The trinity of COVID-19: immunity, inflammation and intervention

Matthew Zirui Tay, Chek Meng Poh, Laurent Rénia, Paul A MacAry, Lisa F P Ng, Matthew Zirui Tay, Chek Meng Poh, Laurent Rénia, Paul A MacAry, Lisa F P Ng

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic. Alongside investigations into the virology of SARS-CoV-2, understanding the fundamental physiological and immunological processes underlying the clinical manifestations of COVID-19 is vital for the identification and rational design of effective therapies. Here, we provide an overview of the pathophysiology of SARS-CoV-2 infection. We describe the interaction of SARS-CoV-2 with the immune system and the subsequent contribution of dysfunctional immune responses to disease progression. From nascent reports describing SARS-CoV-2, we make inferences on the basis of the parallel pathophysiological and immunological features of the other human coronaviruses targeting the lower respiratory tract - severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). Finally, we highlight the implications of these approaches for potential therapeutic interventions that target viral infection and/or immunoregulation.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Chronology of events during SARS-CoV-2…
Fig. 1. Chronology of events during SARS-CoV-2 infection.
When severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects cells expressing the surface receptors angiotensin-converting enzyme 2 (ACE2) and TMPRSS2, the active replication and release of the virus cause the host cell to undergo pyroptosis and release damage-associated molecular patterns, including ATP, nucleic acids and ASC oligomers. These are recognized by neighbouring epithelial cells, endothelial cells and alveolar macrophages, triggering the generation of pro-inflammatory cytokines and chemokines (including IL-6, IP-10, macrophage inflammatory protein 1α (MIP1α), MIP1β and MCP1). These proteins attract monocytes, macrophages and T cells to the site of infection, promoting further inflammation (with the addition of IFNγ produced by T cells) and establishing a pro-inflammatory feedback loop. In a defective immune response (left side) this may lead to further accumulation of immune cells in the lungs, causing overproduction of pro-inflammatory cytokines, which eventually damages the lung infrastructure. The resulting cytokine storm circulates to other organs, leading to multi-organ damage. In addition, non-neutralizing antibodies produced by B cells may enhance SARS-CoV-2 infection through antibody-dependent enhancement (ADE), further exacerbating organ damage. Alternatively, in a healthy immune response (right side), the initial inflammation attracts virus-specific T cells to the site of infection, where they can eliminate the infected cells before the virus spreads. Neutralizing antibodies in these individuals can block viral infection, and alveolar macrophages recognize neutralized viruses and apoptotic cells and clear them by phagocytosis. Altogether, these processes lead to clearance of the virus and minimal lung damage, resulting in recovery. G-CSF, granulocyte colony-stimulating factor; TNF, tumour necrosis factor.
Fig. 2. The structure of the trimeric…
Fig. 2. The structure of the trimeric spike protein of SARS-CoV-2.
The receptor-binding domain (RBD) is shown interacting with its receptor, human angiotensin-converting enzyme 2 (ACE2). SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. Adapted from Protein Data Bank IDs 6VSB and 6VW1 (ref.150).
Fig. 3. Potential therapeutic approaches against SARS-CoV-2.
Fig. 3. Potential therapeutic approaches against SARS-CoV-2.
(1) Antibodies against the spike protein (raised through vaccination or by adoptive transfer) could block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from interacting with the angiotensin-converting enzyme 2 (ACE2) receptor on host cells. (2) Protease inhibitors against the serine protease TMPRSS2 can prevent spike protein cleavage, which is necessary for viral fusion into the host cell. Blocking either ACE2 interaction or viral fusion could prevent the virus from infecting the host cell. (3) Virus-specific memory CD8+ T cells from a previous vaccination or infection can differentiate into effector cells during rechallenge. When they identify infected cells presenting virus-specific epitopes, they degranulate and kill infected cells before they can produce mature virions. (4) In a novel treatment method that targets the cytokine storm symptoms, the blood of patients with coronavirus disease 2019 (COVID-19) can be passed through customized columns that are specially designed to trap pro-inflammatory cytokines, before the purified blood is passed back into patients.
Fig. 4. Sequence alignment and structural comparison…
Fig. 4. Sequence alignment and structural comparison of SARS-CoV and SARS-CoV-2 spike proteins.
a | Sequence alignment of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, with conserved amino acid residues shown in black and non-conserved residues shown in colours. b | The 3D structure of SARS-CoV-2 (Protein Data Bank ID 6VSB, peach ribbon) is superimposed on the SARS-CoV receptor-binding motif (RBM) complex with the neutralizing antibody (nAb; red ribbon) interfacing with the RBM (Protein Data Bank 2DD8 (ref.), purple ribbon). Peach and purple spheres denote the RBMs of SARS-CoV-2 and SARS-CoV, respectively. Magenta spheres denote non-synonymous alterations in the SARS-CoV-2 spike protein that have been reported.

References

    1. World Health Organization. WHO Director-General’s opening remarks at the media briefing on COVID-19 - 11 March 2020. WHO (2020).
    1. Fehr AR, Perlman S. Coronaviruses: an overview of their replication and pathogenesis. Methods. Mol. Biol. 2015;1282:1–23. doi: 10.1007/978-1-4939-2438-7_1.
    1. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020;5:536–544. doi: 10.1038/s41564-020-0695-z.
    1. Zhou P, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. doi: 10.1038/s41586-020-2012-7.
    1. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat. Med. 2020 doi: 10.1038/s41591-020-0820-9.
    1. Guan WJ, et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2002032.
    1. Pung R, et al. Investigation of three clusters of COVID-19 in Singapore: implications for surveillance and response measures. Lancet. 2020;395:1039–1046. doi: 10.1016/S0140-6736(20)30528-6.
    1. Lauer SA, et al. The incubation period of coronavirus disease 2019 (COVID-19) from publicly reported confirmed cases: estimation and application. Ann. Intern. Med. 2020 doi: 10.7326/m20-0504.
    1. Li Q, et al. Early transmission dynamics in Wuhan, China, of Novel Coronavirus-infected pneumonia. N. Engl. J. Med. 2020;382:1199–1207. doi: 10.1056/NEJMoa2001316.
    1. Chan JF, et al. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster. Lancet. 2020;395:514–523. doi: 10.1016/S0140-6736(20)30154-9.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Chen Guang, Wu Di, Guo Wei, Cao Yong, Huang Da, Wang Hongwu, Wang Tao, Zhang Xiaoyun, Chen Huilong, Yu Haijing, Zhang Xiaoping, Zhang Minxia, Wu Shiji, Song Jianxin, Chen Tao, Han Meifang, Li Shusheng, Luo Xiaoping, Zhao Jianping, Ning Qin. Clinical and immunological features of severe and moderate coronavirus disease 2019. Journal of Clinical Investigation. 2020;130(5):2620–2629. doi: 10.1172/JCI137244.
    1. Liu Y, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci. China Life Sci. 2020;63:364–374. doi: 10.1007/s11427-020-1643-8.
    1. Chen N, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–513. doi: 10.1016/S0140-6736(20)30211-7.
    1. Phan LT, et al. Importation and human-to-human transmission of a novel coronavirus in Vietnam. N. Engl. J. Med. 2020;382:872–874. doi: 10.1056/NEJMc2001272.
    1. Pan Y, Zhang D, Yang P, Poon LLM, Wang Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect. Dis. 2020 doi: 10.1016/s1473-3099(20)30113-4.
    1. Kim JY, et al. Viral load kinetics of SARS-CoV-2 infection in first two patients in Korea. J. Korean Med. Sci. 2020;35:e86. doi: 10.3346/jkms.2020.35.e86.
    1. Zou L, et al. SARS-CoV-2 viral load in upper respiratory specimens of infected patients. N. Engl. J. Med. 2020;382:1177–1179. doi: 10.1056/NEJMc2001737.
    1. Peiris JS, et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet. 2003;361:1767–1772. doi: 10.1016/S0140-6736(03)13412-5.
    1. Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323:1061–1069. doi: 10.1001/jama.2020.1585.
    1. Wong CK, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin. Exp. Immunol. 2004;136:95–103. doi: 10.1111/j.1365-2249.2004.02415.x.
    1. Zhang, B. et al. Clinical characteristics of 82 death cases with COVID-19. Preprint at medRxiv10.1101/2020.02.26.20028191 (2020).
    1. Chu KH, et al. Acute renal impairment in coronavirus-associated severe acute respiratory syndrome. Kidney Int. 2005;67:698–705. doi: 10.1111/j.1523-1755.2005.67130.x.
    1. Jia HP, et al. ACE2 receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J. Virol. 2005;79:14614–14621. doi: 10.1128/JVI.79.23.14614-14621.2005.
    1. Xu H, et al. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral. Sci. 2020;12:8. doi: 10.1038/s41368-020-0074-x.
    1. Hamming I, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004;203:631–637. doi: 10.1002/path.1570.
    1. Zhao, Y. et al. Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov. Preprint at bioRxiv10.1101/2020.01.26.919985 (2020).
    1. Walls AC, et al. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020 doi: 10.1016/j.cell.2020.02.058.
    1. Imai Y, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–116. doi: 10.1038/nature03712.
    1. Imai Y, Kuba K, Penninger JM. The discovery of angiotensin-converting enzyme 2 and its role in acute lung injury in mice. Exp. Physiol. 2008;93:543–548. doi: 10.1113/expphysiol.2007.040048.
    1. Kuba K, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879. doi: 10.1038/nm1267.
    1. Kuba K, Imai Y, Penninger JM. Angiotensin-converting enzyme 2 in lung diseases. Curr. Opin. Pharmacol. 2006;6:271–276. doi: 10.1016/j.coph.2006.03.001.
    1. Epidemiology Working Group for NCIP Epidemic Response The epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) in China. Chin. J. Epidemiol. 2020;41:145–151.
    1. Taneja V. Sex hormones determine immune response. Front. Immunol. 2018;9:1931. doi: 10.3389/fimmu.2018.01931.
    1. Xiao X, Chakraborti S, Dimitrov AS, Gramatikoff K, Dimitrov DS. The SARS-CoV S glycoprotein: expression and functional characterization. Biochem. Biophys. Res. Commun. 2003;312:1159–1164. doi: 10.1016/j.bbrc.2003.11.054.
    1. Babcock GJ, Esshaki DJ, Thomas WD, Jr., Ambrosino DM. Amino acids 270 to 510 of the severe acute respiratory syndrome coronavirus spike protein are required for interaction with receptor. J. Virol. 2004;78:4552–4560. doi: 10.1128/JVI.78.9.4552-4560.2004.
    1. Wong SK, Li W, Moore MJ, Choe H, Farzan M. A 193-amino acid fragment of the SARS coronavirus S protein efficiently binds angiotensin-converting enzyme 2. J. Biol. Chem. 2004;279:3197–3201. doi: 10.1074/jbc.C300520200.
    1. Simmons G, et al. Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl Acad. Sci. USA. 2005;102:11876–11881. doi: 10.1073/pnas.0505577102.
    1. Bosch BJ, et al. Severe acute respiratory syndrome coronavirus (SARS-CoV) infection inhibition using spike protein heptad repeat-derived peptides. Proc. Natl Acad. Sci. USA. 2004;101:8455–8460. doi: 10.1073/pnas.0400576101.
    1. Liu S, et al. Interaction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: implications for virus fusogenic mechanism and identification of fusion inhibitors. Lancet. 2004;363:938–947. doi: 10.1016/S0140-6736(04)15788-7.
    1. Chen Y, Guo Y, Pan Y, Zhao ZJ. Structure analysis of the receptor binding of 2019-nCoV. Biochem. Biophys. Res. Commun. 2020;525:135–140. doi: 10.1016/j.bbrc.2020.02.071.
    1. Wrapp D, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263. doi: 10.1126/science.abb2507.
    1. Coutard B, et al. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020;176:104742. doi: 10.1016/j.antiviral.2020.104742.
    1. Hoffmann M, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020 doi: 10.1016/j.cell.2020.02.052.
    1. Richardson P, et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020;395:e30–e31. doi: 10.1016/S0140-6736(20)30304-4.
    1. Chinese Clinical Trial Register. (2020).
    1. Pipeline Review. APEIRON’s respiratory drug product to start pilot clinical trial to treat coronavirus disease COVID-19 in China. Pipeline Review (2020).
    1. Wang M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271. doi: 10.1038/s41422-020-0282-0.
    1. Yamamoto M, et al. Identification of nafamostat as a potent inhibitor of Middle East respiratory syndrome coronavirus S protein-mediated membrane fusion using the split-protein-based cell-cell fusion assay. Antimicrob. Agents Chemother. 2016;60:6532–6539. doi: 10.1128/AAC.01043-16.
    1. Zhang H, Penninger JM, Li Y, Zhong N, Slutsky AS. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46:586–590. doi: 10.1007/s00134-020-05985-9.
    1. Park WB, et al. Virus isolation from the first patient with SARS-CoV-2 in Korea. J. Korean Med. Sci. 2020;35:e84. doi: 10.3346/jkms.2020.35.e84.
    1. Zhang H, et al. Histopathologic changes and SARS-CoV-2 immunostaining in the lung of a patient with COVID-19. Ann. Intern. Med. 2020 doi: 10.7326/m20-0533.
    1. Chen IY, Moriyama M, Chang MF, Ichinohe T. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front. Microbiol. 2019;10:50. doi: 10.3389/fmicb.2019.00050.
    1. Fink SL, Cookson BT. Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells. Infect. Immun. 2005;73:1907–1916. doi: 10.1128/IAI.73.4.1907-1916.2005.
    1. Yang, M. Cell pyroptosis, a potential pathogenic mechanism of 2019-nCoV infection. SSRN10.2139/ssrn.3527420 (2020).
    1. Huang KJ, et al. An interferon-gamma-related cytokine storm in SARS patients. J. Med. Virol. 2005;75:185–194. doi: 10.1002/jmv.20255.
    1. Xu Z, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020;8:420–422. doi: 10.1016/S2213-2600(20)30076-X.
    1. Tian S, et al. Pulmonary pathology of early phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J. Thorac. Oncol. 2020 doi: 10.1016/j.jtho.2020.02.010.
    1. Qin C, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa248.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. Liao, M. et al. The landscape of lung bronchoalveolar immune cells in COVID-19 revealed by single-cell RNA sequencing. Preprint at medRxiv10.1101/2020.02.23.20026690 (2020).
    1. Zhou Y, et al. Pathogenic T cells and inflammatory monocytes incite inflammatory storm in severe COVID-19 patients. Natl Sci. Rev. 2020 doi: 10.1093/nsr/nwaa041.
    1. Siu KL, Chan CP, Kok KH, Chiu-Yat Woo P, Jin DY. Suppression of innate antiviral response by severe acute respiratory syndrome coronavirus M protein is mediated through the first transmembrane domain. Cell. Mol. Immunol. 2014;11:141–149. doi: 10.1038/cmi.2013.61.
    1. Versteeg GA, Bredenbeek PJ, van den Worm SH, Spaan WJ. Group 2 coronaviruses prevent immediate early interferon induction by protection of viral RNA from host cell recognition. Virology. 2007;361:18–26. doi: 10.1016/j.virol.2007.01.020.
    1. Sun L, et al. Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. PLoS One. 2012;7:e30802. doi: 10.1371/journal.pone.0030802.
    1. Frieman M, Ratia K, Johnston RE, Mesecar AD, Baric RS. Severe acute respiratory syndrome coronavirus papain-like protease ubiquitin-like domain and catalytic domain regulate antagonism of IRF3 and NF-kappaB signaling. J. Virol. 2009;83:6689–6705. doi: 10.1128/JVI.02220-08.
    1. Frieman M, et al. Severe acute respiratory syndrome coronavirus ORF6 antagonizes STAT1 function by sequestering nuclear import factors on the rough endoplasmic reticulum/Golgi membrane. J. Virol. 2007;81:9812–9824. doi: 10.1128/JVI.01012-07.
    1. Narayanan K, et al. Severe acute respiratory syndrome coronavirus nsp1 suppresses host gene expression, including that of type I interferon, in infected cells. J. Virol. 2008;82:4471–4479. doi: 10.1128/JVI.02472-07.
    1. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020 doi: 10.1007/s00134-020-05991-x.
    1. Zhao J, Zhao J, Legge K, Perlman S. Age-related increases in PGD2 expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J. Clin. Invest. 2011;121:4921–4930. doi: 10.1172/JCI59777.
    1. Kam KQ, et al. A well infant with coronavirus disease 2019 (COVID-19) with high viral load. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa201.
    1. Dong Y, et al. Epidemiological characteristics of 2143 pediatric patients with 2019 coronavirus disease in China. Pediatrics. 2020 doi: 10.1542/peds.2020-0702.
    1. Gu J, et al. Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 2005;202:415–424. doi: 10.1084/jem.20050828.
    1. Cheung CY, et al. Cytokine responses in severe acute respiratory syndrome coronavirus-infected macrophages in vitro: possible relevance to pathogenesis. J. Virol. 2005;79:7819–7826. doi: 10.1128/JVI.79.12.7819-7826.2005.
    1. Yilla M, et al. SARS-coronavirus replication in human peripheral monocytes/macrophages. Virus Res. 2005;107:93–101. doi: 10.1016/j.virusres.2004.09.004.
    1. Tseng CT, Perrone LA, Zhu H, Makino S, Peters CJ. Severe acute respiratory syndrome and the innate immune responses: modulation of effector cell function without productive infection. J. Immunol. 2005;174:7977–7985. doi: 10.4049/jimmunol.174.12.7977.
    1. Law HK, et al. Chemokine up-regulation in SARS-coronavirus-infected, monocyte-derived human dendritic cells. Blood. 2005;106:2366–2374. doi: 10.1182/blood-2004-10-4166.
    1. US National Library of Medicine. (2020).
    1. Stockman LJ, Bellamy R, Garner P. SARS: systematic review of treatment effects. PLoS Med. 2006;3:e343. doi: 10.1371/journal.pmed.0030343.
    1. Tai DY. Pharmacologic treatment of SARS: current knowledge and recommendations. Ann. Acad. Med. Singap. 2007;36:438–443.
    1. Chinese Clinical Trial Register. (2020).
    1. Liu, A. China turns Roche arthritis drug Actemra against COVID-19 in new treatment guidelines. FiercePharma (2020).
    1. Roivant Sciences. Roivant announces development of anti-GM-CSF monoclonal antibody to prevent and treat acute respiratory distress syndrome (ARDS) in patients with COVID-19. Roivant Sciences (2020).
    1. Humanigen. Humanigen partners with CTI, a leading contract research organization, for planned phase III study for lenzilumab for coronavirus treatment. Humanigen (2020).
    1. Izana Bioscience. Initiation of two-centre compassionate use study involving namilumab in the treatment of individual patients with rapidly worsening COVID-19 infection in Italy. Izana Bioscience (2020).
    1. CytoSorbents Corportation. CytoSorb, the Wuhan coronavirus, and cytokine Newswire (2020).
    1. Chen, C. et al. Thalidomide combined with low-dose glucocorticoid in the treatment of COVID-19 pneumonia. Preprints (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. Tobinick E. TNF-alpha inhibition for potential therapeutic modulation of SARS coronavirus infection. Curr. Med. Res. Opin. 2004;20:39–40. doi: 10.1185/030079903125002757.
    1. Gautret P, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents. 2020 doi: 10.1016/j.ijantimicag.2020.105949.
    1. Yao X, 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 doi: 10.1093/cid/ciaa237.
    1. Shukla AM, Wagle Shukla A. Expanding horizons for clinical applications of chloroquine, hydroxychloroquine, and related structural analogues. Drugs Context. 2019;8:2019-9-1. doi: 10.7573/dic.2019-9-1.
    1. Cortegiani A, Ingoglia G, Ippolito M, Giarratano A, Einav S. A systematic review on the efficacy and safety of chloroquine for the treatment of COVID-19. J. Crit. Care. 2020 doi: 10.1016/j.jcrc.2020.03.005.
    1. Wong RS, et al. Haematological manifestations in patients with severe acute respiratory syndrome: retrospective analysis. BMJ. 2003;326:1358–1362. doi: 10.1136/bmj.326.7403.1358.
    1. Cui W, et al. Expression of lymphocytes and lymphocyte subsets in patients with severe acute respiratory syndrome. Clin. Infect. Dis. 2003;37:857–859. doi: 10.1086/378587.
    1. Li T, et al. Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome. J. Infect. Dis. 2004;189:648–651. doi: 10.1086/381535.
    1. Zheng H-Y, et al. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell. Mol. Immunol. 2020 doi: 10.1038/s41423-020-0401-3.
    1. Libraty DH, O’Neil KM, Baker LM, Acosta LP, Olveda RM. Human CD4+ memory T-lymphocyte responses to SARS coronavirus infection. Virology. 2007;368:317–321. doi: 10.1016/j.virol.2007.07.015.
    1. Yang LT, et al. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin. Immunol. 2006;120:171–178. doi: 10.1016/j.clim.2006.05.002.
    1. Oh HLJ, Gan K-ES, Bertoletti A, Tan YJ. Understanding the T cell immune response in SARS coronavirus infection. Emerg. Microbes. Infect. 2012;1:e23.
    1. Shin HS, et al. Immune responses to Middle East respiratory syndrome coronavirus during the acute and convalescent phases of human infection. Clin. Infect. Dis. 2019;68:984–992. doi: 10.1093/cid/ciy595.
    1. Chen J, et al. Cellular immune responses to severe acute respiratory syndrome coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J. Virol. 2010;84:1289–1301. doi: 10.1128/JVI.01281-09.
    1. Roberts A, et al. A mouse-adapted SARS-coronavirus causes disease and mortality in BALB/c mice. PLoS Pathog. 2007;3:e5. doi: 10.1371/journal.ppat.0030005.
    1. Zhao J, Zhao J, Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J. Virol. 2010;84:9318–9325. doi: 10.1128/JVI.01049-10.
    1. Deming D, et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med. 2006;3:e525. doi: 10.1371/journal.pmed.0030525.
    1. Yasui F, et al. Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV. J. Immunol. 2008;181:6337–6348. doi: 10.4049/jimmunol.181.9.6337.
    1. Bolles M, et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 2011;85:12201–12215. doi: 10.1128/JVI.06048-11.
    1. Lurie N, Saville M, Hatchett R, Halton J. Developing Covid-19 vaccines at pandemic speed. N. Engl. J. Med. 2020 doi: 10.1056/NEJMp2005630.
    1. Thevarajan I, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat. Med. 2020 doi: 10.1038/s41591-020-0819-2.
    1. Tan YJ, et al. Profiles of antibody responses against severe acute respiratory syndrome coronavirus recombinant proteins and their potential use as diagnostic markers. Clin. Diagn. Lab. Immunol. 2004;11:362–371. doi: 10.1128/CDLI.11.2.362-371.2004.
    1. Wu HS, et al. Early detection of antibodies against various structural proteins of the SARS-associated coronavirus in SARS patients. J. Biomed. Sci. 2004;11:117–126. doi: 10.1007/BF02256554.
    1. Nie Y, et al. Neutralizing antibodies in patients with severe acute respiratory syndrome-associated coronavirus infection. J. Infect. Dis. 2004;190:1119–1126. doi: 10.1086/423286.
    1. Temperton NJ, et al. Longitudinally profiling neutralizing antibody response to SARS coronavirus with pseudotypes. Emerg. Infect. Dis. 2005;11:411–416. doi: 10.3201/eid1103.040906.
    1. CGTN. Expert: recovered coronavirus patients are still prone to reinfection. YouTube (2020).
    1. The Straits Times. Japanese woman reinfected with coronavirus weeks after initial recovery. The Straits Times (2020).
    1. NHK World-Japan. Japanese man tests positive for coronavirus again. NHK (2020).
    1. Xinhua. China puts 245 COVID-19 patients on convalescent plasma therapy. Xinhuanet (2020).
    1. Cheng Y, et al. Use of convalescent plasma therapy in SARS patients in Hong Kong. Eur. J. Clin. Microbiol. Infect. Dis. 2005;24:44–46. doi: 10.1007/s10096-004-1271-9.
    1. Soo YO, et al. Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients. Clin. Microbiol. Infect. 2004;10:676–678. doi: 10.1111/j.1469-0691.2004.00956.x.
    1. Yeh KM, et al. Experience of using convalescent plasma for severe acute respiratory syndrome among healthcare workers in a Taiwan hospital. J. Antimicrob. Chemother. 2005;56:919–922. doi: 10.1093/jac/dki346.
    1. Zhu Z, et al. Potent cross-reactive neutralization of SARS coronavirus isolates by human monoclonal antibodies. Proc. Natl Acad. Sci. USA. 2007;104:12123–12128. doi: 10.1073/pnas.0701000104.
    1. Wang, C. et al. A human monoclonal antibody blocking SARS-CoV-2 infection. Preprint at bioRxiv10.1101/2020.03.11.987958 (2020).
    1. Tian X, et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg. Microbes Infect. 2020;9:382–385. doi: 10.1080/22221751.2020.1729069.
    1. Li F, Li W, Farzan M, Harrison SC. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 2005;309:1864–1868. doi: 10.1126/science.1116480.
    1. Tai W, et al. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunology. 2020 doi: 10.1038/s41423-020-0400-4.
    1. Li XY, et al. The keypoints in treatment of the critical coronavirus disease 2019 patient. Chin. J. Tuberculosis Respir. Dis. 2020;43:E026.
    1. CNA. Chinese doctors ‘using plasma therapy’ on COVID-19 patients. CNA (2020).
    1. Johnson RF, et al. 3B11-N, a monoclonal antibody against MERS-CoV, reduces lung pathology in rhesus monkeys following intratracheal inoculation of MERS-CoV Jordan-n3/2012. Virology. 2016;490:49–58. doi: 10.1016/j.virol.2016.01.004.
    1. VIR. Press release details: Vir Biotechnology applying multiple platforms to address public health risk from Wuhan coronavirus. VIR (2020).
    1. Cohen, J. Can an anti-HIV combination or other existing drugs outwit the new coronavirus? Science (2020).
    1. Duddu, P. Coronavirus treatment: vaccines/drugs in the pipeline for COVID-19. Clinical Trials Arena (2020).
    1. Berry JD, et al. Neutralizing epitopes of the SARS-CoV S-protein cluster independent of repertoire, antigen structure or mAb technology. MAbs. 2010;2:53–66. doi: 10.4161/mabs.2.1.10788.
    1. Qiu M, et al. Antibody responses to individual proteins of SARS coronavirus and their neutralization activities. Microbes Infect. 2005;7:882–889. doi: 10.1016/j.micinf.2005.02.006.
    1. GISAID. Receptor binding surveillance for high quality genomes. GISAID (2020).
    1. Kleine-Weber H, et al. Mutations in the spike protein of Middle East respiratory syndrome coronavirus transmitted in Korea increase resistance to antibody-mediated neutralization. J. Virol. 2019;93:e01381-18. doi: 10.1128/JVI.01381-18.
    1. Rockx B, et al. Escape from human monoclonal antibody neutralization affects in vitro and in vivo fitness of severe acute respiratory syndrome coronavirus. J. Infect. Dis. 2010;201:946–955. doi: 10.1086/651022.
    1. Yang ZY, et al. Evasion of antibody neutralization in emerging severe acute respiratory syndrome coronaviruses. Proc. Natl. Acad. Sci. USA. 2005;102:797–801. doi: 10.1073/pnas.0409065102.
    1. Ho MS, et al. Neutralizing antibody response and SARS severity. Emerg. Infect. Dis. 2005;11:1730–1737. doi: 10.3201/eid1111.040659.
    1. Tetro JA. Is COVID-19 receiving ADE from other coronaviruses? Microbes Infect. 2020;22:72–73. doi: 10.1016/j.micinf.2020.02.006.
    1. Liu L, et al. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight. 2019 doi: 10.1172/jci.insight.123158.
    1. Zhang L, et al. Antibody responses against SARS coronavirus are correlated with disease outcome of infected individuals. J. Med. Virol. 2006;78:1–8. doi: 10.1002/jmv.20499.
    1. Arabi YM, et al. Feasibility of using convalescent plasma immunotherapy for MERS-CoV infection, Saudi Arabia. Emerg. Infect. Dis. 2016;22:1554. doi: 10.3201/eid2209.151164.
    1. Drosten C, et al. Transmission of MERS-coronavirus in household contacts. N. Engl. J. Med. 2014;371:828–835. doi: 10.1056/NEJMoa1405858.
    1. Park WB, et al. Kinetics of serologic responses to MERS coronavirus infection in humans, South Korea. Emerg. Infect. Dis. 2015;21:2186–2189. doi: 10.3201/eid2112.151421.
    1. Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol. 2008;8:34–47. doi: 10.1038/nri2206.
    1. Bournazos S, DiLillo DJ, Ravetch JV. The role of Fc-FcgammaR interactions in IgG-mediated microbial neutralization. J. Exp. Med. 2015;212:1361–1369. doi: 10.1084/jem.20151267.
    1. Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science. 2006;313:670–673. doi: 10.1126/science.1129594.
    1. Zhang Q, Wang Y, Qi C, Shen L, Li J. Clinical trial analysis of 2019-nCoV therapy registered in China. J. Med. Virol. 2020 doi: 10.1002/jmv.25733.
    1. Shang J, et al. Structural basis of receptor recognition by SARS-CoV-2. Nature. 2020 doi: 10.1038/s41586-020-2179-y.
    1. Prabakaran Ponraj, Gan Jianhua, Feng Yang, Zhu Zhongyu, Choudhry Vidita, Xiao Xiaodong, Ji Xinhua, Dimitrov Dimiter S. Structure of Severe Acute Respiratory Syndrome Coronavirus Receptor-binding Domain Complexed with Neutralizing Antibody. Journal of Biological Chemistry. 2006;281(23):15829–15836. doi: 10.1074/jbc.M600697200.

Source: PubMed

3
Se inscrever