Biomarkers of endothelial dysfunction and outcomes in coronavirus disease 2019 (COVID-19) patients: a systematic review and meta-analysis ========================================================================================================================================= * Andrianto * Makhyan Jibril Al-Farabi * Ricardo Adrian Nugraha * Bagas Adhimurda Marsudi * Yusuf Azmi ## Abstract **Background** Several studies have reported that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can directly infect endothelial cells, and endothelial dysfunction is often found in severe cases of coronavirus disease 2019 (COVID-19). To better understand the pathological mechanisms underlying endothelial dysfunction in COVID-19-associated coagulopathy, we conducted a systematic review and meta-analysis to assess biomarkers of endothelial cells in patients with COVID-19. **Methods** A literature search was conducted on online databases for observational studies evaluating biomarkers of endothelial dysfunction and composite poor outcomes in COVID-19 patients. **Results** A total of 1187 patients from 17 studies were included in this analysis. The estimated pooled means for von Willebrand Factor (VWF) antigen levels in COVID-19 patients was higher compared to healthy control (306.42 [95% confidence interval (CI) 291.37-321.48], p<0.001; I2:86%), with the highest VWF antigen levels was found in deceased COVID-19 patients (448.57 [95% CI 407.20-489.93], p<0.001; I2:0%). Meta-analysis showed that higher plasma levels of VWF antigen, tissue-type plasminogen activator (t-PA), plasminogen activator inhibitor-1 antigen (PAI-1) antigen, and soluble thrombomodulin (sTM) were associated with composite poor outcome in COVID-19 patients ([standardized mean difference (SMD) 0.74 [0.33-1.16], p<0.001; I2:80.4%], [SMD 0.55 [0.19-0.92], p=0.003; I2:6.4%], [SMD 0.33 [0.04-0.62], p=0.025; I2:7.9%], and [SMD 0.55 [0.10-0.99], p=0.015; I2:23.6%], respectively). **Conclusion** The estimated pooled means shows increased levels of VWF antigen in COVID-19 patients. Several biomarkers of endothelial dysfunction, including VFW antigen, t-PA, PAI-1, and sTM, are significantly associated with increased composite poor outcome in patients with COVID-19. **PROSPERO registration number** CRD42021228821 Keywords * Endothelial dysfunction * von Willebrand Factor * tissue-type plasminogen activator * plasminogen activator inhibitor-1 * thrombomodulin * COVID-19 ## Introduction Although initially recognized as a disease affecting the respiratory system, the coronavirus disease 2019 (COVID-19) often manifests as cardiovascular complications such as myocarditis, myocardial injuries, arrhythmias, and venous thromboembolism events (VTE) [1]. There are several possible mechanisms for these phenomena, one of which may be an unrestrained and unbalanced innate immune response, which in turn negates effective adaptive immunity, supporting the progression of COVID-19. Frequent laboratory abnormalities in patients with unfavorable progression of COVID-19 include abnormal cytokine profiles, which led to the initial presumption that the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection involved a cytokine storm [2]. However, recent evidence suggests that increased inflammatory cytokines including interleukin-6 (IL-6) in patients with severe and critical COVID-19 are significantly lower compared to patients with sepsis and acute respiratory distress syndrome (ARDS) not associated with COVID-19, thus doubting the role of a cytokine storm in COVID-19-related multiple organ damage [3]. Several studies have reported that the SARS-CoV-2 can directly infect endothelial cells, and endothelial dysfunction is often found in severe cases of COVID-19 [4]. Autopsy findings have also demonstrated endothelial dysfunction and microvascular thrombosis, together with pulmonary embolism (PE) and deep-vein thrombosis in COVID-19 patients [5]. These findings suggest that endothelial injury, endotheliitis, and microcirculatory dysfunction in different vascular beds contribute significantly to life-threatening complications of COVID-19, such as VTE and multiple organ involvement [6]. To better understand the pathological mechanisms underlying endothelial dysfunction in COVID-19-associated coagulopathy, we conducted a systematic review and meta-analysis to assess biomarkers of endothelial cells in patients with COVID-19. ## Methods ### Search strategy and study selection A systematic literature search of PubMed, PMC Europe, and the Cochrane Library Database from 1 January 2020 to 20 December 2020 was performed using the search strategy outlined in Supplementary Table S1. Additional records were retrieved from Google Scholar. Duplicate articles were removed after the initial search. Two authors (MJA and YA) independently screened the title and abstract of the articles. Articles that passed the screening were assessed in full text based on the eligibility criteria. Disagreements were resolved by discussion with the senior author (A). This study was conducted following the Preferred Reporting Item for Systematic Reviews and Meta-Analysis (PRISMA) statement and registered with the International Prospective Register of Systematic Reviews (PROSPERO) database (registration number CRD42021228821). ### Eligibility criteria We included all observational studies examining biomarkers of endothelial dysfunction and outcomes from patients who tested positive for SARS-CoV-2 using the reverse transcription-polymerase chain reaction (RT-PCR) test. The following types of articles were excluded from the analysis: case reports, review articles, non-English language articles, research articles on the pediatric population, animal or in-vitro studies, unpublished studies, and studies with irrelevant or non-extractable results. ### Data collection process Data extraction was carried out by two authors (MJA and YA) independently using piloted data extraction forms consisting of the author, year of publication, study design, number and characteristics of samples, levels of several biomarkers of endothelial dysfunction, and patient outcomes. The biomarkers of endothelial dysfunction analyzed in this study were von Willebrand Factor (VWF) antigen, tissue-type plasminogen activator (t-PA), plasminogen activator inhibitor-1 antigen (PAI-1), and soluble thrombomodulin (sTM). The primary endpoint was composite poor outcomes consisting of ICU admission, severe illness, worsening of the respiratory status, the need for mechanical ventilation, and mortality. Moreover, if the included studies reported the data using median and quartile values, we used the formula developed by Wan *et al*. to estimate mean and standard deviation [7]. Disease severity was defined based on the WHO R&D Blueprint on COVID-19 [8]. ### Quality assessment The quality and risk of bias assessment of included studies were conducted using the Newcastle-Ottawa score (NOS) [9] by all authors independently, and discrepancies were resolved through discussion. This scoring system consists of three domains: the selection of sample cohorts, comparability of cohorts, and assessment of outcomes (Supplementary Table S2). ### Data analysis Stata software V.14.0 (College Station) was used for meta-analysis, and figure of estimated pooled means were produced using GraphPad Prism 9. We pooled multiple means and standard errors of the same population characteristic from different studies into a single group using the fixed-effect model of meta-analysis algorithm. Pooled effect estimates of the outcomes were reported as standardized means differences (SMD). Fixed-effects and random-effects models were used for pooled analysis with low heterogeneity (I2 statistic <50% or P-value <0.1) and high heterogeneity (I2 statistic> 50% or P-value ≤ 0.1), respectively. Statistical significance was determined with P-value < 0.05. We performed a sensitivity analysis to test the robustness of the pooled effect estimates for VWF antigen levels by excluding each study sequentially and rerunning the meta-analysis. The funnel-plot analysis was used to assess the symmetrical distribution of effect sizes, and the regression-based Egger test was performed to assess publication bias on continuous endpoints. ## Results ### Study characteristics We identified 697 articles from the database search, and 492 articles remained after the duplication was removed. Screening on titles and abstracts excluded 465 articles, and the remaining 27 full-text articles were assessed according to eligibility criteria. As a result, 17 studies [10–26] with a total of 1187 patients were subjected to qualitative analysis and meta-analysis (Figure 1 and Table 1). Quality assessment with NOS showed that 13 studies [10,13– 19,21–24,26] were of good quality with ≥ 7 NOS score and four studies [11,12,20,25] were considered as moderate quality with six NOS score (Supplementary Table S2). View this table: [Table 1.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/T1) Table 1. Characteristics of the included studies. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/26/2021.01.24.21250389/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/F1) Figure 1. PRISMA flowchart. ### Biomarkers of endothelial dysfunction and outcome There was an increase in the VWF antigen levels in COVID-19 patients with different levels for each outcome group of COVID-19 patients. The pooled means plasma levels of VWF antigen in COVID-19 patients treated at the general wards and ICU patients or severely ill patients ([306.42 [95% confidence interval (CI) 291.37-321.48], p<0.001; I2:86%] and [398.56 [95% CI 386.84-410.30], p<0.001; I2:92%], respectively) were higher than healthy controls (103.24 [95% CI 91.31-115.17], p<0.001; I2:0%). Moreover, deceased COVID-19 patients had the highest pooled means of VWF antigen levels (448.57 [95% CI 407.20-489.93], p<0.001; I2:0%) (Figure 2). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/26/2021.01.24.21250389/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/F2) Figure 2. The estimated pooled mean for plasma levels of Von Willebrand Factor antigen in patients with COVID-19. For individual studies, circle markers indicate study means and error bars indicate 95% confidence intervals. Markers are sized proportionately to the weight of the study in the analysis. Estimated pooled means for grouped studies are represented by the square markers. Meta-analysis showed that higher plasma levels of VWF antigen was associated with composite poor outcome (SMD 0.74 [0.33-1.16], p<0.001; I2:80.4%). Patients with poor outcome had significantly a higher level of t-PA and PAI-1 antigen compared to patients with good outcomes ([SMD 0.55 [0.19-0.92], p=0.003; I2:6.4%] and [SMD 0.33 [0.04-0.62], p=0.025; I2:7.9%], respectively). The plasma levels of sTM were found to be higher in COVID-19 patients with poor outcome ([SMD 0.55 [0.10-0.99], p=0.015; I2:23.6%]) (Figure 3). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/26/2021.01.24.21250389/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/F3) Figure 3. Several biomarkers for endothelial dysfunction and the outcome of COVID-19. Patients presenting with a higher plasma levels of (A) von Willebrand Factor (VWF) antigen; (B) tissue-type plasminogen activator (t-PA); (C) plasminogen activator inhibitor-1 antigen (PAI-1) antigen; and (D) soluble thrombomodulin (sTM) have an increased risk of composite poor outcome. We found substantial heterogeneity for VWF antigen analysis (I2:80.4%) and low heterogeneity for t-PA, PAI-1 antigen, and sTM analysis (I2:6.4%, I2:7.9%, and I2:23.6%, respectively). However, subgroup analyses to evaluate potential sources of heterogeneity of VWF levels were not performed due to the small amount of primary data included in the group analysis. The sensitivity analysis of VWF levels after excluding two studies [19,24] at risk of bias decreased the heterogeneity considerably while maintaining the significance of pooled effect estimate (SMD 0.34 [0.17-0.62], p <0.001; I2:9.2%). ### Publication bias The visual assessment of the funnel plot showed an asymmetrical shape for the analysis of the vWF antigen levels, which indicated the possibility of publication bias (Figure 4). This asymmetrical shape was due to the inclusion of the studies by Goshua *et al*. [19] and Mancini *et al*. [24]. However, quantitative analysis using regression-based Egger’s test for the same variable showed no significant result of small-study effects (p=0.063). Regression-based Harbord’s test for other biomarkers and composite poor outcome also showed no significant result of small-study effects. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/26/2021.01.24.21250389/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/F4) Figure 4. Funnel-plot analysis for the analysis of the von Willebrand Factor (VWF) antigen. SMD, standardized mean difference. ## Discussion A single layer of healthy endothelial cells lines the entire vascular system and plays an essential role in maintaining laminar blood flow. This is attributed to its anti-inflammatory properties and non-thrombotic surface with vasodilatory homeostasis [27]. Under normal conditions, the endothelium is impermeable to large molecules, inhibits adhesion of leukocytes, provides anti-inflammatory properties, prevents thrombosis, and promotes vasodilation. Conditions that cause endothelial activation, such as infection, inflammation, or other insults, support proatherogenic mechanisms that promote plaque development by stimulating thrombin formation, coagulation, and deposition of fibrin in blood vessel walls [28]. Endothelial dysfunction is one of the manifestations of COVID-19, leading to various systemic complications through several mechanisms. Preliminary studies showed that SARS-CoV-2 directly infects endothelial cells due to the high expression of the angiotensin-converting enzyme 2 (ACE2) receptor and transmembrane protease serine 2 (TMPRSS2). After binding to SARS-CoV-2, the ACE2 receptor is internalized and downregulated, which causes a reduction of ACE2 expression in the surface of endothelial cells [2]. Inflammation in COVID-19 activates the renin-angiotensin system (RAS) either directly by increasing angiotensin I or indirectly due to the reduction of ACE2 expression on the surface of the endothelial cells. Reduced ACE2 will inhibit the hydrolysis of angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), a vasoactive ligand of the Mas receptor (MasR), which exhibit anti-inflammatory, vasodilatory, and anti-fibrosis effects. Reduced activation of MasR will increase the Ang II type 1 receptor (AT1R) activation, thereby perpetuating the proinflammatory phenotype [29]. In addition, AT1R activation and direct viral infection of endothelial cells can increase reactive oxygen species (ROS) generation via activation of NADPH and activate nuclear factor kappa B, thereby reducing nitric oxide (NO) production [4]. Porcine animal models have recently revealed a vicious self-perpetuating pathway, whereby ROS production increases expression of ET-1, which results in further ROS production. Increased vWF expression has also been shown to increase ET-1 expression, alternatively, knockdown of vWF expression has been shown to decrease AngII-induced-ET-1 production [30]. Decreased ACE2 levels can also promote prothrombotic signaling by limiting the degradation of des-Arg9 bradykinin, thereby increasing the activated bradykinin receptors [31]. Activation of several proinflammatory cytokine receptors such as tumor necrosis factor-alpha (TNF-α) and IL-6 can directly or indirectly interfere with endothelial function. TNF-α can increase hyaluronic acid deposition in the extracellular matrix and cause fluid retention, whereas IL-6 increases vascular permeability and promote the secretion of proinflammatory cytokines by endothelial cells [32]. Subsequently, TNF-α and IL-6 induce the adhesive phenotype of endothelial cells and promote neutrophil infiltration, resulting in multiple histotoxic mediators, including neutrophil extracellular traps (NETs) and ROS to be produced, which eventually leads to endothelial cell injury. Activated endothelial cells stimulate coagulation by expressing fibrinogen, VWF, and P-selectin [32,33]. Activated endothelial by inflammatory stimuli can also induce thrombosis by favoring the expression of antifibrinolytics (e.g., PAI-1) and procoagulants (e.g., tissue factor) over the expression of profibrinolytic mediators (e.g., t-PA) and anticoagulants (e.g., heparin-like molecules and thrombomodulin) [28]. Taken together, these mechanisms contribute to massive platelet binding and formation of fibrin, leading to deposition of blood clots in the microvasculature and systemic thrombosis [2]. However, viral RNA of SARS-CoV-2 is rarely detected in the blood [34], which suggests that rather than direct viral infection of endothelial cells, additional host-dependence factors may contribute to systemic endothelial dysfunction and vasculopathy in COVID-19. Von Willebrand factor (VWF) is a large multidomain adhesive glycoprotein produced by megakaryocytes and endothelial cells. It binds to platelet glycoproteins Ibα, αIIbβ3, and subendothelial collagen to activate platelets and initiate platelet aggregation [35]. Endothelial cell-derived VWF contributes to VWF-dependent atherosclerosis by increasing vascular inflammation and platelet adhesion and [36]. COVID-19 and endothelial activation of VWF are recognized as acute-phase proteins released from endothelial cells in response to inflammation [37], but high levels of VWF, in this case, indicated a suspicion of endothelial disturbance [38]. Expression of VWF and its release from the Weibel-Palade body of endothelial cells may also be stimulated by hypoxia. Hypoxia-induced upregulation of VWF is associated with the presence of thrombus in cardiac and pulmonary vessels that promotes leukocyte recruitment [39]. Since VWF is a significant determinant of platelet adhesion after the vascular injury leading to clot formation, high plasma levels of VWF antigen are an independent risk factor for ischemic stroke and myocardial infarction [40]. Several studies have shown that COVID-19 patients have higher plasma levels of VWF antigen than healthy controls [10,12,18,41]. The pooled means of VWF antigen levels in COVID-19 patients obtained in this study were higher than VWF levels reported in patients with acute coronary syndromes [42,43], acute ischemic stroke [44], and active ulcerative colitis [45]. Meanwhile, similar levels of VWF antigen were reported in patients with disseminated intravascular coagulation [46], severe sepsis, and septic shock not associated with COVID-19 [47,48], thus supporting the role of endothelial dysfunction in COVID-19-induced organ dysfunction. In addition to the VWF antigen, VWF activity has also been shown to increase in COVID-19 patients, thus further explaining the role of endothelial cell injury in COVID-19-associated coagulopathy [12,18,20,22]. The procoagulant state resulting from endothelial activation can also be measured from changes in the balance of tissue plasminogen activator and its endogenous inhibitor. Activated endothelial cells produce increased levels of PAI-1, which inhibits the activity of t-PA and urokinase-type plasminogen activator, causing a shift from the hemostatic balance towards the procoagulant state [49]. Increased levels of PAI-1 produced by endothelial cells and possibly other tissues, such as adipose tissue, are risk factors for atherosclerosis and thrombosis [27]. A study by Nougier *et al*. demonstrated that an impaired balance of coagulation and fibrinolysis in COVID-19 patients with significant hypercoagulability was associated with hypofibrinolysis caused by increased plasma levels of PAI-1 [26]. In COVID-19 patients, plasma levels of PAI-1 were found to be 3.7 times higher than in healthy controls [41]. A study by Kang et al. demonstrated that COVID-19 patients with severe respiratory dysfunction had significantly higher levels of PAI-1 compared to patients with burns, ARDS, and sepsis [50]. In addition to endothelial activation due to proinflammatory cytokines, direct infection by SARS-CoV-2 may cause endotheliitis, which indicates vascular endothelial damage, thereby potentiating t-PA and PAI-1 release [51]. Soluble thrombomodulin (sTM) is a soluble form in the plasma as the result of proteolytic cleavage of the intact thrombomodulin protein from the surface of endothelial cells after endothelial injury and dysfunction, such as inflammation, sepsis, ARDS, and atherosclerosis. Thrombomodulin is a vasculoprotective integral membrane type-1 glycoprotein that plays a vital role in endothelial thromboresistance [52]. The thrombomodulin-mediated binding will activate protein C, which provides anti-inflammatory, antifibrinolytic, and anticoagulant benefits for blood vessel walls [53]. In the hyperinflammatory state, increased sTM levels could be due to direct damage to the endothelial cells [54]. Elevated plasma levels of sTM have also been reported in patients with severe acute respiratory syndrome (SARS), indicating endothelial injury [55]. The plasma levels of sTM might be too low to have an impact on the coagulation process, but the consistent increase in sTM levels during pathologies has been widely considered a biomarker for endothelial dysfunction and vascular risk assessment [52]. Although the relative impact of thrombomodulin release due to endothelial injury in COVID- 19 will require further research, several studies have reported the role of sTM as a prognostic biomarker in COVID-19 patients [16,19]. These findings support the hypothesis of endotheliopathy as an important event in the transition to severity and mortality in COVID-19 patients. ### Impact for clinical practice The elevated biomarkers of endothelial cells, which indicate a manifestation of endothelial dysfunction in COVID-19, might increase the risk of vascular complications, poor outcomes, and death. Plasma levels of VWF antigen, t-PA, PAI-1, and sTM can be used as laboratory markers for vascular risk assessment and predictors of poor outcome in COVID-19. ### Limitation One of the 17 studies included in this meta-analysis was a preprint article. Nevertheless, a thorough assessment has been made to make sure that only sound studies are included. Most of the included studies had a retrospective observational design, and the data were not sufficiently matched or adjusted for confounders. Moreover, our primary endpoints of composite poor outcomes vary widely from ICU admission, severe illness, worsening of the respiratory status, the need for mechanical ventilation to death. Therefore, the results must be cautiously interpreted. ## Conclusion There was an increase in the VWF antigen levels in COVID-19 patients, and the highest levels of VWF antigen were found in deceased COVID-19 patients. Biomarkers of endothelial dysfunction, including VWF antigen, t-PA, PAI-1 antigen, and sTM, are significantly associated with an increased composite poor outcome in patients with COVID-19. ## Data Availability All data generated or analyzed during this study are included in this published article. The corresponding author (A) can be contacted for more information. [https://www.crd.york.ac.uk/prospero/display\_record.php?RecordID=228821](https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=228821) ## Data availability The data used to support the findings of this study are included in the article and supplementary data. ## Funding statement None. ## Ethics approval and consent to participate Not Applicable. ## Consent for publication Not Applicable. ## Availability of data and materials All data generated or analyzed during this study are included in this published article. The corresponding author (A) can be contacted for more information. ## Declaration of competing interest The authors declared no conflict of interest. ## Supplementary data View this table: [Table S1.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/T2) Table S1. Details of the search strategy. View this table: [Table S2.](http://medrxiv.org/content/early/2021/01/26/2021.01.24.21250389/T3) Table S2. Assessment of included studies in the Newcastle-Ottawa Scale (NOS). ## Acknowledgments None. ## Footnotes * Email: andrianto{at}fk.unair.ac.id * Email: m.farabi.17{at}ucl.ac.uk * Email: Ricardo.adrian.nugraha-2019{at}fk.unair.ac.id * Email: bagasmarsudi{at}gmail.com * Email: yusuf.azmi-13{at}fk.unair.ac.id * Received January 24, 2021. * Revision received January 24, 2021. * Accepted January 26, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. [1].Evans PC, Rainger GE, Mason JC, Guzik TJ, Osto E, Stamataki Z, et al. Endothelial dysfunction in COVID-19: a position paper of the ESC Working Group for Atherosclerosis and Vascular Biology, and the ESC Council of Basic Cardiovascular Science. Cardiovasc Res 2020;116:2177–84. [https://doi.org/10.1093/cvr/cvaa230](https://doi.org/10.1093/cvr/cvaa230). 2. [2].Perico L, Benigni A, Casiraghi F, Ng LFP, Renia L, Remuzzi G. Immunity, endothelial injury and complement-induced coagulopathy in COVID-19. Nat Rev Nephrol 2020;17. [https://doi.org/10.1038/s41581-020-00357-4](https://doi.org/10.1038/s41581-020-00357-4). 3. [3].Leisman DE, Ronner L, Pinotti R, Taylor MD, Sinha P, Calfee CS, et al. Cytokine elevation in severe and critical COVID-19: a rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir Med 2020. 4. [4].Nägele MP, Haubner B, Tanner FC, Ruschitzka F, Flammer AJ. Endothelial dysfunction in COVID-19: Current findings and therapeutic implications. Atherosclerosis 2020;314:58–62. [https://doi.org/10.1016/j.atherosclerosis.2020.10.014](https://doi.org/10.1016/j.atherosclerosis.2020.10.014). 5. [5].Gavriilaki E, Anyfanti P, Gavriilaki M, Lazaridis A, Douma S, Gkaliagkousi E. Endothelial Dysfunction in COVID-19: Lessons Learned from Coronaviruses. Curr Hypertens Rep 2020;22. [https://doi.org/10.1007/s11906-020-01078-6](https://doi.org/10.1007/s11906-020-01078-6). 6. [6].Huertas A, Montani D, Savale L, Pichon J, Tu L, Parent F, et al. Endothelial cell dysfunction: A major player in SARS-CoV-2 infection (COVID-19)? Eur Respir J 2020;56. [https://doi.org/10.1183/13993003.01634-2020](https://doi.org/10.1183/13993003.01634-2020). 7. [7].Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol 2014;14:135. [https://doi.org/10.1186/1471-2288-14-135](https://doi.org/10.1186/1471-2288-14-135). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2288-14-135&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25524443&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 8. [8].Organization WH. WHO R&D Blueprint-Novel Coronavirus COVID-19 Therapeutic Trial Synopsis. World Heal Organ [Https://Www.Who.Int/Blueprint/Priority-Diseases/Key-Action/COVID-19\_Treatment\_Trial\_Design\_Master\_Protocol\_synopsis\_Final\_18022020Pdf](https://www.who.int/blueprint/priority-diseases/key-action/COVID-19\_treatment\_trial\_design\_master\_protocol_synopsis_final_18022020Pdf) 2020. 9. [9].Wells GA, Shea B, O’connell D, Peterson J, Welch V, Losos M, et al. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality if Nonrandomized Studies in Meta-Analyses 2015. 10. [10].Cugno M, Meroni PL, Gualtierotti R, Griffini S, Grovetti E, Torri A, et al. Complement activation and endothelial perturbation parallel COVID-19 severity and activity. J Autoimmun 2020:102560. 11. [11].Fan BE, Ng J, Chan SSW, Christopher D, Tso ACY, Ling LM, et al. COVID-19 associated coagulopathy in critically ill patients: A hypercoagulable state demonstrated by parameters of haemostasis and clot waveform analysis. J Thromb Thrombolysis 2020:1–12. 12. [12].Panigada M, Bottino N, Tagliabue P, Grasselli G, Novembrino C, Chantarangkul V, et al. Hypercoagulability of COVID-19 patients in Intensive Care Unit. A Report of Thromboelastography Findings and other Parameters of Hemostasis. J Thromb Haemost 2020. [https://doi.org/10.1111/jth.14850](https://doi.org/10.1111/jth.14850). 13. [13].Pine AB, Meizlish ML, Goshua G, Chang C-H, Zhang H, Bishai J, et al. Circulating Markers of Angiogenesis and Endotheliopathy in COVID-19. MedRxiv Prepr Serv Heal Sci 2020;10:2045894020966547. [https://doi.org/10.1101/2020.06.29.20140376](https://doi.org/10.1101/2020.06.29.20140376). 14. [14].Ranucci M, Sitzia C, Baryshnikova E, Di Dedda U, Cardani R, Martelli F, et al. Covid-19-Associated Coagulopathy: Biomarkers of Thrombin Generation and Fibrinolysis Leading the Outcome. J Clin Med 2020;9. [https://doi.org/10.3390/jcm9113487](https://doi.org/10.3390/jcm9113487). 15. [15].Rauch A, Labreuche J, Lassalle F, Goutay J, Caplan M, Charbonnier L, et al. Coagulation biomarkers are independent predictors of increased oxygen requirements in COVID-19. J Thromb Haemost 2020;18:2942–53. [https://doi.org/10.1111/jth.15067](https://doi.org/10.1111/jth.15067). 16. [16].Rovas A, Osiaevi I, Buscher K, Sackarnd J, Tepasse P-R, Fobker M, et al. Microvascular dysfunction in COVID-19: the MYSTIC study. Angiogenesis 2020:1–13. [https://doi.org/10.1007/s10456-020-09753-7](https://doi.org/10.1007/s10456-020-09753-7). 17. [17].Sweeney JM, Barouqa M, Krause GJ, Gonzalez-Lugo JD, Rahman S, Gil MR. Evidence for secondary thrombotic microangiopathy in COVID-19. MedRxiv Prepr Serv Heal Sci 2020. [https://doi.org/10.1101/2020.10.20.20215608](https://doi.org/10.1101/2020.10.20.20215608). 18. [18].Taus F, Salvagno G, Canè S, Fava C, Mazzaferri F, Carrara E, et al. Platelets Promote Thromboinflammation in SARS-CoV-2 Pneumonia. Arterioscler Thromb Vasc Biol 2020;40:2975–89. [https://doi.org/10.1161/ATVBAHA.120.315175](https://doi.org/10.1161/ATVBAHA.120.315175). 19. [19].Goshua G, Pine AB, Meizlish ML, Chang C-H, Zhang H, Bahel P, et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol 2020;7:e575–82. [https://doi.org/10.1016/S2352-3026(20)30216-7](https://doi.org/10.1016/S2352-3026(20)30216-7). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s2352-3026(20)30216-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 20. [20].Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020;46:1089–98. [https://doi.org/10.1007/s00134-020-06062-x](https://doi.org/10.1007/s00134-020-06062-x). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00134-020-06062-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 21. [21].Henry BM, Benoit SW, de Oliveira MHS, Lippi G, Favaloro EJ, Benoit JL. ADAMTS13 activity to von Willebrand factor antigen ratio predicts acute kidney injury in patients with COVID-19: Evidence of SARS-CoV-2 induced secondary thrombotic microangiopathy. Int J Lab Hematol 2020. [https://doi.org/10.1111/ijlh.13415](https://doi.org/10.1111/ijlh.13415). 22. [22].Hoechter DJ, Becker-Pennrich A, Langrehr J, Bruegel M, Zwissler B, Schaefer S, et al. Higher procoagulatory potential but lower DIC score in COVID-19 ARDS patients compared to non-COVID-19 ARDS patients. Thromb Res 2020;196:186–92. [https://doi.org/10.1016/j.thromres.2020.08.030](https://doi.org/10.1016/j.thromres.2020.08.030). 23. [23].Ladikou EE, Sivaloganathan H, Milne KM, Arter WE, Ramasamy R, Saad R, et al. Von Willebrand factor (vWF): marker of endothelial damage and thrombotic risk in COVID-19? Clin Med 2020;20:e178–82. [https://doi.org/10.7861/clinmed.2020-0346](https://doi.org/10.7861/clinmed.2020-0346). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImNsaW5tZWRpY2luZSI7czo1OiJyZXNpZCI7czo5OiIyMC81L2UxNzgiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wMS8yNi8yMDIxLjAxLjI0LjIxMjUwMzg5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 24. [24].Mancini I, Baronciani L, Artoni A, Colpani P, Biganzoli M, Cozzi G, et al. The ADAMTS13-von Willebrand factor axis in COVID-19 patients. J Thromb Haemost 2020. [https://doi.org/10.1111/jth.15191](https://doi.org/10.1111/jth.15191). 25. [25].Morici N, Bottiroli M, Fumagalli R, Marini C, Cattaneo M. Role of von Willebrand Factor and ADAMTS-13 in the Pathogenesis of Thrombi in SARS-CoV-2 Infection: Time to Rethink. Thromb Haemost 2020;120:1339–42. [https://doi.org/10.1055/s-0040-1713400](https://doi.org/10.1055/s-0040-1713400). 26. [26].Nougier C, Benoit R, Simon M, Desmurs-Clavel H, Marcotte G, Argaud L, et al. Hypofibrinolytic state and high thrombin generation may play a major role in SARS-COV2 associated thrombosis. J Thromb Haemost 2020;18:2215–9. [https://doi.org/10.1111/jth.15016](https://doi.org/10.1111/jth.15016). 27. [27].Wang M, Hao H, Leeper NJ, Zhu L. Thrombotic Regulation From the Endothelial Cell Perspectives. Arterioscler Thromb Vasc Biol 2018;38:e90–5. [https://doi.org/10.1161/ATVBAHA.118.310367](https://doi.org/10.1161/ATVBAHA.118.310367). [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYXR2YmFoYSI7czo1OiJyZXNpZCI7czo4OiIzOC82L2U5MCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAxLzI2LzIwMjEuMDEuMjQuMjEyNTAzODkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 28. [28].Lilly LS. Pathophysiology of heart disease: a collaborative project of medical students and faculty. Lippincott Williams & Wilkins; 2012. 29. [29].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. [https://doi.org/10.1016/j.ejim.2020.04.037](https://doi.org/10.1016/j.ejim.2020.04.037). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ejim.2020.04.037&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32336612&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 30. [30].Dushpanova A, Agostini S, Ciofini E, Cabiati M, Casieri V, Matteucci M, et al. Gene silencing of endothelial von Willebrand Factor attenuates angiotensin II-induced endothelin-1 expression in porcine aortic endothelial cells. Sci Rep 2016;6:30048. 31. [31].Chung MK, Karnik S, Saef J, Bergmann C, Barnard J, Lederman MM, et al. SARS-CoV-2 and ACE2: The biology and clinical data settling the ARB and ACEI controversy. EBioMedicine 2020;58:102907. 32. [32].Teuwen L-A, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat Rev Immunol 2020:1–3. 33. [33].Radermecker C, Detrembleur N, Guiot J, Cavalier E, Henket M, d’Emal C, et al. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J Exp Med 2020;217. 34. [34].Wölfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Müller MA, et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020;581:465–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2196-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 35. [35].Löf A, Müller JP, Brehm MA. A biophysical view on von Willebrand factor activation. J Cell Physiol 2018;233:799–810. [https://doi.org/10.1002/jcp.25887](https://doi.org/10.1002/jcp.25887). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jcp.25887&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28256724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 36. [36].Doddapattar P, Dhanesha N, Chorawala MR, Tinsman C, Jain M, Nayak MK, et al. Endothelial cell-derived von Willebrand factor, but not platelet-derived, promotes atherosclerosis in Apoe-deficient mice 2019;38:319–35. [https://doi.org/10.1161/ATVBAHA.117.309918.Endothelial](https://doi.org/10.1161/ATVBAHA.117.309918.Endothelial). 37. [37].Kawecki C, Lenting PJ, Denis C V. von Willebrand factor and inflammation. J Thromb Haemost 2017;15:1285–94. [https://doi.org/10.1111/jth.13696](https://doi.org/10.1111/jth.13696). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/jth.13696&link_type=DOI) 38. [38].Breakey N, Escher R. D-dimer and mortality in COVID-19: a self-fulfilling prophecy or a pathophysiological clue? Swiss Med Wkly 2020;150:w20293. [https://doi.org/10.4414/smw.2020.20293](https://doi.org/10.4414/smw.2020.20293). 39. [39].Mojiri A, Alavi P, Lorenzana Carrillo MA, Nakhaei-Nejad M, Sergi CM, Thebaud B, et al. Endothelial cells of different organs exhibit heterogeneity in von Willebrand factor expression in response to hypoxia. sAtherosclerosis 2019;282:1–10. [https://doi.org/10.1016/j.atherosclerosis.2019.01.002](https://doi.org/10.1016/j.atherosclerosis.2019.01.002). 40. [40].Andersson HM, Siegerink B, Luken BM, Crawley JTB, Algra A, Lane DA, et al. High VWF, low ADAMTS13, and oral contraceptives increase the risk of ischemic stroke and myocardial infarction in young women. Blood 2012;119:1555–60. [https://doi.org/10.1182/blood-2011-09-380618](https://doi.org/10.1182/blood-2011-09-380618). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMDoiMTE5LzYvMTU1NSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAxLzI2LzIwMjEuMDEuMjQuMjEyNTAzODkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 41. [41].Blasi A, von Meijenfeldt FA, Adelmeijer J, Calvo A, Ibañez C, Perdomo J, et al. In vitro hypercoagulability and ongoing in vivo activation of coagulation and fibrinolysis in COVID-19 patients on anticoagulation. J Thromb Haemost 2020;18:2646–53. [https://doi.org/10.1111/jth.15043](https://doi.org/10.1111/jth.15043). 42. [42].Lee KW, Blann AD, Lip GYH. Plasma markers of endothelial damage/dysfunction, inflammation and thrombogenesis in relation to TIMI risk stratification in acute coronary syndromes. Thromb Haemost 2005;94:1077–83. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16363252&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000233377900028&link_type=ISI) 43. [43].Boos CJ, Balakrishnan B, Blann AD, Lip GYH. The relationship of circulating endothelial cells to plasma indices of endothelial damage/dysfunction and apoptosis in acute coronary syndromes: implications for prognosis. J Thromb Haemost 2008;6:1841–50. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1538-7836.2008.03148.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18761720&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000260138400001&link_type=ISI) 44. [44].Kozuka K, Kohriyama T, Nomura E, Ikeda J, Kajikawa H, Nakamura S. Endothelial markers and adhesion molecules in acute ischemic stroke—sequential change and differences in stroke subtype. Atherosclerosis 2002;161:161–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0021-9150(01)00635-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11882328&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 45. [45].Zezos P, Papaioannou G, Nikolaidis N, Vasiliadis T, Giouleme O, Evgenidis N. Elevated plasma von Willebrand factor levels in patients with active ulcerative colitis reflect endothelial perturbation due to systemic inflammation. World J Gastroenterol 2005;11:7639–45. [https://doi.org/10.3748/wjg.v11.i48.7639](https://doi.org/10.3748/wjg.v11.i48.7639). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16437691&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 46. [46].Habe K, Wada H, Ito-Habe N, Hatada T, Matsumoto T, Ohishi K, et al. Plasma ADAMTS13, von Willebrand factor (VWF) and VWF propeptide profiles in patients with DIC and related diseases. Thromb Res 2012;129:598–602. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.thromres.2011.10.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22070827&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) 47. [47].Kremer Hovinga JA, Zeerleder S, Kessler P, Romani DE Wit T, Van Mourik JA, Hack CE, et al. ADAMTS-13, von Willebrand factor and related parameters in severe sepsis and septic shock. J Thromb Haemost 2007;5:2284–90. [https://doi.org/10.1111/j.1538-7836.2007.02743.x](https://doi.org/10.1111/j.1538-7836.2007.02743.x). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1538-7836.2007.02743.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17764538&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250718300017&link_type=ISI) 48. [48].Fukushima H, Nishio K, Asai H, Watanabe T, Seki T, Matsui H, et al. Ratio of von Willebrand factor propeptide to ADAMTS13 is associated with severity of sepsis. Shock 2013;39:409–14. 49. [49].Shapiro NI, Schuetz P, Yano K, Sorasaki M, Parikh SM, Jones AE, et al. The association of endothelial cell signaling, severity of illness, and organ dysfunction in sepsis. Crit Care 2010;14. [https://doi.org/10.1186/cc9290](https://doi.org/10.1186/cc9290). 50. [50].Kang S, Tanaka T, Inoue H, Ono C, Hashimoto S, Kioi Y, et al. IL-6 trans-signaling induces plasminogen activator inhibitor-1 from vascular endothelial cells in cytokine release syndrome. Proc Natl Acad Sci U S A 2020;117:22351–6. [https://doi.org/10.1073/pnas.2010229117](https://doi.org/10.1073/pnas.2010229117). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTE3LzM2LzIyMzUxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDEvMjYvMjAyMS4wMS4yNC4yMTI1MDM4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 51. [51].Varga Z, Flammer A, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID-19. Ann Oncol 2020. 52. [52].Martin FA, Murphy RP, Cummins PM. Thrombomodulin and the vascular endothelium: insights into functional, regulatory, and therapeutic aspects. Am J Physiol Circ Physiol 2013;304:H1585–97. 53. [53].Conway EM. Thrombomodulin and its role in inflammation. Semin. Immunopathol., vol. 34, Springer; 2012, p. 107–25. 54. [54].Nawroth PP, Häring H-U. Thrombomodulin and coronary heart disease. Lancet (London, England) 1999;353:1722–3. 55. [55].Liu Z-H, Wei R, Wu Y-P, Lisman T, Wang Z-X, Han J-J, et al. Elevated plasma tissue-type plasminogen activator (t-PA) and soluble thrombomodulin in patients suffering from severe acute respiratory syndrome (SARS) as a possible index for prognosis and treatment strategy. Biomed Environ Sci 2005;18:260–4. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16274108&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F26%2F2021.01.24.21250389.atom)