88
views
0
recommends
+1 Recommend
3 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Rational Drug Repurposing: Focus on Lysosomotropism, Targets in Disease Process, Drug Profile, and Pulmonary Tissue Accumulation in SARS-CoV-2 Infection/COVID-19

      discussion

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction The pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been identified as the disease-causing pathogen of Coronavirus disease 2019 (COVID-19). (Pre)clinical research to identify rapidly available small molecules for the treatment of SARS-CoV-2 infections/COVID-19 has focused to date on the approved lysosomotropic antimalarials chloroquine and hydroxychloroquine, the investigational remdesivir (GS-5734, compassionate use), and the anti-inflammatory corticosteroid dexamethasone (COVID-19 Treatment Guidelines Panel, 2020). Lopinavir/ritonavir and other HIV protease inhibitors, however, were discontinued as treatment options in COVID-19 demonstrating no clinical benefit in clinical trials. Despite encouraging results in treating hospitalized patients with COVID-19 requiring supplemental oxygen, mechanical ventilation, or extracorporeal membrane oxygenation (ECMO) with remdesivir and dexamethasone, there is still a lack of active compounds exhibiting pan-coronavirus antiviral activity, tackling or preventing host cell infection, forming syncytia, endotheliitis, or the cytokine release syndrome (CRS)/cytokine storm syndrome in COVID-19. Target-oriented and in particular site of action-oriented drug repurposing of small molecules has the potential to close the gap in prophylaxis and treatment of mild and moderate COVID-19 and to reduce mortality in severe cases. Oxidative Stress, Apoptosis, Multinucleate Syncytia, Host Cell Entry, and Cytokine Storm Syndrome Define Drug Repurposing Targets Oxidative stress (e.g., enhanced ROS levels) has been demonstrated in animal models of SARS (Delgado-Roche and Mesta, 2020) and serves as a possible explanation why SARS-CoV-2 patients with Glucose-6-phosphate dehydrogenase (G6PD) deficiency develop intravascular hemolysis and methemoglobinemia (Palmer et al., 2020). Both, chloroquine and hydroxychloroquine, are supposed to trigger severe drug-induced hemolytic anemia in G6PD-deficient COVID-19 patients (Beauverd et al., 2020; Kuipers et al., 2020). Severe COVID-19 is associated with an atypical diffuse alveolar damage, ending in the acute respiratory distress syndrome (ARDS) (Huang et al., 2020), most likely accompanied by occurrence of syncytia as a result of a direct infection of cells by an infected neighboring cell without releasing a complete virus (Ou et al., 2020). Ceramides, in particular C18-ceramide, are present in (sepsis-induced) cardiac dysfunction (Chung et al., 2017), and are effective in triggering exocytosis in rat PC12 cells (Tang et al., 2007); further they may contribute to SARS-CoV-2-related cell–cell fusion by exocytosis of viral S protein fractions and development of multinucleate syncytia. Non-structural protein nsp2 of SARS-CoV-2 was associated with host cell cell cycle progression, and apoptosis in host cells, suggesting an impact on disrupting the host cell environment (Yoshimoto, 2020) and apoptosis of endothelial cells (Varga et al., 2020). According to current knowledge, cleavage-mediated fusion of viral S protein with host cells can occur either immediately at the cell surface by TMPRSS2 or within the lysosome catalyzed by lysosomal cathepsin L (Belouzard et al., 2012). The lysosomal cathepsin L induced fusion of SARS particles bound to ACE2 with host cells (Millet and Whittaker, 2015) is sensitive to lysosomal pH. Hence both, TMPRSS2 and cathepsin L, display promising targets of prophylaxis and treatment of SARS-CoV-2 infection/COVID-19. In severe COVID-19, SARS-CoV-2 is likely to cause both, pulmonary and systemic inflammation, thus leading to multi-organ dysfunction in high risk populations. Significantly higher concentrations of IL-8, TNFα, and IL-6 in deceased patients (Chen et al., 2020) are suggesting a rapid and severe deterioration during SARS-CoV-2 infection associated with CRS/cytokine storm syndrome (Mehta et al., 2020). Lysosomotropic (Active) Compounds Are Valuable Drug Candidates Lysosomotropism is a biological characteristic of small molecules and always present in addition to intrinsic pharmacological effects. Various well-known approved drugs such as amitriptyline, chlorpromazine, sertraline, and imipramine share lysosomotropic characteristics (Figure 1A) (Kornhuber et al., 2008; Blaess et al., 2018). Regardless of their pharmacological effects, they are accumulating in lysosomes raising the lysosomal pH from 4.5–5 to 6–6.5, beyond the optimum of most of the lysosomal enzymes, including cathepsin L. Since no effects of lysosomotropic aminoglycoside antibiotics on free cathepsin L (Zhou et al., 2016) or other lysosomotropic drugs on lysosomal enzymes such as acid sphingomyelinase exist (Blaess et al., 2018), a selective inhibition is unlikely. FIGURE 1 (A) Variety of approved lysosomotropic compounds for various indications (Kornhuber et al., 2008; Blaess et al., 2018). Achievement of the desired lysosomotropic effect depends on the active compound, the dosage, and accumulation in lysosomes. Unless indicated, maximum daily doses are split into three applications. *Lysosomotropism very likely, but not yet confirmed, lysosomal drug concentration (effect) within the therapeutic margin expected; dosage: #single dose per day; x in vitro anti-SARS‐CoV tested, xx in vitro anti-SARS-CoV and anti-SARS-CoV-2 tested (Vincent et al., 2005; Kornhuber et al., 2008; Dyall et al., 2014; Zhou et al., 2016; Blaess et al., 2018; Liu et al., 2020; Weston et al., 2020). (B) Cellular targets, cellular effects, and effects related effects of lysosomotropic active compounds in SARS-CoV-2 infection/COVID-19 (Vincent et al., 2005; Masters, 2006; Mingo et al., 2015; Zhou et al., 2016; Blaess et al., 2018; Varga et al., 2020; Zhou et al., 2020). Lysosomotropic compounds target in mammalian cells three major targets related to SARS-CoV-2 infection/COVID-19: cathepsin L (1), gene expression of inflammation-relevant genes (2), C16-ceramide and C18-ceramide synthesis, and apoptosis of host cells (3). Addressing targets 1–3 results in various disease process interfering effects supposed to improve SARS-CoV-2 infection/COVID-19 outcome; (°) in viral infection and bacterial superinfection, (°°) only in bacterial superinfection. Lysosomotropic compounds are not limited to mediate inactivation of cathepsin L Figure 1B. Moreover, lysosomotropic compounds are assumed to suppress the CRS/cytokine storm syndrome and to attenuate the transition from mild to severe SARS-CoV-2 infection/COVID-19 (Zhou et al., 2020). Data of the lysosomotropic model compound NB 06 in LPS-induced inflammation in monocytic cells (Blaess et al., 2018) supports the hypothesis. NB 06 affects gene expression of the prominent inflammatory messengers IL1B, IL23A, CCL4, CCL20, and IL6; likewise, it has beneficial effects in (systemic) infections involving bacterial endotoxins by targeting the TLR4 receptor pathway in sepsis. Similarly, desipramine reduces endothelial stress response in systemic inflammation (Chung et al., 2017). Apoptosis of (infected) mammalian cells is characterized by an increase in C16-ceramide (Thomas et al., 1999) and can be blocked via lysosomotropic compounds such as NB 06, chlorpromazine, and imipramine (Blaess et al., 2018). Furthermore, C18-ceramide triggered exocytosis and forming of syncytia is blocked by chlorpromazine as well (Garner et al., 2010). Suitable Drug Profiles and Routes of Administration According to current knowledge, in therapy inhibition of lysosomal pH dependent processes (e.g., cathepsin L dependent viral entry into host cells) can be obtained only through off-label use of lysosomotropic drugs. Systemic application in lysosomotropic drug concentrations and obtaining an efficacious blood level is sometimes accompanied by severe adverse effects and/or (in this case) undesirable (intrinsic) pharmacological effects. Chloroquine was among the first lysosomotropic active compounds exerting antiviral effects on SARS-CoV-2 (Liu et al., 2020) and during SARS-CoV pre- and post-infection conditions (Vincent et al., 2005). Owing to an unfavorable drug profile (G6PD patients, insufficient lysosomotropism, elimination half-life of 45 ± 15 days), a recommendation against (hydroxy)chloroquine, but not against lysosomotropic active compounds in principle was issued (COVID-19 Treatment Guidelines Panel, 2020). Chlorpromazine displayed anti-SARS-CoV-2 effects in vitro (Weston et al., 2020) and protective effects on COVID-19 in patients in a psychiatry hospital (NCT04366739). Consequently, chlorpromazine is rated as a promising candidate in COVID-19/CRS treatment. In case of treatment of people without mental illness, however, a premature termination of treatment due to severe side effects by systemic application of chlorpromazine is extremely likely. This raises the question of how to handle this issue to provide well tolerated lysosomotropic drugs in SARS-CoV-2 infection/COVID-19. Personalized Bench to Bedside Treatment Concept Numerous available approved drugs with lysosomotropic characteristics permit tailor-made therapy. The individual pre-existing conditions are a criterion for the selection and combination of lysosomotropic drugs. For choosing suitable lysosomotropic drugs some issues have to be considered: Tolerable Intrinsic Pharmacology and Drug Profile Various lysosmotropic drugs in Figure 1A demonstrated anti-SARS-CoV(-2) efficacy (Dyall et al., 2014; Zhou et al., 2016; Liu et al., 2020; Weston et al., 2020), offer a more favorable drug profile than the initially investigated chloroquine and hydroxychloroquine. Accumulation In Lysosomes of Pulmonary Tissue Imipramine and chlorpromazine are accumulating in isolated perfused lung tissue and imipramine in alveolar macrophages (Wilson et al., 1982; Macintyre and Cutler, 1988) suggesting that lysosomotropic drug concentrations in pulmonary alveoli and protective effects on SARS-CoV-2 infection of particular drugs are likely. Of the lysosomotropic in vitro anti-SARS-CoV-2 antibiotics teicoplanin, oritavancin, dalbavancin, and telavancin (Zhou et al., 2016), solely teicoplanin and telavancin are in accumulating pulmonary tissue and are expected to be a treatment option. Additional Therapeutic Benefits In Sars-Cov-2 Infection/Covid-19 Beside lysosomotropism certain intrinsic pharmacological effects are advantageously in SARS-CoV-2 infection/COVID-19. The incidence of CRS/cytokine storm syndrome associated with secondary gram-positive bacterial infections is likely to be minimized by using the pulmonary tissue accumulating antibacterials teicoplanin and telavancin or the antifungal itraconazole in systemic mycoses in appropriate systemic drug levels. Choosing A Suitable Route of Administration Systemic application of chlorpromazine (NCT04366739) and fluoxetine (NCT04377308) as lysosomotropic drugs may provoke severe and unfavorable adverse effects in mental healthy patients. Since the respiratory tract is both, the gateway for SARS-CoV-2 infection/COVID-19 and an internal surface, the expedient is a local application in the airways and/or the respiratory tract. Local application of small molecules is possible, preferably as inhalant or via nebulizers to avoid (undesirable) systemic effects. The majority of lysosomotropic drugs should be suitable for inhalation. Combination With Antivirals and Tmprss2 Inhibitors COVID-19 originates from a SARS-CoV-2 infection that could not be tackled successfully by the immune system. The antiviral remdesivir proved to be effective in infection prophylaxis (phase 0) (de Wit et al., 2020) and viral (SARS-CoV-2) infection (phase 1) within a limited period (5–6 days), shortly after the symptoms emerge and viral shedding occurs (Mitjà and Clotet, 2020). In severe COVID-19 neither a lower mortality nor a faster clearance of viruses was observed (Wang et al., 2020). As soon as the infection initiates a CRS/cytokine storm, it is likely that the transition toward COVID-19 (phase 2), a disseminated intravascular coagulation/thrombotic microangiopathy, or a bacterial secondary infection occurs. An effective multi-drug therapy, focusing on the progression of COVID-19 and emerging severe complications, can be implemented by lysosomotropic drugs, TMPRSS2 inhibitors and antivirals. Nafamostat: An In Vivo TMPRSS2 Inhibitor? Nafamostat is an approved protease inhibitor that inhibits TMPRSS2 (in vitro) (Hoffmann et al., 2020), prevents (sepsis-related) disseminated intravascular coagulation, and thrombotic microangiopathy (Okajima et al., 1995; Levi and Thachil, 2020), appears to be useful in SARS-CoV-2 infection and prophylaxis, and for patients subjected to extracorporeal circulation such as ECMO (Han et al., 2011). It is doubtful, however, whether the pulmonary concentration in therapeutically dosage (Ono Pharmaceuticals, 2020) is sufficient to generate a TMPRSS2 inhibition in vivo as demonstrated in vitro due to poor accumulation in pulmonary tissue (Midgley et al., 1994). Single or Multi Target Approach: Lysosomotropic Drugs vs. Antibodies Various clinical trials are currently under way using immunomodulatory IL-1 and IL-6 inhibitors or anti-IL-6R antibodies (anakinra, tocilizumab, siltuximab, and sarilumab) in patients with COVID-19 (COVID-19 Treatment Guidelines Panel, 2020); limited data, however, is yet available. In a retrospective study using tocilizumab and hydroxychloroquine, both demonstrated a limited benefit in survival (Ip et al., 2020). Tocilizumab shortens mechanical ventilation and hospital stay in severe COVID-19 (Eimer et al., 2020), while tocilizumab is often accompanied by bacterial pneumonia 2 days after application (23%) (Pettit et al., 2020). To improve outcome, antibody cocktails consisting of anti-IL-6, IL-1 receptor blocker, IL-1 type 1 receptor, and TNF-α are suggested (Harrison, 2020), irrespective of the risk of serious adverse effects (e.g., bacterial pneumonia) due to more pronounced interference with the immune defense. Such cocktails are intended to tackle the release of pro-inflammatory cytokines IL-1β and IL-6 mediating lung and tissue inflammation, fever, and fibrosis, as they are supposed to be responsible for the emergence of COVID-19. Although lysosomotropic drugs likewise interfere with the immune defense, such adverse effects are not reported. In contrast to antibodies, however, only the resynthesis of IL-6 and thus the available amount is reduced, but not completely obstructed, still allowing a moderate immune response. Multitargeting on core processes of the viral infection addressing the formation of multinucleate syncytia and alteration of tissue structure, ceramide metabolism, and the release of virions could be a key advantage of lysosomotropic drugs compared to current strategies. Future Directions Daunting results of (hydroxy)chloroquine in clinical trials are closely related to their drug profile and minor lysosomotropism, but not to the mode of action (lysosomotropism) in general. Observations in patients treated with chlorpromazine and the extensive accumulation of imipramine in alveolar macrophages and of both, imipramine and chlorpromazine in isolated perfused lung tissue supports the benefits of lysosomotropic drugs that are accumulating in pulmonary tissue in SARS-CoV-2 infection/COVID-19. Promising candidates among lysosomotropic drugs in fact require more than adequate lysosomotropism; accumulation in pulmonary tissue is a prerequisite as well. It is, however, likely irrelevant whether the drug or its metabolite(s) is accumulating given the broad structural requirements for this activity. Since a large number of compounds has not yet been evaluated for lysosomotropism, many compounds beside those listed in Figure 1A are expected to meet the requirements described here and may (partially) be responsible for background immunity to SARS-CoV infection.

          Related collections

          Most cited references39

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China

          Summary Background A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by WHO and the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0–58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0–13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found

            COVID-19: consider cytokine storm syndromes and immunosuppression

            As of March 12, 2020, coronavirus disease 2019 (COVID-19) has been confirmed in 125 048 people worldwide, carrying a mortality of approximately 3·7%, 1 compared with a mortality rate of less than 1% from influenza. There is an urgent need for effective treatment. Current focus has been on the development of novel therapeutics, including antivirals and vaccines. Accumulating evidence suggests that a subgroup of patients with severe COVID-19 might have a cytokine storm syndrome. We recommend identification and treatment of hyperinflammation using existing, approved therapies with proven safety profiles to address the immediate need to reduce the rising mortality. Current management of COVID-19 is supportive, and respiratory failure from acute respiratory distress syndrome (ARDS) is the leading cause of mortality. 2 Secondary haemophagocytic lymphohistiocytosis (sHLH) is an under-recognised, hyperinflammatory syndrome characterised by a fulminant and fatal hypercytokinaemia with multiorgan failure. In adults, sHLH is most commonly triggered by viral infections 3 and occurs in 3·7–4·3% of sepsis cases. 4 Cardinal features of sHLH include unremitting fever, cytopenias, and hyperferritinaemia; pulmonary involvement (including ARDS) occurs in approximately 50% of patients. 5 A cytokine profile resembling sHLH is associated with COVID-19 disease severity, characterised by increased interleukin (IL)-2, IL-7, granulocyte-colony stimulating factor, interferon-γ inducible protein 10, monocyte chemoattractant protein 1, macrophage inflammatory protein 1-α, and tumour necrosis factor-α. 6 Predictors of fatality from a recent retrospective, multicentre study of 150 confirmed COVID-19 cases in Wuhan, China, included elevated ferritin (mean 1297·6 ng/ml in non-survivors vs 614·0 ng/ml in survivors; p 39·4°C 49 Organomegaly None 0 Hepatomegaly or splenomegaly 23 Hepatomegaly and splenomegaly 38 Number of cytopenias * One lineage 0 Two lineages 24 Three lineages 34 Triglycerides (mmol/L) 4·0 mmol/L 64 Fibrinogen (g/L) >2·5 g/L 0 ≤2·5 g/L 30 Ferritin ng/ml 6000 ng/ml 50 Serum aspartate aminotransferase <30 IU/L 0 ≥30 IU/L 19 Haemophagocytosis on bone marrow aspirate No 0 Yes 35 Known immunosuppression † No 0 Yes 18 The Hscore 11 generates a probability for the presence of secondary HLH. HScores greater than 169 are 93% sensitive and 86% specific for HLH. Note that bone marrow haemophagocytosis is not mandatory for a diagnosis of HLH. HScores can be calculated using an online HScore calculator. 11 HLH=haemophagocytic lymphohistiocytosis. * Defined as either haemoglobin concentration of 9·2 g/dL or less (≤5·71 mmol/L), a white blood cell count of 5000 white blood cells per mm3 or less, or platelet count of 110 000 platelets per mm3 or less, or all of these criteria combined. † HIV positive or receiving longterm immunosuppressive therapy (ie, glucocorticoids, cyclosporine, azathioprine).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Endothelial cell infection and endotheliitis in COVID-19

              Cardiovascular complications are rapidly emerging as a key threat in coronavirus disease 2019 (COVID-19) in addition to respiratory disease. The mechanisms underlying the disproportionate effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on patients with cardiovascular comorbidities, however, remain incompletely understood.1, 2 SARS-CoV-2 infects the host using the angiotensin converting enzyme 2 (ACE2) receptor, which is expressed in several organs, including the lung, heart, kidney, and intestine. ACE2 receptors are also expressed by endothelial cells. 3 Whether vascular derangements in COVID-19 are due to endothelial cell involvement by the virus is currently unknown. Intriguingly, SARS-CoV-2 can directly infect engineered human blood vessel organoids in vitro. 4 Here we demonstrate endothelial cell involvement across vascular beds of different organs in a series of patients with COVID-19 (further case details are provided in the appendix). Patient 1 was a male renal transplant recipient, aged 71 years, with coronary artery disease and arterial hypertension. The patient's condition deteriorated following COVID-19 diagnosis, and he required mechanical ventilation. Multisystem organ failure occurred, and the patient died on day 8. Post-mortem analysis of the transplanted kidney by electron microscopy revealed viral inclusion structures in endothelial cells (figure A, B ). In histological analyses, we found an accumulation of inflammatory cells associated with endothelium, as well as apoptotic bodies, in the heart, the small bowel (figure C) and lung (figure D). An accumulation of mononuclear cells was found in the lung, and most small lung vessels appeared congested. Figure Pathology of endothelial cell dysfunction in COVID-19 (A, B) Electron microscopy of kidney tissue shows viral inclusion bodies in a peritubular space and viral particles in endothelial cells of the glomerular capillary loops. Aggregates of viral particles (arrow) appear with dense circular surface and lucid centre. The asterisk in panel B marks peritubular space consistent with capillary containing viral particles. The inset in panel B shows the glomerular basement membrane with endothelial cell and a viral particle (arrow; about 150 nm in diameter). (C) Small bowel resection specimen of patient 3, stained with haematoxylin and eosin. Arrows point to dominant mononuclear cell infiltrates within the intima along the lumen of many vessels. The inset of panel C shows an immunohistochemical staining of caspase 3 in small bowel specimens from serial section of tissue described in panel D. Staining patterns were consistent with apoptosis of endothelial cells and mononuclear cells observed in the haematoxylin-eosin-stained sections, indicating that apoptosis is induced in a substantial proportion of these cells. (D) Post-mortem lung specimen stained with haematoxylin and eosin showed thickened lung septa, including a large arterial vessel with mononuclear and neutrophilic infiltration (arrow in upper inset). The lower inset shows an immunohistochemical staining of caspase 3 on the same lung specimen; these staining patterns were consistent with apoptosis of endothelial cells and mononuclear cells observed in the haematoxylin-eosin-stained sections. COVID-19=coronavirus disease 2019. Patient 2 was a woman, aged 58 years, with diabetes, arterial hypertension, and obesity. She developed progressive respiratory failure due to COVID-19 and subsequently developed multi-organ failure and needed renal replacement therapy. On day 16, mesenteric ischaemia prompted removal of necrotic small intestine. Circulatory failure occurred in the setting of right heart failure consequent to an ST-segment elevation myocardial infarction, and cardiac arrest resulted in death. Post-mortem histology revealed lymphocytic endotheliitis in lung, heart, kidney, and liver as well as liver cell necrosis. We found histological evidence of myocardial infarction but no sign of lymphocytic myocarditis. Histology of the small intestine showed endotheliitis (endothelialitis) of the submucosal vessels. Patient 3 was a man, aged 69 years, with hypertension who developed respiratory failure as a result of COVID-19 and required mechanical ventilation. Echocardiography showed reduced left ventricular ejection fraction. Circulatory collapse ensued with mesenteric ischaemia, and small intestine resection was performed, but the patient survived. Histology of the small intestine resection revealed prominent endotheliitis of the submucosal vessels and apoptotic bodies (figure C). We found evidence of direct viral infection of the endothelial cell and diffuse endothelial inflammation. Although the virus uses ACE2 receptor expressed by pneumocytes in the epithelial alveolar lining to infect the host, thereby causing lung injury, the ACE2 receptor is also widely expressed on endothelial cells, which traverse multiple organs. 3 Recruitment of immune cells, either by direct viral infection of the endothelium or immune-mediated, can result in widespread endothelial dysfunction associated with apoptosis (figure D). The vascular endothelium is an active paracrine, endocrine, and autocrine organ that is indispensable for the regulation of vascular tone and the maintenance of vascular homoeostasis. 5 Endothelial dysfunction is a principal determinant of microvascular dysfunction by shifting the vascular equilibrium towards more vasoconstriction with subsequent organ ischaemia, inflammation with associated tissue oedema, and a pro-coagulant state. 6 Our findings show the presence of viral elements within endothelial cells and an accumulation of inflammatory cells, with evidence of endothelial and inflammatory cell death. These findings suggest that SARS-CoV-2 infection facilitates the induction of endotheliitis in several organs as a direct consequence of viral involvement (as noted with presence of viral bodies) and of the host inflammatory response. In addition, induction of apoptosis and pyroptosis might have an important role in endothelial cell injury in patients with COVID-19. COVID-19-endotheliitis could explain the systemic impaired microcirculatory function in different vascular beds and their clinical sequelae in patients with COVID-19. This hypothesis provides a rationale for therapies to stabilise the endothelium while tackling viral replication, particularly with anti-inflammatory anti-cytokine drugs, ACE inhibitors, and statins.7, 8, 9, 10, 11 This strategy could be particularly relevant for vulnerable patients with pre-existing endothelial dysfunction, which is associated with male sex, smoking, hypertension, diabetes, obesity, and established cardiovascular disease, all of which are associated with adverse outcomes in COVID-19.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                20 November 2020
                2020
                20 November 2020
                : 11
                : 584881
                Affiliations
                [ 1 ]Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Villingen-Schwenningen, Germany
                [ 2 ]Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
                [ 3 ]Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
                [ 4 ]Department of Organic Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
                [ 5 ]EXIM Department, Fraunhofer Institute IZI Leipzig, Rostock, Germany
                [ 6 ]Faculty of Science, Tuebingen University, Tübingen, Germany
                Author notes

                Edited by: Rafael Maldonado, Pompeu Fabra University, Spain

                Reviewed by: Nadezhda A. German, Texas Tech University Health Sciences Center, United States

                *Correspondence: Hans-Peter Deigner, Hans-Peter.Deigner@ 123456hs-furtwangen.de

                This article was submitted to Experimental Pharmacology and Drug Discovery, a section of the journal Frontiers in Pharmacology

                Article
                584881
                10.3389/fphar.2020.584881
                7938238
                c7a18bd7-546f-4b71-8f16-5396d07bd406
                Copyright © 2020 Blaess, Kaiser, Sommerfeld, Rentschler, Csuk and Deigner.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 18 July 2020
                : 06 October 2020
                Page count
                Pages: 0
                Categories
                Pharmacology
                Opinion

                Pharmacology & Pharmaceutical medicine
                sars-cov-2,covid-19,lysosomotropic compounds,approved active compounds,cytokine storm syndrome,lysosomotropism,repurposing approved drugs,lysosome

                Comments

                Comment on this article