespread thrombosis in the micro- and macro-circulation, including the pulmonary circulation compromising the lung perfusion [17, 18]. This double hit affecting the ventilation and perfusion simultaneously underlies the intractable hypoxemia that contributed to the high mortality. None of the animal models replicated the respiratory failure, thromboembolic manifestations, and their pathological expression, hence, indicating that a wide gap separates the animal models from the full spectrum of COVID-19 in humans.
The mechanisms of the lung injury and coagulopathy are not well understood, although several known pathways were postulated including cytokine storm leading to upregulation of tissue factor [5, 9, 24], activation/injury of the endothelium infected by the virus [30, 67, 71], complement activation [72], alveolar hypoxia promoting thrombosis [73], and autoantibodies against phospholipid and lupus anticoagulant [74, 75] modulating the hemostasis and coagulation cascade directly. Hence, the development of animal models that replicate the dysregulation of the inflammation and coagulation could be important, as these would allow the deciphering of the intimate mechanisms at play. This, in turn, may aid in identifying therapeutic targets and the testing of immunotherapy, anticoagulation, and thrombolytic interventions and thereby may improve the outcome.
Both antiviral and vaccine therapies were tested in rhesus macaques and mice infected with SARS-CoV-2 [4042]. The antiviral drug stopped the viral replication and improved the pneumonitis [42, 55]. The vaccines induced an increase in titers of neutralizing antibodies in the sera that correlated with the decrease of viral replication and prevented the lung pathology [3941]. These results represent a substantial proof of the concept of antiviral or vaccine efficacy against SARS-CoV-2 in animal models. However, because of the lack of overt clinical illness, the rapid clearance of the virus, and spontaneous improvement of the pneumonitis without lethality, the models do not permit the full assessment of the duration of the protection of the vaccines, or the effect of antiviral therapy on survival.
Since the emergence of SARS-CoV infection in 2003 [76], followed by the MERS-CoV in 2012 [77], and now with COVID-19, researchers have not been able to develop a model of coronavirus infection that reproduces the severity and lethality seen in humans [65, 66]. One of the well-known reasons lies in the difference of ACE-2 receptor binding domain structure across species [78]. Human and primates have conserved a comparable structure that allows binding with high affinity to the SARS-CoV-2 [78]. The hamsters, ferrets, and cats maintained an intermediate affinity, while mice exhibit very low affinity [78]. The latter explains why wild-type mouse does not support SARS-CoV-2 replication, and hence, the necessity to create a chimera that expresses human ACE-2, to enable the use of this species as a model of COVID-19 [50]. More recently, a study applying single-cell RNA sequencing to nonhuman primate uncovered another explanation that may underlie the difference between nonhuman primates and humans in expressing the complex phenotype of COVID-19 [79]. The study reveals that the cellular expression and distribution of ACE2 and TMPRSS2 which are essential for virus entry in the cells and its spread inside the body differ in the lung, liver, and kidney between the two species. ACE2 expression was found lower in pneumocytes type II and higher in ciliated cells in nonhuman primate lung as compared to humans [40]. This is particularly significant as type II pneumocytes are critical targets of SARS-CoV-2 in humans and the pathogenesis of lung injury/damage. Finally, the innate immune response including the defense system against viruses diverged during evolution both at the transcriptional levels and cellular levels, which may also explain why the SARS-CoV-2 hardly progresses in these animals outside the respiratory system [80]. Taken together, these fundamental differences represent a real challenge to the successful development of an animal model that reproduces human COVID-19.
This systematic review has a few limitations. First, it is the high number of preprints included in this study that have not been peer-reviewed. Second, the animal models from the same species were difficult to compare across studies, as they used different viral strain, inoculum size, route of administration, and timing of tissue collection.

Conclusion: failure to reproduce a severe form of human COVID-19

This systematic review revealed that animal models of COVID19 mimic mild human COVID-19, but not the severe form COVID-19 associated with mortality. It also disclosed the knowledge generated by these models of COVID-19 including viral dynamic and transmission, pathogenesis, and testing of therapy and vaccines. Likewise, the study underlines the distinct advantages and limitations of each model, which should be considered when designing studies, interpreting pathogenic mechanisms, or extrapolating therapy or vaccines results to humans. Finally, harmonization of animal research protocols to generate results that are consistent, reproducible, and comparable across studies is needed.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13054-020-03304-8.

Acknowledgements

Not applicable
Not applicable
Not applicable

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, Si HR, Zhu Y, Li B, Huang CL, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270–3.PubMedPubMedCentralCrossRef Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, Si HR, Zhu Y, Li B, Huang CL, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270–3.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat 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(8):727–33.PubMedPubMedCentralCrossRef 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(8):727–33.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020;323(13):1239–2. Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020;323(13):1239–2.
4.
Zurück zum Zitat Zhang X, Tan Y, Ling Y, Lu G, Liu F, Yi Z, Jia X, Wu M, Shi B, Xu S, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature. 2020;583(7816):437–40. Zhang X, Tan Y, Ling Y, Lu G, Liu F, Yi Z, Jia X, Wu M, Shi B, Xu S, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature. 2020;583(7816):437–40.
5.
Zurück zum Zitat Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–9.PubMedPubMedCentralCrossRef Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–9.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, et al. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy Region, Italy. JAMA. 2020;323(16):1574–81.PubMedPubMedCentralCrossRef Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, et al. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy Region, Italy. JAMA. 2020;323(16):1574–81.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Guo T, Fan Y, Chen M, Wu X, Zhang L, He T, Wang H, Wan J, Wang X, Lu Z. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020;5(7):811–8. Guo T, Fan Y, Chen M, Wu X, Zhang L, He T, Wang H, Wan J, Wang X, Lu Z. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020;5(7):811–8.
8.
Zurück zum Zitat Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, Merdji H, Clere-Jehl R, Schenck M, Fagot Gandet F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1–10. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, Merdji H, Clere-Jehl R, Schenck M, Fagot Gandet F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1–10.
9.
Zurück zum Zitat Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506.PubMedPubMedCentralCrossRef Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367(6483):1260–3.PubMedPubMedCentralCrossRef Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367(6483):1260–3.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, 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(2):271–280.e278.PubMedPubMedCentralCrossRef Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, 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(2):271–280.e278.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Sungnak W, Huang N, Bécavin C, Berg M, Queen R, Litvinukova M, Talavera-López C, Maatz H, Reichart D, Sampaziotis F, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26(5):681–7.PubMedCrossRefPubMedCentral Sungnak W, Huang N, Bécavin C, Berg M, Queen R, Litvinukova M, Talavera-López C, Maatz H, Reichart D, Sampaziotis F, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26(5):681–7.PubMedCrossRefPubMedCentral
13.
Zurück zum Zitat Muus C, Luecken MD, Eraslan G, Waghray A, Heimberg G, Sikkema L, Kobayashi Y, Vaishnav ED, Subramanian A, Smilie C, et al. Integrated analyses of single-cell atlases reveal age, gender, and smoking status associations with cell type-specific expression of mediators of SARS-CoV-2 viral entry and highlights inflammatory programs in putative target cells. bioRxiv. https://doi.org/10.1101/2020.04.19.049254. Muus C, Luecken MD, Eraslan G, Waghray A, Heimberg G, Sikkema L, Kobayashi Y, Vaishnav ED, Subramanian A, Smilie C, et al. Integrated analyses of single-cell atlases reveal age, gender, and smoking status associations with cell type-specific expression of mediators of SARS-CoV-2 viral entry and highlights inflammatory programs in putative target cells. bioRxiv. https://​doi.​org/​10.​1101/​2020.​04.​19.​049254.
14.
Zurück zum Zitat Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, Cao Y, Yousif AS, Bals J, Hauser BM, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020;181(5):1016–1035.e1019.PubMedPubMedCentralCrossRef Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, Cao Y, Yousif AS, Bals J, Hauser BM, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020;181(5):1016–1035.e1019.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Li C, Ji F, Wang L, Wang L, Hao J, Dai M, Liu Y, Pan X, Fu J, Li L, et al. Asymptomatic and human-to-human transmission of SARS-CoV-2 in a 2-family cluster, Xuzhou, China. Emerg Infect Dis. 2020; 26(7):1626–8. Li C, Ji F, Wang L, Wang L, Hao J, Dai M, Liu Y, Pan X, Fu J, Li L, et al. Asymptomatic and human-to-human transmission of SARS-CoV-2 in a 2-family cluster, Xuzhou, China. Emerg Infect Dis. 2020; 26(7):1626–8.
16.
Zurück zum Zitat Rothe C, Schunk M, Sothmann P, Bretzel G, Froeschl G, Wallrauch C, Zimmer T, Thiel V, Janke C, Guggemos W, et al. Transmission of 2019-nCoV infection from an asymptomatic contact in Germany. N Engl J Med. 2020;382(10):970–1.PubMedPubMedCentralCrossRef Rothe C, Schunk M, Sothmann P, Bretzel G, Froeschl G, Wallrauch C, Zimmer T, Thiel V, Janke C, Guggemos W, et al. Transmission of 2019-nCoV infection from an asymptomatic contact in Germany. N Engl J Med. 2020;382(10):970–1.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Wichmann D, Sperhake JP, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, et al. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann Intern Med 2020;173(4):268–77. Wichmann D, Sperhake JP, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, et al. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann Intern Med 2020;173(4):268–77.
21.
Zurück zum Zitat Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054–62.PubMedPubMedCentralCrossRef Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054–62.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Liu Y, Yan LM, Wan L, Xiang TX, Le A, Liu JM, Peiris M, Poon LLM, Zhang W. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis. 2020;20(6):656–7.PubMedPubMedCentralCrossRef Liu Y, Yan LM, Wan L, Xiang TX, Le A, Liu JM, Peiris M, Poon LLM, Zhang W. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis. 2020;20(6):656–7.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Thevarajan I, Nguyen THO, Koutsakos M, Druce J, Caly L, van de Sandt CE, Jia X, Nicholson S, Catton M, Cowie B, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453–5.PubMedCrossRef Thevarajan I, Nguyen THO, Koutsakos M, Druce J, Caly L, van de Sandt CE, Jia X, Nicholson S, Catton M, Cowie B, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453–5.PubMedCrossRef
24.
Zurück zum Zitat Zhou Z, Ren L, Zhang L, Zhong J, Xiao Y, Jia Z, Guo L, Yang J, Wang C, Jiang S, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020;27(6):883–90.e882. Zhou Z, Ren L, Zhang L, Zhong J, Xiao Y, Jia Z, Guo L, Yang J, Wang C, Jiang S, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020;27(6):883–90.e882.
26.
Zurück zum Zitat Zheng M, Gao Y, Wang G, Song G, Liu S, Sun D, Xu Y, Tian Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol. 2020;17(5):533–5.PubMedPubMedCentralCrossRef Zheng M, Gao Y, Wang G, Song G, Liu S, Sun D, Xu Y, Tian Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol. 2020;17(5):533–5.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, Wang Q, Miao H. Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct Target Ther. 2020;5(1):33.PubMedPubMedCentralCrossRef Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, Wang Q, Miao H. Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct Target Ther. 2020;5(1):33.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Grillet F, Behr J, Calame P, Aubry S, Delabrousse E. Acute pulmonary embolism associated with COVID-19 pneumonia detected with pulmonary CT angiography. Radiology. 2020;296(3):E186–8.. Grillet F, Behr J, Calame P, Aubry S, Delabrousse E. Acute pulmonary embolism associated with COVID-19 pneumonia detected with pulmonary CT angiography. Radiology. 2020;296(3):E186–8..
29.
Zurück zum Zitat Klok FA, Kruip M, van der Meer NJM, Arbous MS, Gommers D, Kant KM, Kaptein FHJ, van Paassen J, Stals MAM, Huisman MV, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–7.PubMedPubMedCentralCrossRef Klok FA, Kruip M, van der Meer NJM, Arbous MS, Gommers D, Kant KM, Kaptein FHJ, van Paassen J, Stals MAM, Huisman MV, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–7.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Poor HD, Ventetuolo CE, Tolbert T, Chun G, Serrao G, Zeidman A, Dangayach NS, Olin J, Kohli-Seth R, Powell CA. COVID-19 critical illness pathophysiology driven by diffuse pulmonary thrombi and pulmonary endothelial dysfunction responsive to thrombolysis. medRxiv.. https://doi.org/10.1101/2020.04.17.20057125. Poor HD, Ventetuolo CE, Tolbert T, Chun G, Serrao G, Zeidman A, Dangayach NS, Olin J, Kohli-Seth R, Powell CA. COVID-19 critical illness pathophysiology driven by diffuse pulmonary thrombi and pulmonary endothelial dysfunction responsive to thrombolysis. medRxiv.. https://​doi.​org/​10.​1101/​2020.​04.​17.​20057125.
31.
Zurück zum Zitat Whyte CS, Morrow GB, Mitchell JL, Chowdary P, Mutch NJ. Fibrinolytic abnormalities in acute respiratory distress syndrome (ARDS) and versatility of thrombolytic drugs to treat COVID-19. J Thromb Haemost. 2020;18(7):1548–55. Whyte CS, Morrow GB, Mitchell JL, Chowdary P, Mutch NJ. Fibrinolytic abnormalities in acute respiratory distress syndrome (ARDS) and versatility of thrombolytic drugs to treat COVID-19. J Thromb Haemost. 2020;18(7):1548–55.
32.
Zurück zum Zitat Bhayana R, Som A, Li MD, Carey DE, Anderson MA, Blake MA, Catalano O, Gee MS, Hahn PF, Harisinghani M, et al. Abdominal imaging findings in COVID-19: preliminary observations. Radiology. 2020;297(1):E207–15. Bhayana R, Som A, Li MD, Carey DE, Anderson MA, Blake MA, Catalano O, Gee MS, Hahn PF, Harisinghani M, et al. Abdominal imaging findings in COVID-19: preliminary observations. Radiology. 2020;297(1):E207–15.
33.
Zurück zum Zitat Oxley TJ, Mocco J, Majidi S, Kellner CP, Shoirah H, Singh IP, De Leacy RA, Shigematsu T, Ladner TR, Yaeger KA, et al. Large-vessel stroke as a presenting feature of COVID-19 in the young. N Engl J Med. 2020;382(20):e60.PubMedCrossRef Oxley TJ, Mocco J, Majidi S, Kellner CP, Shoirah H, Singh IP, De Leacy RA, Shigematsu T, Ladner TR, Yaeger KA, et al. Large-vessel stroke as a presenting feature of COVID-19 in the young. N Engl J Med. 2020;382(20):e60.PubMedCrossRef
34.
Zurück zum Zitat Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18(5):1094–9.PubMedCrossRef Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18(5):1094–9.PubMedCrossRef
35.
Zurück zum Zitat Lakdawala SS, Menachery VD. The search for a COVID-19 animal model. Science. 2020;368(6494):942–3.PubMedCrossRef Lakdawala SS, Menachery VD. The search for a COVID-19 animal model. Science. 2020;368(6494):942–3.PubMedCrossRef
36.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.PubMedPubMedCentralCrossRef Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Munster VJ, Feldmann F, Williamson BN, 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(7824):268–272. Munster VJ, Feldmann F, Williamson BN, 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(7824):268–272.
38.
Zurück zum Zitat Yu P, Qi F, Xu Y, Li F, Liu P, Liu J, Bao L, Deng W, Gao H, Xiang Z, et al. Age-related rhesus macaque models of COVID-19. Anim Models Exp Med. 2020;3(1):93–7.CrossRef Yu P, Qi F, Xu Y, Li F, Liu P, Liu J, Bao L, Deng W, Gao H, Xiang Z, et al. Age-related rhesus macaque models of COVID-19. Anim Models Exp Med. 2020;3(1):93–7.CrossRef
39.
Zurück zum Zitat Yu J, Tostanoski LH, Peter L, Mercado NB, McMahan K, Mahrokhian SH, Nkolola JP, Liu J, Li Z, Chandrashekar A. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science. 2020;369(6505):806–811. Yu J, Tostanoski LH, Peter L, Mercado NB, McMahan K, Mahrokhian SH, Nkolola JP, Liu J, Li Z, Chandrashekar A. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science. 2020;369(6505):806–811.
41.
Zurück zum Zitat 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(6499):77–81. 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(6499):77–81.
43.
Zurück zum Zitat Chandrashekar A, Liu J, Martinot AJ, McMahan K, Mercado NB, Peter L, Tostanoski LH, Yu J, Maliga Z, Nekorchuk M, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369(6505):812–17. Chandrashekar A, Liu J, Martinot AJ, McMahan K, Mercado NB, Peter L, Tostanoski LH, Yu J, Maliga Z, Nekorchuk M, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369(6505):812–17.
47.
Zurück zum Zitat Finch CL, Crozier I, Lee JH, Byrum R, Cooper TK, Liang J, Sharer K, Solomon J, Sayre PJ, Kocher G, et al. Characteristic and quantifiable COVID-19-like abnormalities in CT- and PET/CT-imaged lungs of SARS-CoV-2-infected crab-eating macaques. bioRxiv. https://doi.org/10.1101/2020.05.14.096727. Finch CL, Crozier I, Lee JH, Byrum R, Cooper TK, Liang J, Sharer K, Solomon J, Sayre PJ, Kocher G, et al. Characteristic and quantifiable COVID-19-like abnormalities in CT- and PET/CT-imaged lungs of SARS-CoV-2-infected crab-eating macaques. bioRxiv. https://​doi.​org/​10.​1101/​2020.​05.​14.​096727.
48.
Zurück zum Zitat Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science. 2020;368(6494):1012–5.PubMedPubMedCentralCrossRef Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science. 2020;368(6494):1012–5.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat 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(7818):830–3. 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(7818):830–3.
53.
Zurück zum Zitat Lv H, Wu NC, Tsang OT, Yuan M, Perera R, Leung WS, So RTY, Chan JMC, Yip GK, Chik TSH, et al. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep. 2020;31(9):107725.PubMedPubMedCentralCrossRef Lv H, Wu NC, Tsang OT, Yuan M, Perera R, Leung WS, So RTY, Chan JMC, Yip GK, Chik TSH, et al. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep. 2020;31(9):107725.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Pruijssers AJ, George AS, Schäfer A, Leist SR, Gralinksi LE, Dinnon KH, Yount BL, Agostini ML, Stevens LJ, Chappell JD, et al. Remdesivir potently inhibits SARS-CoV-2 in human lung cells and chimeric SARS-CoV expressing the SARS-CoV-2 RNA polymerase in mice. bioRxiv. https://doi.org/10.1101/2020.04.27.064279. Pruijssers AJ, George AS, Schäfer A, Leist SR, Gralinksi LE, Dinnon KH, Yount BL, Agostini ML, Stevens LJ, Chappell JD, et al. Remdesivir potently inhibits SARS-CoV-2 in human lung cells and chimeric SARS-CoV expressing the SARS-CoV-2 RNA polymerase in mice. bioRxiv. https://​doi.​org/​10.​1101/​2020.​04.​27.​064279.
56.
Zurück zum Zitat Boudewijns R, Thibaut HJ, Kaptein SJF, Li R, Vergote V, Seldeslachts L, De Keyzer C, Sharma S, Jansen S, Weyenbergh JV, et al. STAT2 signaling as double-edged sword restricting viral dissemination but driving severe pneumonia in SARS-CoV-2 infected hamsters. bioRxiv. https://doi.org/10.1101/2020.04.23.056838. Boudewijns R, Thibaut HJ, Kaptein SJF, Li R, Vergote V, Seldeslachts L, De Keyzer C, Sharma S, Jansen S, Weyenbergh JV, et al. STAT2 signaling as double-edged sword restricting viral dissemination but driving severe pneumonia in SARS-CoV-2 infected hamsters. bioRxiv. https://​doi.​org/​10.​1101/​2020.​04.​23.​056838.
57.
Zurück zum Zitat Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW, Wen L, Liang R, Cao J, Chen Y, Tang K, Luo C, Cai JP, Kok KH, Chu H, Chan KH, Sridhar S, Chen Z, Chen H, To KK, Yuen KY. 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:ciaa325. https://doi.org/10.1093/cid/ciaa325. Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW, Wen L, Liang R, Cao J, Chen Y, Tang K, Luo C, Cai JP, Kok KH, Chu H, Chan KH, Sridhar S, Chen Z, Chen H, To KK, Yuen KY. 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:ciaa325. https://​doi.​org/​10.​1093/​cid/​ciaa325.
59.
Zurück zum Zitat Sia SF, Yan L-M, Chin AWH, Fung K, Choy K-T, Wong AYL, Kaewpreedee P, Perera RAPM, Poon LLM, Nicholls JM, et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. 2020;583(7818):834–8. Sia SF, Yan L-M, Chin AWH, Fung K, Choy K-T, Wong AYL, Kaewpreedee P, Perera RAPM, Poon LLM, Nicholls JM, et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. 2020;583(7818):834–8.
60.
Zurück zum Zitat Kim YI, Kim SG, Kim SM, Kim EH, Park SJ, Yu KM, Chang JH, Kim EJ, Lee S, Casel MAB, et al. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe. 2020;27(5):704–709.e702.PubMedPubMedCentralCrossRef Kim YI, Kim SG, Kim SM, Kim EH, Park SJ, Yu KM, Chang JH, Kim EJ, Lee S, Casel MAB, et al. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe. 2020;27(5):704–709.e702.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181(5):1036–1045.e1039.PubMedPubMedCentralCrossRef Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181(5):1036–1045.e1039.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat 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(6494):1016–20.PubMedPubMedCentralCrossRef 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(6494):1016–20.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Hanley B, Lucas SB, Youd E, Swift B, Osborn M. Autopsy in suspected COVID-19 cases. J Clin Pathol. 2020;73(5):239–42.PubMedCrossRef Hanley B, Lucas SB, Youd E, Swift B, Osborn M. Autopsy in suspected COVID-19 cases. J Clin Pathol. 2020;73(5):239–42.PubMedCrossRef
66.
Zurück zum Zitat Sutton TC, Subbarao K. Development of animal models against emerging coronaviruses: from SARS to MERS coronavirus. Virology. 2015;479-480:247–58.PubMedCrossRef Sutton TC, Subbarao K. Development of animal models against emerging coronaviruses: from SARS to MERS coronavirus. Virology. 2015;479-480:247–58.PubMedCrossRef
67.
Zurück zum Zitat Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med. 2020;383(2):120–8. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med. 2020;383(2):120–8.
68.
Zurück zum Zitat Marini JJ, Gattinoni L. Management of COVID-19 respiratory distress. JAMA. 2020;323(22):2329–30. Marini JJ, Gattinoni L. Management of COVID-19 respiratory distress. JAMA. 2020;323(22):2329–30.
69.
Zurück zum Zitat Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420–2.PubMedPubMedCentralCrossRef Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420–2.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Nicholls JM, Poon LL, Lee KC, Ng WF, Lai ST, Leung CY, Chu CM, Hui PK, Mak KL, Lim W, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet. 2003;361(9371):1773–8.PubMedPubMedCentralCrossRef Nicholls JM, Poon LL, Lee KC, Ng WF, Lai ST, Leung CY, Chu CM, Hui PK, Mak KL, Lim W, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet. 2003;361(9371):1773–8.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, Mehra MR, Schuepbach RA, Ruschitzka F, Moch H. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395(10234):1417–8.PubMedPubMedCentralCrossRef Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, Mehra MR, Schuepbach RA, Ruschitzka F, Moch H. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395(10234):1417–8.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Mulvey JJ, Magro CM, Ma LX, Nuovo GJ, Baergen RN. Analysis of complement deposition and viral RNA in placentas of COVID-19 patients. Ann Diagn Pathol. 2020;46:151530.PubMedPubMedCentralCrossRef Mulvey JJ, Magro CM, Ma LX, Nuovo GJ, Baergen RN. Analysis of complement deposition and viral RNA in placentas of COVID-19 patients. Ann Diagn Pathol. 2020;46:151530.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Gupta N, Zhao YY, Evans CE. The stimulation of thrombosis by hypoxia. Thromb Res. 2019;181:77–83.PubMedCrossRef Gupta N, Zhao YY, Evans CE. The stimulation of thrombosis by hypoxia. Thromb Res. 2019;181:77–83.PubMedCrossRef
74.
Zurück zum Zitat Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, Chen H, Ding X, Zhao H, Zhang H, et al. Coagulopathy and antiphospholipid antibodies in patients with COVID-19. N Engl J Med. 2020;382(17):e38.PubMedCrossRef Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, Chen H, Ding X, Zhao H, Zhang H, et al. Coagulopathy and antiphospholipid antibodies in patients with COVID-19. N Engl J Med. 2020;382(17):e38.PubMedCrossRef
75.
Zurück zum Zitat Bowles L, Platton S, Yartey N, Dave M, Lee K, Hart DP, MacDonald V, Green L, Sivapalaratnam S, Pasi KJ, et al. Lupus anticoagulant and abnormal coagulation tests in patients with COVID-19. N Engl J Med. 2020;383(3):288–90. Bowles L, Platton S, Yartey N, Dave M, Lee K, Hart DP, MacDonald V, Green L, Sivapalaratnam S, Pasi KJ, et al. Lupus anticoagulant and abnormal coagulation tests in patients with COVID-19. N Engl J Med. 2020;383(3):288–90.
76.
Zurück zum Zitat Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, Ahuja A, Yung MY, Leung CB, To KF, et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348(20):1986–94.PubMedCrossRef Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, Ahuja A, Yung MY, Leung CB, To KF, et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348(20):1986–94.PubMedCrossRef
77.
Zurück zum Zitat Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME, Fouchier RAM. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):1814–20.PubMedCrossRef Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME, Fouchier RAM. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):1814–20.PubMedCrossRef
80.
Zurück zum Zitat Hagai T, Chen X, Miragaia RJ, Rostom R, Gomes T, Kunowska N, Henriksson J, Park J-E, Proserpio V, Donati G, et al. Gene expression variability across cells and species shapes innate immunity. Nature. 2018;563(7730):197–202.PubMedPubMedCentralCrossRef Hagai T, Chen X, Miragaia RJ, Rostom R, Gomes T, Kunowska N, Henriksson J, Park J-E, Proserpio V, Donati G, et al. Gene expression variability across cells and species shapes innate immunity. Nature. 2018;563(7730):197–202.PubMedPubMedCentralCrossRef
Metadaten
Titel
Evidence of a wide gap between COVID-19 in humans and animal models: a systematic review
verfasst von
Salleh N. Ehaideb
Mashan L. Abdullah
Bisher Abuyassin
Abderrezak Bouchama
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Schlagwort
COVID-19
Erschienen in
Critical Care / Ausgabe 1/2020
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03304-8

Weitere Artikel der Ausgabe 1/2020

Critical Care 1/2020 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.