key: cord-0852833-qd5pzxtx authors: Grimes, Joseph M.; Grimes, Kevin V. title: p38 MAPK inhibition: A promising therapeutic approach for COVID-19 date: 2020-05-16 journal: J Mol Cell Cardiol DOI: 10.1016/j.yjmcc.2020.05.007 sha: 89e4efea500c7f98cd67b0ff34d416b9d49b6459 doc_id: 852833 cord_uid: qd5pzxtx COVID-19, caused by the SARS-CoV-2 virus, is a major source of morbidity and mortality due to its inflammatory effects in the lungs and heart. The p38 MAPK pathway plays a crucial role in the release of pro-inflammatory cytokines such as IL-6 and has been implicated in acute lung injury and myocardial dysfunction. The overwhelming inflammatory response in COVID-19 infection may be caused by disproportionately upregulated p38 activity, explained by two mechanisms. First, angiotensin-converting enzyme 2 (ACE2) activity is lost during SARS-CoV-2 viral entry. ACE2 is highly expressed in the lungs and heart and converts Angiotensin II into Angiotensin 1–7. Angiotensin II signals proinflammatory, pro-vasoconstrictive, pro-thrombotic activity through p38 MAPK activation, which is countered by Angiotensin 1–7 downregulation of p38 activity. Loss of ACE2 upon viral entry may tip the balance towards destructive p38 signaling through Angiotensin II. Second, SARS-CoV was previously shown to directly upregulate p38 activity via a viral protein, similar to other RNA respiratory viruses that may hijack p38 activity to promote replication. Given the homology between SARS-CoV and SARS-CoV-2, the latter may employ a similar mechanism. Thus, SARS-CoV-2 may induce overwhelming inflammation by directly activating p38 and downregulating a key inhibitory pathway, while simultaneously taking advantage of p38 activity to replicate. Therapeutic inhibition of p38 could therefore attenuate COVID-19 infection. Interestingly, a prior preclinical study showed protective effects of p38 inhibition in a SARS-CoV mouse model. A number of p38 inhibitors are in the clinical stage and should be considered for clinical trials in serious COVID-19 infection. COVID-19, caused by the SARS-CoV-2 virus, is a major source of morbidity and mortality due to its inflammatory effects in the lungs and heart. The p38 MAPK pathway plays a crucial role in the release of pro-inflammatory cytokines such as IL-6 and has been implicated in acute lung injury and myocardial dysfunction. The overwhelming inflammatory response in COVID-19 infection may be due to caused by disproportionately upregulated p38 activity, explained by two mechanisms. First, angiotensin-converting enzyme 2 (ACE2) activity is lost during SARS-CoV-2 viral entry. ACE2 is highly expressed in the lungs and heart and converts Angiotensin II into Angiotensin 1-7. Angiotensin II signals proinflammatory, pro-vasoconstrictive, pro-thrombotic activity through p38 MAPK activation, which is countered by Angiotensin 1-7 downregulation of p38 activity. Loss of ACE2 during upon viral entry may tip the balance towards destructive p38 signaling through Angiotensin II. Second, SARS-CoV was previously shown to directly upregulate p38 activity via a viral protein, similar to other RNA respiratory viruses that may hijack p38 activity to promote replication. Given the homology between SARS-CoV and SARS-CoV-2, the latter may employ a similar mechanism. Thus, SARS-CoV-2 may induce overwhelming inflammation by directly activating p38 and downregulating a key inhibitory pathway, while simultaneously taking advantage of p38 activity to replicate. Therapeutic inhibition of p38 could therefore attenuate COVID-19 infection. Interestingly, a prior preclinical study showed protective effects of p38 inhibition in a SARS-CoV mouse model. A number of p38 inhibitors are in the clinical stage and should be considered for clinical trials in serious The COVID-19 pandemic, caused by the SARS-CoV-2 coronavirus, has caused resulted in substantial ICU admissions and excess mortality worldwide. Similar to the SARS-CoV virus implicated in the 2003 SARS outbreak, SARS-CoV-2 facilitates cell entry by attaching to angiotensin converting enzyme 2 (ACE2) located on the cell surface [1] . ACE2 is present in multiple tissues and is highly expressed in the lungs and heart [2] . Given this distribution, it is unsurprising that Acute Respiratory Distress Syndrome (ARDS) and myocarditis are the primary causes of death in COVID-19 patients [3] . The cause of overwhelming inflammation induced by SARS-CoV-2 is unclear, however. One particular pathway that has been previously implicated in animal models of acute lung injury and myocardial injury is the p38 MAPK system [4, 5] . Upregulation of the p38 MAPK pathway activates pro-inflammatory cytokines such as IL-6, TNF-α and IL-1β [6] . Herein, we argue that the inflammatory injury provoked by SARS-CoV-2 may be due to a disproportionate upregulation of p38 MAPK activity. This inflammation may be attenuated by p38 small molecule inhibitors, many of which have been safely studied in clinical trials for other indications. Like SARS-CoV, SARS-CoV-2 binds and downregulates ACE2 upon cell entry [7] . ACE2 converts Angiotensin II (Ang II) into Angiotensin 1-7 (Ang 1-7), which then binds to the Mas receptor to counterbalance the vasoconstrictive and pro-inflammatory effects of Ang II [8, 9] . Ang II mediates its effects through p38 MAPK activation [10] . Ang 1-7 stimulation of the Mas receptor decreases p38 MAPK activation to attenuate inflammation [11] . The loss of ACE2 activity upon viral entry may therefore allow Ang II mediated activation of p38 to predominate in the lungs and heart as Ang 1-7 is downregulated. This allows unchecked inflammation and J o u r n a l P r e -p r o o f Journal Pre-proof produces a positive feedback loop as p38 activation also upregulates ADAM17, a protease known to cleave the ACE2 ectodomain to further reduce local ACE2 protective activity [12] . It is worth noting that while the role of the renin-angiotensin system (RAS) is still being fully elucidated in the pathogenesis of SARS-CoV-2, a recent study found that inhibition of RAS via angiotensin-converting enzyme inhibitors or angiotensin receptor blockers may reduce all-cause mortality in COVID-19 patients [13] . In another study of COVID-19 patients, Ang II levels were linearly associated with degree of lung injury and viral load, further implicating RAS imbalance in the COVID-19 pathogenesis [14] . Furthermore, the previous SARS-CoV virus was shown to express a protein that directly upregulates p38 MAPK in vitro, a pathogenic step thought to be utilized in the lifecycle of many RNA respiratory viruses, including influenza strains associated with severe inflammatory responses such as H5N1 [15, 16] . It has been suggested that, among other functions, viral p38 MAPK activation induces endocytosis of viral receptors to facilitate cell entry [17] . Interestingly, p38 MAPK activation has been implicated in the endocytosis of ACE2 [18] . Given its homology to SARS-CoV, SARS-CoV-2 could use a similar mechanism to directly upregulate p38 MAPK. If this is the case, then SARS-CoV-2 could be propagating severe inflammation and organ damage both by both directly upregulating p38 MAPK activity and by downregulating a key p38 MAPK shut off mechanism by reducing ACE2 activity. Furthermore, p38 MAPK activation could simultaneously be propagating the SARS-CoV-2 viral lifecycle. Interestingly, a small preclinical study with a p38 inhibitor in SARS-CoV infected mice showed 80% survival in the treatment group (N=5) and 0% in the control (N=5) [19] . Importantly, overactivation of p38 MAPK activity can also explain additional clinical findings in patients of with SARS-CoV-2 infection. There are reports of significant thrombotic J o u r n a l P r e -p r o o f events in severely ill COVID-19 patients, and a recent study in COVID-19 patients notes that the vascular endothelium is directly infected in target organs of SARS-CoV-2 [20] [21] [22] . Elevated p38 MAPK activity in the endothelium has been implicated in platelet aggregation, and arterial thrombosis, and apoptosis of endothelial cells, a clinical finding also shown in the histology of the aforementioned study [23] . Cardiac dysfunction in COVID-19 patients could partly be due to this endothelial dysfunction, or due to overactivation of p38 MAPK in infected cardiomyocytes, which has been shown to induce apoptosis, impair contractility, and increase fibrosis [24] . Additionally, elevated p38 MAPK levels are also implicated in hypoxic pulmonary vasoconstriction and vascular remodeling, which are poor predictors of outcome in acute lung injury [25, 26] . These effects are not entirely unexpected as excessive Ang II signaling, which transmits through p38 MAPK, also produces similar effects. However, in this instance, both direct viral activation of p38 MAPK and overactive Ang II p38 signaling may combine to create severe clinical complications. A number of direct p38 MAPK inhibitors are in clinical trials for other indications and could be repurposed for randomized, controlled trials (RCT) in patients at risk for serious COVID-19 infection. Losmapimod is the most clinically studied p38 inhibitor and it has a favorable reasonable safety profile. In 12 healthy volunteers, a single IV infusion of 3 mg of losmapimod and a subsequent 15 mg oral dose were safe and well-tolerated. Headache was the only adverse event reported more than once. Also reported were nausea, fatigue, dry mouth, neuralgia and nasopharyngitis. Nausea was the only adverse event thought to be drug related [27] . In a 28-day RCT comparing 7.5 mg of losmapimod twice daily versus placebo in hypercholesterolemic patients, losmapimod was well-tolerated and safe in 27 patients, with no J o u r n a l P r e -p r o o f Journal Pre-proof serious adverse events. The most commonly reported side effect was headache and there were no differences between treatment groups in other safety data-labs, vital signs, and ECGincluding liver function tests [28] . In a 12-week RCT of 7.5 mg of losmapimod twice daily in 3503 patients previously hospitalized with acute myocardial infarction, 16% of the 1738 patients in the on-treatment group experienced serious adverse events compared to 14.2% in the placebo group, requiring study drug discontinuation in 4.4% of on-drug patients and 3.9% of placebo patients [29] . Dilmapimod is another p38 inhibitor previously studied in 77 patients at risk of acute lung injury following trauma in a clinical trial with no relevant safety findings [30] . Various other p38 MAPK inhibitors are currently in clinical trials for a range of indications. The p38 MAPK pathway is a key mediator of inflammation implicated in lung and heart injury. The SARS-CoV-2 virus, like SARS-CoV and other RNA respiratory viruses, may directly upregulate p38 MAPK, perhaps for replication purposes. However, the unique viral entry mechanism of SARS-CoV-2 may also shut off disables a key counterbalancing mechanism employed by the cell to dampen p38 signaling through ACE2 activity. This combination may allow a perfect storm of unrestrained p38 MAPK activity, promoting inflammation, vasoconstriction, and thrombosis, while simultaneously benefiting the viral lifecycle. As such, p38 MAPK inhibition may be an appropriate treatment for SARS-CoV-2 infection. This is supported by a small preclinical study of SARS-CoV infection in a mouse model. Further preclinical trials should be performed to implicate elucidate the role of p38 activation in animal models of SARS-CoV-2. More importantly, safe, and well tolerated tolerable p38 MAPK inhibitors that are already in clinical stages of development. These drugs should be considered J o u r n a l P r e -p r o o f Journal Pre-proof for could be readily repurposed in randomized, controlled trials enrolling in patients at risk for serious COVID-19 complications. No conflicts of interest exist for either author. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. J o u r n a l P r e -p r o o f SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China Modulation of mitogen-activated protein kinase attenuates sepsis-induced acute lung injury in acute respiratory distress syndrome rats Inhibition of p38 Mitogen-Activated Protein Kinase Decreases Cardiomyocyte Apoptosis and Improves Cardiac Function After Myocardial Ischemia and Reperfusion Activation and signaling of the p38 MAP kinase pathway Angiotensin Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System Immunologic Effects of the Renin-Angiotensin System ACE2, angiotensin-(1-7) and Mas receptor axis in inflammation and fibrosis Wellner Maren, et al. p38 Mitogen-Activated Protein Kinase Inhibition Ameliorates Angiotensin II-Induced Target Organ Damage Angiotensin-converting enzyme 2-angiotensin (1-7)-Mas axis prevents pancreatic acinar cell inflammatory response via inhibition of the p38 mitogen-activated protein kinase/nuclear factor-κB pathway Reactive Oxygen Species and p38 Mitogen-activated Protein Kinase Mediate Tumor Necrosis Factor α-Converting Enzyme (TACE/ADAM-17) Activation in Primary Human Monocytes Association of Inpatient Use of Angiotensin Converting Enzyme Inhibitors and Angiotensin II Receptor Blockers with Mortality Among Patients With Hypertension Hospitalized With COVID-19 Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury 7a Protein of Severe Acute Respiratory Syndrome Coronavirus Inhibits Cellular Protein Synthesis and Activates p38 Mitogen-Activated Protein Kinase Mitogen-activated Protein Kinase Impairs Influenza Virus-induced Primary and Secondary Host Gene Responses and Protects Mice from Lethal H5N1 Infection Toll-Like Receptor 4-Mediated Activation of p38 Mitogen-Activated Protein Kinase Is a Determinant of Respiratory Virus Entry and Tropism Angiotensin II regulates ACE and ACE2 in neurons through p38 mitogen-activated protein kinase and extracellular signal-regulated kinase 1/2 signaling The PDZ-binding motif of severe acute respiratory syndrome coronavirus envelope protein is a determinant of viral pathogenesis Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. The Lancet Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19 Endothelial cell infection and endotheliitis in COVID-19. The Lancet. 2020; Ahead of print Arterial Thrombosis Is Accompanied by Elevated Mitogen-Activated Protein Kinase (MAPK) and Cyclooxygenase-2 Expression via Toll-Like Receptor 4 (TLR-4) Activation by S100A8/A9 The p38 mitogen-activated protein kinase pathway--a potential target for intervention in infarction, hypertrophy, and heart failure The reversal of pulmonary vascular remodeling through inhibition of p38 MAPK-alpha: a potential novel antiinflammatory strategy in pulmonary hypertension p38 mitogen-activated protein kinase mediates the sustained phase of hypoxic pulmonary vasoconstriction and plays a role in phase I vasodilation Safety, tolerability, pharmacokinetics and pharmacodynamics of losmapimod following a single intravenous or oral dose in healthy volunteers Inhibition of p38 Mitogen-Activated Protein Kinase Improves Nitric Oxide-Mediated Vasodilatation and Reduces Inflammation in Hypercholesterolemia Effect of Losmapimod on Cardiovascular Outcomes in Patients Hospitalized With Acute Myocardial Infarction: A Randomized Clinical Trial  The p38 MAPK pathway is a proinflammatory pathway implicated in lung and heart injury  Loss of ACE2 activity in COVID-19 infection allows for p38 upregulation via Ang II  SARS-CoV-2 may also directly activate p38 via viral proteins to promote replication  Overactive p38 may explain inflammation, thrombosis, and vasoconstriction in COVID-19  p38 inhibitors are in clinical development and could be trialed in COVID-19 patients J o u r n a l P r e -p r o o f