Immunity, endothelial injury and complement-induced coagulopathy in COVID-19

Luca Perico, Ariela Benigni, Federica Casiraghi, Lisa F P Ng, Laurent Renia, Giuseppe Remuzzi, Luca Perico, Ariela Benigni, Federica Casiraghi, Lisa F P Ng, Laurent Renia, Giuseppe Remuzzi

Abstract

In December 2019, a novel coronavirus was isolated from the respiratory epithelium of patients with unexplained pneumonia in Wuhan, China. This pathogen, named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causes a pathogenic condition that has been termed coronavirus disease 2019 (COVID-19) and has reached pandemic proportions. As of 17 September 2020, more than 30 million confirmed SARS-CoV-2 infections have been reported in 204 different countries, claiming more than 1 million lives worldwide. Accumulating evidence suggests that SARS-CoV-2 infection can lead to a variety of clinical conditions, ranging from asymptomatic to life-threatening cases. In the early stages of the disease, most patients experience mild clinical symptoms, including a high fever and dry cough. However, 20% of patients rapidly progress to severe illness characterized by atypical interstitial bilateral pneumonia, acute respiratory distress syndrome and multiorgan dysfunction. Almost 10% of these critically ill patients subsequently die. Insights into the pathogenic mechanisms underlying SARS-CoV-2 infection and COVID-19 progression are emerging and highlight the critical role of the immunological hyper-response - characterized by widespread endothelial damage, complement-induced blood clotting and systemic microangiopathy - in disease exacerbation. These insights may aid the identification of new or existing therapeutic interventions to limit the progression of early disease and treat severe cases.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. SARS-CoV-2 structure, genome composition and…
Fig. 1. SARS-CoV-2 structure, genome composition and life cycle.
a | Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a typical betacoronavirus belonging the Orthocoronavirinae family. Each SARS-CoV-2 virion has a diameter of approximately 100–200 nm. Like other coronaviruses, the SARS-CoV-2 envelope comprises a lipid membrane and three structural components: the spike (S) glycoprotein, the envelope (E) protein and the membrane (M) protein. Within the viral envelope, the nucleocapsid (N) protein holds the positive-sense single-stranded RNA, which is 29,903 bases in length. b | The SARS-CoV-2 genome is composed of ten open reading frames (ORFs). At least two-thirds of the viral genome are contained in ORF1a and ORF1b, which together encode a polyprotein, pp1ab, which is further cleaved into 16 non-structural proteins that are involved in genome transcription and replication. Of these proteins, papain-like protease (PLpro) and 3C-like protease (3CLpro) are encoded by ORF1a, whereas RNA-dependent RNA polymerase (RdRp), helicase (Hel) and exonuclease (ExoN) are encoded by ORF1b. The remaining ORFs encode the structural S glycoproteins, and the E protein, M protein and N protein, as well as several accessory proteins with unknown functions. c | SARS-CoV-2 enters the host cell using the endosomal pathway and the cell surface non-endosomal pathway. In the setting of endosomal entry, the SARS-CoV-2 virion attaches to its target cells by direct binding of the S glycoprotein to the host receptor angiotensin-converting enzyme 2 (ACE2). Upon binding, the transmembrane protease serine 2 (TMPRSS2) cleaves and primes S glycoprotein, leading to the fusion of the viral and cell membranes. In addition to canonical viral entry via the endosomal pathway, non-endosomal entry at the plasma membrane may be an additional infection route for SARS-CoV-2. Within the target cells, SARS-CoV-2 is disassembled to release nucleocapsid and viral RNA into the cytoplasm for translation and replication. Translated viral proteins are then assembled in the endoplasmic reticulum (ER) to form the new virions, which are then released from the Golgi membrane system by exocytosis into the extracellular compartment.
Fig. 2. Pathogenesis and outcomes of COVID-19.
Fig. 2. Pathogenesis and outcomes of COVID-19.
a | Following infection of the lungs, (severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces the death of epithelial cells, in particular, type II pneumocytes, as part of the viral replication cycle. Macrophages and neutrophils elicit a specific innate immune response to eradicate the pathogen and kill virus-infected cells. The increase in pro-inflammatory cytokines within the lung leads to the recruitment of leukocytes, further propagating the local inflammatory response. Among these cytokines, interleukin-2 (IL-2), IL-6, granulocyte-colony-stimulating factor (GCSF), interferon-γ (IFNγ), IFNγ inducible protein 10 (IP-10), monocyte chemoattractant protein 1 and 3 (MCP1 and 3), macrophage inflammatory protein 1α (MIP-1α) and tumour necrosis factor (TNF) stimulate the adaptive immune response. At this stage, infiltration of lymphocytes (CD4+ and CD8+ T cells) and natural killer (NK) cells is required to ensure an optimal defence response against SARS-CoV-2. CD4+ T cells mediate antibody production by B cells and also enhance effector CD8+ T cell and NK responses during viral infections. This orchestrated immune response leads to viral eradication and resolution of the disease. In these patients, coronavirus disease 2019 (COVID-19) seems to manifest as a mild disease with symptoms similar to the common flu that resolve spontaneously. b | For unknown reasons, but possibly individual predisposition or differences in viral loads during primary SARS-CoV-2 infection, some patients experience more severe disease. A maladaptive immune response, characterized by lymphopenia and suppression of CD4+ T cells, is likely accountable for the poor prognosis of these patients. In the absence of robust CD4+ T cell activation, B cells generate a polyclonal antibody response that may be ineffective in neutralizing SARS-CoV-2. Increased numbers of exhausted T cells that express high levels of programmed cell death protein 1 (PD1), suggest decreased proliferation and activity of CD8+ T cells. Similarly, NK cells exhibit increased levels of the inhibitory CD94–NK group 2 member A (NKG2A). Impaired cytotoxic activity results in persistent viral shedding that amplifies macrophage and neutrophil activation, leading to the massive production of cytokines (a process referred to as hypercytokinaemia). In these patients, COVID-19 manifests as a severe disease, consisting of advanced pneumonia and acute respiratory distress syndrome. The generation of excess cytokines and persistent viral infection leads to systemic vascular damage, disseminated intravascular coagulation (DIC) and the failure of vital organs, including the kidney and the heart.
Fig. 3. Effect of SARS-CoV-2 infection on…
Fig. 3. Effect of SARS-CoV-2 infection on endothelial cell function, systemic coagulation and thrombosis.
a | Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly infects endothelial cells owing to their high expression levels of angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). After binding by SARS-CoV-2, ACE2 is internalized, and the lack of ACE2 on endothelial cells favours the progression of inflammatory and thrombotic processes triggered by local angiotensin II (Ang II) hyperactivity. Inhibition of ACE2 by binding of SARS-CoV-2 reduces the ACE2-mediated conversion of Ang II to Ang 1–7, the vasoactive ligand of the MAS receptor. The reduction of MAS receptor activation induces a pro-inflammatory phenotype through increased activation of type 1 angiotensin receptors (AT1R). Additionally, the reduction in levels of ACE2 limits the degradation of des-Arg9 bradykinin (DABK) into inactive peptides, ultimately leading to increased pro-thrombotic signalling via the activation of bradykinin receptors (BKRs). b | SARS-CoV-2 also activates the complement system — an integral component of the innate immune response. The complement cascade can be activated by three different pathways, the classical, the lectin and the alternative pathway, that resolve around the formation of the C3 convertases that cleave C3, generating the pro-inflammatory peptide C3a and large amount of C3b that opsonizes pathogens. C3b also forms the C5 convertase, which leads to release of the potent anaphylatoxin C5a, as well as the fragment C5b, responsible for the formation of the membrane attack complex (MAC) C5b–9 on target cells, which is considered to be the terminal event of complement activation. In addition, complexes of SARS-CoV-2-specific antibodies and viral antigens might induce endothelial cell injury through activation of the C1 complex of the classical pathway and induction of antibody-dependent cytotoxicity (ADC). c | Pro-inflammatory cytokines and chemokines released by activated macrophages amplify the vicious cycle of vascular integrity disruption, vessel coagulation and thrombosis by degrading the endothelial glycocalyx, activating the coagulation system and dampening anticoagulant mechanisms. The adhesive phenotype of endothelial cells induced by inflammatory cytokines and chemokines promotes infiltration of neutrophils, which produce large amounts of histotoxic mediators, including reactive oxygen species (ROS) and neutrophil extracellular traps (NETs), ultimately leading to injury of endothelial cells. d | Activated endothelial cells initiate coagulation by expressing P-selectin, von Willebrand factor (vWf) and fibrinogen, leading to massive platelet binding, fibrin formation and clotting of red blood cells (RBCs), ultimately resulting in systemic thrombosis and disseminated intravascular coagulation.
Fig. 4. The effects of COVID-19 on…
Fig. 4. The effects of COVID-19 on the kidney.
The kidney expresses high levels of angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) and has been identified as a target organ for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by several studies. a | Within the glomerulus, podocytes and endothelial cells have been identified as specific sites for viral infection. SARS-CoV-2-induced podocyte dysfunction is likely to induce impairment of glomerular filtration, leading to proteinuria and haematuria, which are often observed in patients with coronavirus disease 2019 (COVID-19). Infection of endothelial cells alters glomerular capillary haemostasis and induces the formation of fibrin thrombi. b | SARS-CoV-2 has also been found in proximal tubular cells, which exhibit high levels of ACE2 in the apical brush border. SARS-CoV-2 infection leads to loss of the brush border and vacuolar degeneration in tubular epithelial cells, with luminal debris composed of necrotic epithelium with evidence of complement activation and membrane attack complex (MAC) (C5b–9) deposition on tubular cells and massive macrophage infiltration in the kidney interstitium. c | In addition, a non-viral-dependent mechanism might also be accountable for kidney dysfunction in the context of COVID-19, including a possible contribution of the APOL1 ‘risk’ genotype in inducing focal segmental glomerulosclerosis following SARS-CoV-2 infection, as well as the role of haemodynamic factors, cardiac dysfunction, high levels of mechanical ventilation, hypovolaemia, nephrotoxic drug treatments and nosocomial sepsis resulting in COVID-19-associated acute kidney injury (AKI).

References

    1. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat. Med. 2020;6:450–452.
    1. Zhou P, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273.
    1. Shi J, et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science. 2020;368:1016–1020.
    1. Zhang YZ, Holmes EC. A genomic perspective on the origin and emergence of SARS-CoV-2. Cell. 2020;181:223–227.
    1. Zhou H, et al. A novel bat coronavirus closely related to SARS-CoV-2 contains natural insertions at the S1/S2 cleavage site of the spike protein. Curr. Biol. 2020;30:2196–2203.e3.
    1. Zhou Y, et al. Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell Discov. 2020;6:14–18.
    1. Peng Q, et al. Structural and biochemical characterization of nsp12-nsp7-nsp8 core polymerase complex from SARS-CoV-2. Cell Rep. 2020;31:107774.
    1. Kirchdoerfer RN, Ward AB. Structure of the SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-factors. Nat. Commun. 2019;10:2342.
    1. Zumla A, Chan JFW, Azhar EI, Hui DSC, Yuen K-Y. Coronaviruses — drug discovery and therapeutic options. Nat. Rev. Drug Discov. 2016;15:327–347.
    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;181:271–280.e8.
    1. Shang J, et al. Cell entry mechanisms of SARS-CoV-2. Proc. Natl Acad. Sci. USA. 2020;117:11727–11734.
    1. Wan, Y., Shang, J., Graham, R., Baric, R. S. & Li, F. Receptor recognition by the novel coronavirus from Wuhan: an analysis based on decade-long structural studies of SARS coronavirus. J. Virol. 10.1128/JVI.00127-20 (2020).
    1. Jaimes JA, Millet JK, Whittaker GR. Proteolytic cleavage of the SARS-CoV-2 spike protein and the role of the novel S1/S2 site. iScience. 2020;23:101212.
    1. Perico L, Benigni A, Remuzzi G. Should COVID-19 concern nephrologists? Why and to what extent? The emerging impasse of angiotensin blockade. Nephron. 2020;144:213–221.
    1. Li Y, et al. The MERS-CoV receptor DPP4 as a candidate binding target of the SARS-CoV-2 spike. iScience. 2020;23:101160.
    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;12:1–5.
    1. Sungnak W, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 2020;26:681–687.
    1. Ziegler CGK, 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;181:1016–1035.e19.
    1. Wölfel R, et al. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020;581:465–469.
    1. Li Q, et al. Early transmission dynamics in Wuhan, China, of novel coronavirus–infected pneumonia. N. Engl. J. Med. 2020;382:1199–1207.
    1. Hou YJ, et al. SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Cell. 2020;182:429–446.e14.
    1. Shi Y, et al. COVID-19 infection: the perspectives on immune responses. Cell Death Differ. 2020;27:1451–1454.
    1. Cao X. COVID-19: immunopathology and its implications for therapy. Nat.Rev. Immunol. 2020;20:269–270.
    1. Ackermann M, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N. Engl. J. Med. 2020;383:120–128.
    1. Aguiar D, Lobrinus JA, Schibler M, Fracasso T, Lardi C. Inside the lungs of COVID-19 disease. Int. J. Legal Med. 2020;34:1271–1274.
    1. Pernazza A, et al. Early histologic findings of pulmonary SARS-CoV-2 infection detected in a surgical specimen. Virchows Arch. 2020 doi: 10.1007/s00428-020-02829-1.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.
    1. Xu Z, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020;8:420–422.
    1. Fan E, et al. COVID-19-associated acute respiratory distress syndrome: is a different approach to management warranted? Lancet Respir. Med. 2020;8:816–821.
    1. Marini JJ, Gattinoni L. Management of COVID-19 respiratory distress. JAMA. 2020;323:2329–2330.
    1. Patel A, et al. New onset anosmia and ageusia in adult patients diagnosed with SARS-CoV-2. Clin. Microbiol. Infect. 2020;26:1236–1241.
    1. Romero-Sánchez CM, et al. Neurologic manifestations in hospitalized patients with COVID-19: The ALBACOVID registry. Neurology. 2020;95:e1060–e1070.
    1. Dockery DM, Rowe SG, Murphy MA, Krzystolik MG. The ocular manifestations and transmission of COVID-19: recommendations for prevention. J. Emerg. Med. 2020;59:137–140.
    1. Suresh Kumar VC, et al. Novelty in the gut: a systematic review and meta-analysis of the gastrointestinal manifestations of COVID-19. BMJ Open Gastroenterol. 2020;7:e000417.
    1. J J Jin X, et al. Epidemiological, clinical and virological characteristics of 74 cases of coronavirus-infected disease 2019 (COVID-19) with gastrointestinal symptoms. Gut. 2020;69:1002–1009.
    1. Shi S, et al. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5:802–810.
    1. Zang R, et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci. Immunol. 2020;5:eabc3582.
    1. Lamers MM, et al. SARS-CoV-2 productively infects human gut enterocytes. Science. 2020;369:50–54.
    1. Grajewski RS, et al. Letter to the editor: a missing link between SARS-CoV-2 and the eye?: ACE2 expression on the ocular surface. J. Med. Virol. 2020 doi: 10.1002/jmv.26136.
    1. Lucas C, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020;584:463–469.
    1. Giamarellos-Bourboulis EJ, et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020;27:992–1000.e3.
    1. Mathew D, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369:eabc8511.
    1. McKechnie JL, Blish CA. The innate immune system: fighting on the front lines or fanning the flames of COVID-19? Cell Host Microbe. 2020;27:863–869.
    1. Vabret N, et al. Immunology of COVID-19: current state of the science. Immunity. 2020;52:910–941.
    1. Blanco-Melo D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181:1036–1045.e9.
    1. Park A, Iwasaki A. Type I. and type III interferons — induction, signaling, evasion, and application to combat COVID-19. Cell Host Microbe. 2020;27:870–878.
    1. Thoms M, et al. Structural basis for translational shutdown and immune evasion by the Nsp1 protein of SARS-CoV-2. Science. 2020;369:1249–1255.
    1. Jiang H, et al. SARS-CoV-2 Orf9b suppresses type I interferon responses by targeting TOM70. Cell. Mol. Immunol. 2020;17:998–1000.
    1. Vazquez C, Horner SM. MAVS coordination of antiviral innate immunity. J. Virol. 2015;89:6974–6977.
    1. Gordon DE, et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020;583:459–468.
    1. Hadjadj J, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369:718–724.
    1. Trouillet-Assant S, et al. Type I IFN immunoprofiling in COVID-19 patients. J. Allergy Clin. Immunol. 2020;146:206–208.e2.
    1. Mehta P, et al. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033–1034.
    1. Yang Y, et al. Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J. Allergy Clin. Immunol. 2020;146:119–127.e4.
    1. Usmani GN, Woda BA, Newburger PE. Advances in understanding the pathogenesis of HLH. Br. J. Haematol. 2013;161:609–622.
    1. McGonagle D, Sharif K, O’Regan A, Bridgewood C. The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-like disease. Autoimmun. Rev. 2020;19:102537.
    1. Sinha P, Matthay MA, Calfee CS. Is a “cytokine storm” relevant to COVID-19? JAMA Intern. Med. 2020;180:1152–1154.
    1. Pedersen SF, Ho Y-C. SARS-CoV-2: a storm is raging. J. Clin. Invest. 2020;130:2202–2205.
    1. Wang C, et al. Aveolar macrophage activation and cytokine storm in the pathogenesis of severe COVID-19. EBioMedicine. 2020;57:102833.
    1. Liao M, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020;26:842–844.
    1. B Bost P, et al. Host-viral infection maps reveal signatures of severe COVID-19 patients. Cell. 2020;181:1475–1488.e12.
    1. Shen B, et al. Proteomic and metabolomic characterization of COVID-19 patient sera. Cell. 2020;182:59–72.e15.
    1. Giordano, S., Kramer, P., Darley-Usmar, V. M., & White C. R. in Apolipoprotein Mimetics in the Management of Human Disease (eds Anantharamaiah, G., Goldberg, D.)(Adis, Springer International Publishing, 2015).
    1. Li J, et al. Virus-host interactome and proteomic survey of PMBCs from COVID-19 patients reveal potential virulence factors influencing SARS-CoV-2 pathogenesis. Med. 2020 doi: 10.1016/j.medj.2020.07.002.
    1. Z Zhou Z, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020;27:883–890.e2.
    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.
    1. Liu, J. et al. Neutrophil-to-lymphocyte ratio predicts severe illness patients with 2019 novel coronavirus in the early stage. Preprint at medRxiv10.1101/2020.02.10.20021584 (2020).
    1. Carissimo, G. et al. Whole blood immunophenotyping uncovers immature neutrophil-to-VD2 T-cell ratio as an early prognostic marker for severe COVID-19. Preprint at bioRxiv10.1101/2020.06.11.147389 (2020).
    1. Barnes BJ, et al. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J. Exp. Med. 2020;217:e20200652.
    1. Chen G, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Invest. 2020;130:2620–2629.
    1. Yang X, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir. Med. 2020;8:475–481.
    1. Tan L, et al. Validation of predictors of disease severity and outcomes in COVID-19 patients: a descriptive and retrospective study. Med. 2020 doi: 10.1016/j.medj.2020.05.002.
    1. Zheng M, et al. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell. Mol. Immunol. 2020;17:533–535.
    1. Diao B, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19) Front. Immunol. 2020;11:827.
    1. Chen, Y. et al. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes. Preprint at medRxiv10.1101/2020.03.27.20045427 (2020).
    1. Bellesi S, et al. Increased CD95 (Fas) and PD-1 expression in peripheral blood T lymphocytes in COVID-19 patients. Br. J.Haematol. 2020 doi: 10.1111/bjh.17034.
    1. Qin C, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 2020;71:762–768.
    1. Bermejo-Martin JF, et al. Lymphopenic community acquired pneumonia as signature of severe COVID-19 infection. J. Infect. 2020;80:e23–e24.
    1. Bermejo-Martin JF, et al. Lymphopenic community acquired pneumonia (L-CAP), an immunological phenotype associated with higher risk of mortality. EBioMedicine. 2017;24:231–236.
    1. Menéndez R, et al. simultaneous depression of immunological synapse and endothelial injury is associated with organ dysfunction in community-acquired pneumonia. J. Clin. Med. 2019;8:1404.
    1. Grifoni A, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181:1489–1501.e15.
    1. Peng Y, et al. Broad and strong memory CD4 + and CD8 + T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020 doi: 10.1038/s41590-020-0782-6.
    1. Le Bert N, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature. 2020;584:457–462.
    1. Weiskopf D, et al. Phenotype and kinetics of SARS-CoV-2–specific T cells in COVID-19 patients with acute respiratory distress syndrome. Sci. Immunol. 2020;5:eabd2071.
    1. Braun J, et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature. 2020;217:e20200206.
    1. Cassotta A, et al. Deciphering and predicting CD4+ T cell immunodominance of influenza virus hemagglutinin. J. Exp. Med. 2020;217:e20200206.
    1. Mateus J, et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science. 2020;26:845–848.
    1. Long QX, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat. Med. 2020;26:845–848.
    1. Wang Y-T, et al. Spiking pandemic potential: structural and immunological aspects of SARS-CoV-2. Trends Microbiol. 2020;28:605–618.
    1. Yuan M, et al. A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV. Science. 2020;368:630–633.
    1. Wu Y, et al. Identification of human single-domain antibodies against SARS-CoV-2. Cell Host Microbe. 2020;27:891–898.e5.
    1. Chandrashekar A, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369:812–817.
    1. Deng W, et al. Primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques. Science. 2020;369:818–823.
    1. Zost, S. J. et al. Potently neutralizing human antibodies that block SARS-CoV-2 receptor binding and protect animals. Preprint at bioRxiv10.1101/2020.05.22.111005 (2020).
    1. Long Q-X, et al. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat. Med. 2020;26:1200–1204.
    1. Seow, J. et al. Longitudinal evaluation and decline of antibody responses in SARS-CoV-2 infection. Preprint at medRxiv10.1101/2020.07.09.20148429 (2020).
    1. Ibarrondo FJ, et al. Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19. N. Engl. J. Med. 2020;83:1085–1087.
    1. Gudbjartsson DF, et al. Humoral immune response to SARS-CoV-2 in Iceland. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2026116.
    1. Seydoux E, et al. Analysis of a SARS-CoV-2-infected individual reveals development of potent neutralizing antibodies with limited somatic mutation. Immunity. 2020;53:98–105.e5.
    1. Juno JA, et al. Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19. Nat. Med. 2020;26:1428–1434.
    1. Robbiani DF, et al. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature. 2020;584:437–442.
    1. Kuri-Cervantes L, et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 2020;5:eabd7114.
    1. Wilk AJ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020;26:1070–1076.
    1. Woodruff, M. et al. Critically ill SARS-CoV-2 patients display lupus-like hallmarks of extrafollicular B cell activation. Preprint at medRxiv10.1101/2020.04.29.20083717 (2020).
    1. Amrun SN, et al. Linear B-cell epitopes in the spike and nucleocapsid proteins as markers of SARS-CoV-2 exposure and disease severity. EBioMedicine. 2020;58:102911.
    1. Wu, F. et al. Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications. Preprint at medRxiv10.1101/2020.03.30.20047365 (2020).
    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;4:e123158.
    1. Yu H, et al. Distinct features of SARS-CoV-2-specific IgA response in COVID-19 patients. Eur. Respir. J. 2020;56:2001526.
    1. Hotez PJ, Corry DB, Bottazzi ME. COVID-19 vaccine design: the Janus face of immune enhancement. Nat. Rev. Immunol. 2020;20:347–348.
    1. Olson JD. D-dimer: an overview of hemostasis and fibrinolysis, assays, and clinical applications. Adv. Clin. Chem. 2015;69:1–46.
    1. Yau JW, Teoh H, Verma S. Endothelial cell control of thrombosis. BMC Cardiovasc. Disord. 2015;15:130.
    1. Reitsma S, Slaaf DW, Vink H, van Zandvoort MAMJ, oude Egbrink MGA. The endothelial glycocalyx: composition, functions, and visualization. Pflugers Arch. 2007;454:345–359.
    1. Teuwen LA, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat. Rev. Immunol. 2020;20:389–391.
    1. Varga Z, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–1418.
    1. Oxley TJ, et al. Large-vessel stroke as a presenting feature of covid-19 in the Young. N. Engl. J. Med. 2020;382:e60.
    1. Cheng. M. European Doctors Warn Rare Kids’ Syndrome May Have Virus Tie. (Associated Press, 2020).
    1. Bunyavanich S, Do A, Vicencio A. Nasal gene expression of angiotensin-converting Enzyme 2 in children and adults. JAMA. 2020;323:2427–2429.
    1. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–1608.
    1. Diorio C, et al. Multisystem inflammatory syndrome in children and COVID-19 are distinct presentations of SARS-CoV-2. J. Clin. Invest. 2020 doi: 10.1172/JCI140970.
    1. Cheung EW, et al. Multisystem inflammatory syndrome related to COVID-19 in previously healthy children and adolescents in New York City. JAMA. 2020;324:294–296.
    1. Verdoni L, et al. An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet. 2020;395:1771–1778.
    1. Zahra B, et al. Acute heart failure in multisystem inflammatory syndrome in children (MIS-C) in the context of global SARS-CoV-2 pandemic. Circulation. 2020 doi: 10.1161/CIRCULATIONAHA.120.048360.
    1. Monteil V, et al. Inhibition of SARS-CoV-2 Infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181:905–913.e7.
    1. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur. J. Intern. Med. 2020;76:14–20.
    1. Garvin MR, et al. A mechanistic model and therapeutic interventions for COVID-19 involving a RAS-mediated bradykinin storm. eLife. 2020;9:e59177.
    1. Schmaier AH. The contact activation and kallikrein/kinin systems: pathophysiologic and physiologic activities. J. Thromb. Haemost. 2016;14:28–39.
    1. Schmaier AH. A novel antithrombotic mechanism mediated by the receptors of the kallikrein/kinin and renin–angiotensin systems. Front. Med. 2016;3:61.
    1. Stavrou EX, et al. Reduced thrombosis in Klkb1−/− mice is mediated by increased Mas receptor, prostacyclin, Sirt1, and KLF4 and decreased tissue factor. Blood. 2015;125:710–719.
    1. Sodhi CP, et al. Attenuation of pulmonary ACE2 activity impairs inactivation of des-Arg9 bradykinin/BKB1R axis and facilitates LPS-induced neutrophil infiltration. Am. J. Physiol. Lung Cell Mol. Physiol. 2018;314:L17–L31.
    1. Chung MK, et al. SARS-CoV-2 and ACE2: the biology and clinical data settling the ARB and ACEI controversy. EBioMedicine. 2020;58:102907.
    1. Fang C, et al. Angiotensin 1-7 and Mas decrease thrombosis in Bdkrb2−/− mice by increasing NO and prostacyclin to reduce platelet spreading and glycoprotein VI activation. Blood. 2013;121:3023–3032.
    1. Veerdonk FLvande, et al. Outcomes associated with use of a kinin B2 receptor antagonist among patients with COVID-19. JAMA Netw. Open. 2020;3:e2017708–e2017708.
    1. van de Veerdonk FL, et al. Kallikrein-kinin blockade in patients with COVID-19 to prevent acute respiratory distress syndrome. eLife. 2020;9:e57555.
    1. Roche JA, Roche R. A hypothesized role for dysregulated bradykinin signaling in COVID-19 respiratory complications. FASEB J. 2020;34:7265–7269.
    1. Buzhdygan, T. P. et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in vitro models of the human blood–brain barrier. Preprint at bioRxiv10.1101/2020.06.15.150912 (2020).
    1. Martini R. The compelling arguments for the need of microvascular investigation in COVID-19 critical patients. Clin. Hemorheol. Microcirc. 2020;75:27–34.
    1. Noris M, Benigni A, Remuzzi G. The case of complement activation in COVID-19 multiorgan impact. Kidney Int. 2020;98:314–322.
    1. Song WC, FitzGerald GA. COVID-19, microangiopathy, hemostatic activation, and complement. J. Clin. Invest. 2020;130:3950–3953.
    1. Gralinski LE, et al. Complement activation contributes to severe acute respiratory syndrome coronavirus pathogenesis. mBio. 2018;9:e01753–18.
    1. Ip WKE, et al. Mannose-binding lectin in severe acute respiratory syndrome coronavirus infection. J. Infect. Dis. 2005;191:1697–1704.
    1. Jiang Y, et al. Blockade of the C5a-C5aR axis alleviates lung damage in hDPP4-transgenic mice infected with MERS-CoV. Emerg. Microbes Infect. 2018;7:77.
    1. Gao, T. et al. Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. Preprint at medRxiv10.1101/2020.03.29.20041962 (2020).
    1. Yang YH, et al. Autoantibodies against human epithelial cells and endothelial cells after severe acute respiratory syndrome (SARS)-associated coronavirus infection. J. Med. Virol. 2005;77:1–7.
    1. Messner CB, et al. Ultra-high-throughput clinical proteomics reveals classifiers of COVID-19 infection. Cell Syst. 2020;11:11–24.e4.
    1. Risitano AM, et al. Complement as a target in COVID-19? Nat. Rev. Immunol. 2020;20:343–344.
    1. McGonagle D, O’Donnell JS, Sharif K, Emery P, Bridgewood C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020;2:e437–e445.
    1. Fox, S. E. et al. Pulmonary and cardiac pathology in Covid-19: the first autopsy series from New Orleans. Preprint at medRxiv10.1101/2020.04.06.20050575 (2020).
    1. Carsana, L. et al. Pulmonary post-mortem findings in a large series of COVID-19 cases from Northern Italy. Preprint at medRxiv10.1101/2020.04.19.20054262 (2020).
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020;18:844–847.
    1. Danzi GB, Loffi M, Galeazzi G, Gherbesi E. Acute pulmonary embolism and COVID-19 pneumonia: a random association? Eur. Heart J. 2020;41:1858.
    1. Llitjos JF, et al. High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J. Thromb. Haemost. 2020;18:1743–1746.
    1. Zuckier LS, Moadel RM, Haramati LB, Freeman L. Diagnostic evaluation of pulmonary embolism during the COVID-19 pandemic. J. Nucl. Med. 2020;61:630–631.
    1. Beun R, Kusadasi N, Sikma M, Westerink J, Huisman A. Thromboembolic events and apparent heparin resistance in patients infected with SARS-CoV-2. Int. J. Lab. Hematol. 2020;42:19–20.
    1. Levi M, van der Poll T. Coagulation and sepsis. Thromb. Res. 2017;149:38–44.
    1. Radermecker C, et al. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J. Exp. Med. 2020;217:e20201012.
    1. Cheng Y, et al. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. 2020;97:829–838.
    1. Gabarre P, et al. Acute kidney injury in critically ill patients with COVID-19. Intensive Care Med. 2020;46:1339–1348.
    1. Chan L, et al. AKI in hospitalized patients with COVID-19. J. Am. Soc. Nephrol. 2020 doi: 10.1681/ASN.2020050615.
    1. Ali H, et al. Survival rate in acute kidney injury superimposed COVID-19 patients: a systematic review and meta-analysis. Ren. Fail. 2020;42:393–397.
    1. Lim J-H, et al. Fatal outcomes of COVID-19 in patients with severe acute kidney injury. J. Clin. Med. 2020;9:1718.
    1. Division of Nephrology, Columbia University Vagelos College of Physicians Disaster Response to the COVID-19 Pandemic for Patients with Kidney Disease in New York City. J. Am. Soc. Nephrol. 2020;31:1371–1379.
    1. Alberici F, et al. Management of patients on dialysis and with kidney transplantation during the SARS-CoV-2 (COVID-19) pandemic in Brescia, Italy. Kidney Int. Rep. 2020;5:580–585.
    1. Su H, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020;98:219–227.
    1. Magro C, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res. 2020;220:1–13.
    1. Diao, B. et al. Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Preprint at medRxiv10.1101/2020.03.04.20031120 (2020).
    1. Larsen CP, Bourne TD, Wilson JD, Saqqa O, Sharshir M. A. collapsing glomerulopathy in a patient with COVID-19. Kidney Int. Rep. 2020;5:935–939.
    1. Magoon S, et al. COVID-19–related glomerulopathy: a report of 2 cases of collapsing focal segmental glomerulosclerosis. Kidney Med. 2020;2:488–492.
    1. Hepokoski ML, Malhotra A, Singh P, Crotty Alexander LE. Ventilator-induced kidney injury: are novel biomarkers the key to prevention? Nephron. 2018;140:90–93.
    1. Durvasula R, Wellington T, McNamara E, Watnick S. COVID-19 and kidney failure in the acute care setting: our experience from Seattle. Am. J. Kidney Dis. 2020;76:4–6.
    1. Naicker S, et al. The novel coronavirus 2019 epidemic and kidneys. Kidney Int. 2020;97:824–828.
    1. Farkash EA, Wilson AM, Jentzen JM. Ultrastructural evidence for direct renal infection with SARS-CoV-2. J. Am. Soc. Nephrol. 2020;31:1683–1687.
    1. Abbate M, Rottoli D, Gianatti A. COVID-19 attacks the kidney: ultrastructural evidence for the presence of virus in the glomerular epithelium. Nephron. 2020;144:341–342.
    1. Xu D, et al. Identification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: a study based on single-cell transcriptome analysis. Intensive Care Med. 2020;46:1114–1116.
    1. Ye M, et al. Glomerular localization and expression of angiotensin-converting enzyme 2 and angiotensin-converting enzyme: implications for albuminuria in diabetes. J. Am. Soc. Nephrol. 2006;17:3067–3075.
    1. Samavati L, Uhal BD. ACE2, much more than just a receptor for SARS-COV-2. Front. Cell. Infect. Microbiol. 2020;10:317.
    1. Puelles VG, et al. Multiorgan and renal tropism of SARS-CoV-2. N. Engl. J. Med. 2020;383:590–592.
    1. Miller SE, Goldsmith CS. Caution in identifying coronaviruses by electron microscopy. J. Am. Soc. Nephrol. 2020;31:2223–2224.
    1. Wu H, et al. AKI and collapsing glomerulopathy associated with COVID-19 and APOL1 high-risk genotype. J. Am. Soc. Nephrol. 2020;31:1688–1695.
    1. Kudose S, et al. Kidney biopsy findings in patients with COVID-19. J. Am. Soc. Nephrol. 2020;31:1959–1968.
    1. Santoriello D, et al. Postmortem kidney pathology findings in patients with COVID-19. J. Am. Soc. Nephrol. 2020;31:2158–2167.
    1. Sharma P, et al. COVID-19-associated kidney injury: a case series of kidney biopsy findings. J. Am. Soc. Nephrol. 2020;31:1948–1958.
    1. Hanley B, et al. Histopathological findings and viral tropism in UK patients with severe fatal COVID-19: a post-mortem study. Lancet Microbe. 2020 doi: 10.1016/S2666-5247(20)30115-4.
    1. Ma, Y. et al. 2019 novel coronavirus disease in hemodialysis (HD) patients: Report from one HD center in Wuhan, China. Preprint at medRxiv10.1101/2020.02.24.20027201 (2020).
    1. Wang R, et al. COVID-19 in hemodialysis patients: a report of 5 cases. Am. J. Kidney Dis. 2020;76:141–143.
    1. Kikuchi K, et al. COVID-19 in dialysis patients in Japan: current status and guidance on preventive measures. Ther. Apher. Dial. 2020;24:361–365.
    1. Corbett RW, et al. Epidemiology of COVID-19 in an urban dialysis center. J. Am. Soc. Nephrol. 2020;31:1815–1823.
    1. World Health Organization. Draft landscape of COVID-19 candidate vaccines (2020).
    1. Begum J, et al. Challenges and prospects of COVID-19 vaccine development based on the progress made in SARS and MERS vaccine development. Transbound Emerg. Dis. 2020 doi: 10.1111/tbed.13804.
    1. Shannon A, et al. Remdesivir and SARS-CoV-2: Structural requirements at both nsp12 RdRp and nsp14 exonuclease active-sites. Antivir. Res. 2020;178:104793.
    1. Liu Z, et al. Composition and divergence of coronavirus spike proteins and host ACE2 receptors predict potential intermediate hosts of SARS-CoV-2. J. Med. Virol. 2020;92:595–601.
    1. Wang Y, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395:1569–1578.
    1. US National Library of Medicine. (2020).
    1. Gilead. Gilead’s Investigational Antiviral Remdesivir Receives U.S. Food and Drug Administration Emergency Use Authorization for the Treatment of COVID-19. (2020).
    1. Wang Q, et al. Structural basis for RNA replication by the SARS-CoV-2 polymerase. Cell. 2020;82:417–428.e13.
    1. Shen Y, Radhakrishnan ML, Tidor B. Molecular mechanisms and design principles for promiscuous inhibitors to avoid drug resistance: lessons learned from HIV-1 protease inhibition. Proteins. 2015;83:351–372.
    1. Cao B, et al. A trial of lopinavir-ritonavir in adults hospitalized with severe COVID-19. N. Engl. J. Med. 2020;382:1787–1799.
    1. Zhang L, et al. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science. 2020;368:409–412.
    1. Gandolfini I, et al. COVID-19 in kidney transplant recipients. Am. J. Transpl. 2020;20:1941–1943.
    1. Bartiromo M, et al. Threatening drug-drug interaction in a kidney transplant patient with Coronavirus Disease 2019 (COVID-19) Transpl. Infect. Dis. 2020;22:e13286.
    1. Vincent MJ, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol. J. 2005;2:69.
    1. Cavalcanti AB, et al. Hydroxychloroquine with or without azithromycin in mild-to-moderate Covid-19. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2019014.
    1. Boulware DR, et al. A randomized trial of hydroxychloroquine as postexposure prophylaxis for Covid-19. N. Engl. J. Med. 2020;383:517–525.
    1. Magagnoli J, et al. Outcomes of hydroxychloroquine usage in United States veterans hospitalized with Covid-19. Med. 2020 doi: 10.1016/j.medj.2020.06.001.
    1. Borba MGS, et al. Effect of high vs low doses of chloroquine diphosphate as adjunctive therapy for patients hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection: a randomized clinical trial. JAMA Netw. Open. 2020;3:e208857.
    1. Kleynberg RL, Schiller GJ. Secondary hemophagocytic lymphohistiocytosis in adults: an update on diagnosis and therapy. Clin. Adv. Hematol. Oncol. 2012;10:726–732.
    1. Cruz-Topete D, Cidlowski JA. One hormone, two actions: anti- and pro-inflammatory effects of glucocorticoids. Neuroimmunomodulation. 2015;22:20–32.
    1. Russell CD, Millar JE, Baillie JK. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet. 2020;395:473–475.
    1. Wu C, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern. Med. 2020;180:1–11.
    1. Russell B, et al. Associations between immune-suppressive and stimulating drugs and novel COVID-19 — a systematic review of current evidence. Ecancermedicalscience. 2020;14:1022.
    1. Shang L, Zhao J, Hu Y, Du R, Cao B. On the use of corticosteroids for 2019-nCoV pneumonia. Lancet. 2020;395:683–684.
    1. Blum CA, et al. Adjunct prednisone therapy for patients with community-acquired pneumonia: a multicentre, double-blind, randomised, placebo-controlled trial. Lancet. 2015;385:1511–1518.
    1. Torres A, et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: a randomized clinical trial. JAMA. 2015;313:677–686.
    1. Lee KY, Rhim J-W, Kang J-H. Early preemptive immunomodulators (corticosteroids) for severe pneumonia patients infected with SARS-CoV-2. Clin. Exp. Pediatr. 2020;63:117–118.
    1. Fadel R, et al. Early short course corticosteroids in hospitalized patients with COVID-19. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa601.
    1. The WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) working group Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-analysis. JAMA. 2020 doi: 10.1001/jama.2020.17023.
    1. RECOVERY Collaborative Group. Horby P, et al. Dexamethasone in hospitalized patients with covid-19 — preliminary report. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2021436.
    1. Jacob KA, et al. Intraoperative high-dose dexamethasone and severe AKI after cardiac surgery. J. Am. Soc. Nephrol. 2015;26:2947–2951.
    1. Ponticelli C, Locatelli F. Glucocorticoids in the treatment of glomerular diseases: pitfalls and pearls. Clin. J. Am. Soc. Nephrol. 2018;13:815–822.
    1. Dusheiko G. Side effects of alpha interferon in chronic hepatitis C. Hepatology. 1997;26:112S–121S.
    1. Alattar R, et al. Tocilizumab for the treatment of severe coronavirus disease 2019. J. Med. Virol. 2020 doi: 10.1002/jmv.25964.
    1. Guaraldi G, et al. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. Lancet Rheumatol. 2020;2:e474–e484.
    1. US National Library of Medicine. (2020).
    1. Furlow B. COVACTA trial raises questions about tocilizumab’s benefit in COVID-19. Lancet Rheumatol. 2020;2:e592.
    1. Dimopoulos G, et al. Favorable anakinra responses in severe Covid-19 patients with secondary hemophagocytic lymphohistiocytosis. Cell HostMicrobe. 2020;28:117–123.e1.
    1. Ahn E, et al. Role of PD-1 during effector CD8 T cell differentiation. Proc. Natl Acad. Sci. USA. 2018;115:4749–4754.
    1. Jin H-T, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc. Natl Acad. Sci. USA. 2010;107:14733–14738.
    1. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc. Natl Acad. Sci. USA. 2017;114:4993–4998.
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. Wei SC, et al. Combination anti-CTLA-4 plus anti–PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc. Natl Acad. Sci. USA. 2019;116:22699–22709.
    1. Goldstein AL, Goldstein AL. From lab to bedside: emerging clinical applications of thymosin α1. Expert Opin. Biol. Ther. 2009;9:593–608.
    1. Liu Y, et al. Thymosin alpha 1 (Tα1) reduces the mortality of severe COVID-19 by restoration of lymphocytopenia and reversion of exhausted T cells. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa630.
    1. Shi, C. et al. The potential of low molecular weight heparin to mitigate cytokine storm in severe COVID-19 patients: a retrospective clinical study. Clin. Transl Sci.10.1111/cts.12880 (2020).
    1. Stein, B. L. Coagulopathy associated with COVID-19. (NEJM Journal Watch, 2020).
    1. Thachil J, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J. Thromb. Haemost. 2020;8:1023–1026.
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    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.
    1. Ansarin K, et al. Effect of bromhexine on clinical outcomes and mortality in COVID-19 patients: a randomized clinical trial. Bioimpacts. 2020;10:209–215.
    1. Liu X, et al. Potential therapeutic effects of dipyridamole in the severely ill patients with COVID-19. Acta Pharm. Sin. B. 2020;10:1205–1215.
    1. Porfidia A, Pola R. Venous thromboembolism and heparin use in COVID-19 patients: juggling between pragmatic choices, suggestions of medical societies. J. Thromb. Thrombolysis. 2020;50:68–71.
    1. Mastaglio S, et al. The first case of COVID-19 treated with the complement C3 inhibitor AMY-101. Clin. Immunol. 2020;215:108450.
    1. O’Brien KB, Morrison TE, Dundore DY, Heise MT, Schultz-Cherry S. A protective role for complement C3 protein during pandemic 2009 H1N1 and H5N1 influenza A virus infection. PLoS ONE. 2011;6:e17377.
    1. Ricklin D, Mastellos DC, Reis ES, Lambris JD. The renaissance of complement therapeutics. Nat. Rev. Nephrol. 2018;14:26–47.
    1. Keshari RS, et al. Complement C5 inhibition blocks the cytokine storm and consumptive coagulopathy by decreasing lipopolysaccharide (LPS) release in E. coli sepsis. Blood. 2015;126:765–765.
    1. US National Library of Medicine. (2020).
    1. Woodruff TM, Nandakumar KS, Tedesco F. Inhibiting the C5–C5a receptor axis. Mol. Immunol. 2011;48:1631–1642.
    1. Williams AL, et al. C5 inhibition prevents renal failure in a mouse model of lethal C3 glomerulopathy. Kidney Int. 2017;91:1386–1397.
    1. Noris M, Remuzzi G. Terminal complement effectors in atypical hemolytic uremic syndrome: C5a, C5b-9, or a bit of both? Kidney Int. 2019;96:13–15.
    1. Gattinoni L, et al. COVID-19 pneumonia: different respiratory treatments for different phenotypes? Intensive Care Med. 2020;46:1099–1102.
    1. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia: ARDS or not? Crit. Care. 2020;24:154.
    1. Copin M-C, et al. Time to consider histologic pattern of lung injury to treat critically ill patients with COVID-19 infection. Intensive Care Med. 2020;46:1124–1126.
    1. Aloisi, S., Beasley, D., Borter, G., Escritt, T. & Kelland K. Special report: as virus advances, doctors rethink rush to ventilate. Tildes (2020).
    1. Robba C, et al. Distinct phenotypes require distinct respiratory management strategies in severe COVID-19. Respir. Physiol. Neurobiol. 2020;279:103455.
    1. Casadevall A, Pirofski L. The convalescent sera option for containing COVID-19. J. Clin. Invest. 2020;130:1545–1548.
    1. Duan K, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc. Natl Acad. Sci. USA. 2020;117:9490–9496.
    1. Shen C, et al. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020;323:1582–1589.
    1. Ye M, et al. Treatment with convalescent plasma for COVID-19 patients in Wuhan, China. J. Med. Virol. 2020 doi: 10.1002/jmv.25882.
    1. Zeng QL, et al. Effect of convalescent plasma therapy on viral shedding and survival in COVID-19 patients. J. Infect. Dis. 2020;222:38–43.
    1. Li L, et al. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: a randomized clinical trial. JAMA. 2020;324:460–470.
    1. Agarwal, A. et al. Convalescent plasma in the management of moderate COVID-19 in India: An open-label parallel-arm phase II multicentre randomized controlled trial (PLACID Trial). Preprint at medRxiv10.1101/2020.09.03.20187252 (2020).
    1. Chi X, et al. A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science. 2020;369:650–655.
    1. Wec AZ, et al. Broad neutralization of SARS-related viruses by human monoclonal antibodies. Science. 2020;369:731–736.
    1. Hansen J, et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science. 2020;369:1010–1014.
    1. Ju B, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature. 2020;584:115–119.
    1. Pinto D, et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature. 2020;583:290–295.
    1. Wang C, et al. A human monoclonal antibody blocking SARS-CoV-2 infection. Nat. Commun. 2020;11:1–6.
    1. Liu Y, et al. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect. Dis. 2020;20:656–657.
    1. Kirby T. Evidence mounts on the disproportionate effect of COVID-19 on ethnic minorities. Lancet Respir. Med. 2020;8:547–548.

Source: PubMed

3
구독하다