6
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Off-label therapy targeting pathogenic inflammation in COVID-19

      brief-report

      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

          The world is facing a pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) for which no proven specific therapies are available other than supportive ones. From the start of the coronavirus disease 2019 (COVID-19) outbreak, in China and in other countries patients have received off-label and compassionate use therapies, such as interferon (IFN)-α combined with the repurposed drug Kaletra, an approved cocktail of the human immunodeficiency virus (HIV) protease inhibitors ritonavir and lopinavir, chloroquine, azithromycin, favipiravir, remdesivir, steroids, and anti-interleukin (IL)-6 inhibitors, based on either their in vitro antiviral or anti-inflammatory properties. SARS-CoV-2 is an enveloped, positive-sense, single-stranded RNA β-coronavirus similar to the severe acute respiratory syndrome (SARS-CoV) and Middle East respiratory syndrome (MERS-CoV) viruses. No clinical evidence currently supports the efficacy and safety of any drugs against coronaviruses in humans, including SARS-CoV-2. Existing antivirals and knowledge gained from the SARS and MERS outbreaks have been employed as the fastest route to fight the current coronavirus epidemic. Testing therapies approved for other indications makes senses. The World Health Organization considered remdesivir the most promising candidate to treat COVID-19, on the basis of its broad spectrum activity and clinical safety from Ebola virus disease trials. However, antivirals known to be acting at targets not playing a role in the replication of coronaviruses may fail in clinical studies. The lack of a concurrent control group prevents any true appreciation of the beneficial versus harmful effects of the off-label use of any drugs, which might be the case for the cardiovascular effects of chloroquine/hydroxychloroquine, azithromycin, and lopinavir–ritonavir. Similarly, the adverse effects associated with the compassionate use of remdesivir could not be anticipated given the paucity of information available from previous trials. Therefore, it is appropriate to propose and test implementable hypotheses to discover new therapies for the current and any future coronavirus pandemics. Most of the drugs in clinical trials inhibit key components of the coronavirus infection lifecycle 1 . However, the occurrence and outcome of COVID-19 infection depend on the interaction between the virus and an individual’s immune system. Many viruses multiply in the host without causing significant damage, including viruses that are capable of causing disease. However, the host response itself may lead to pathological outcomes, which may be relatively nonspecific or may result in a specific injury in target organs via cellular and humoral immune responses. Accumulating evidence suggests that some patients with severe COVID-19 infection might have a cytokine storm-like syndrome, contributing to the often lethal acute respiratory distress syndrome 2 . The severity of COVID-19 disease has been associated with increased chemokines and cytokines, such as tumor necrosis factor (TNF)-α and, to a lesser extent, IL-1β and IL-6, suggesting the occurrence of an uncontrolled inflammation in response to the virus. Of note, bats tolerate coronavirus—no inflammation in the face of an unimpaired viral load—thanks to a dampened transcriptional priming of the inflammasome sensor NLR family pyrin domain containing 3 (NLRP3) 3 , one major executor of the vertebrate inflammatory response. This suggests that targeting selective pathways of the inflammatory response—rather than interfering with the plethora of inflammatory pathways—might be a successful strategy in COVID-19 infection. To achieve this goal, immunomodulatory agents capable of keeping the runaway inflammatory response at bay, without compromising the ability of the immune system to respond to pathogens, are urgently needed. As a matter of fact, with respect to anti-inflammatory therapy in COVID-19 infection, the use of intravenous steroids has been associated with delayed coronavirus clearance in both blood and lungs and steroids were associated with significantly increased risk of mortality and secondary infections in patients with influenza 4 . Furthermore, in spite of the documented efficacy of the IL-6 inhibitor tocilizumab in the treatment of COVID-19 5,6 , IL-6 inhibitors may cause even more profound immunosuppression than steroids, increasing the risk of sepsis, bacterial pneumonia, gastrointestinal perforation, and hepatotoxicity 7 . Preclinical studies have pointed to the efficacy of the selective inflammatory pathways blockade in lung inflammation by drugs that are already in use in humans. Patients with COVID-19 may have features mimicking rheumatic diseases, such as arthralgias, acute interstitial pneumonia, myocarditis, leucopenia, lymphopenia, thrombocytopenia, and cytokine storm. This may suggest that drugs commonly used in rheumatology could be beneficial in COVID-19. Accordingly, several TNF-α-blocking antibodies, successfully used to treat inflammatory diseases, have been recommended for the hospitalized COVID-19 patients 8 . Similarly, IL-1β inhibitors may have significant potential at controlling hyperinflammation in severe COVID-19 disease. Among the latter, anakinra is the recombinant form of the IL-1 receptor antagonist, known to inhibit NLRP3 via blockade of the IL-1 receptor I. Anakinra is currently used to treat a wide range of diseases that goes beyond its approved indications for rheumatoid arthritis and cryopyrin-associated periodic syndromes, to encompass cancer and chronic inflammatory diseases 9 . Anakinra potently inhibited pathogenic NLRP3 activation in the murine lung and human bronchial epithelial cells from patients with lung infection and inflammation, and it concurrently inhibited IL-1β, TNF-α, and IL-6 production 10 . Compared to other biologics, anakinra has an unparalleled safety profile. It is currently being explored in a phase IIa, randomized, placebo-controlled, double-blind, cross-over study in patients with cystic fibrosis (NCT03925194). Thus patients with severe COVID-19 may likely benefit from therapeutic options that include, among others, NLRP3 antagonists and IL-1 inhibitors to inhibit unwanted inflammation while preserving antimicrobial defense. Currently, anakinra is being trialed in a randomized placebo-controlled study in children and adults with COVID-19-associated cytokine storm syndrome in China (NCT02780583) and in a phase 2/3, randomized, open-label, multicenter study investigating the efficacy and safety of intravenous administrations of anakinra in Italy (Sobi.IMMUNO-101, March 20, 2020). AIFA has also recently approved the compassionate use of canakinumab, a fully human monoclonal antibody that neutralizes the bioactivity of human IL-1β, in patients with COVID-19 (April 10, 2020). Consistent with the observation that a robust cytotoxic T lymphocyte response plays a vital role in clearing coronavirus, lymphopenia, affecting both CD4+ and CD8+ T lymphocytes, has been described in patients with both SARS-CoV and MERS-CoV infections and correlated with the severity of the disease 11,12 . For its ability to stimulate innate and adaptive immune responses, thymosin alpha1 (Tα1), a naturally occurring thymic peptide of 28 amino acids, is used worldwide as an immunomodulator in a wide range of clinical indications 13,14 (Fig. 1). Owing to its ability to promote IFN type I production and activation of CD8+ T cells via stimulation of innate receptors, Tα1 has been registered as a treatment or prevention of respiratory viral infections and as an immune adjuvant in influenza vaccination of the elderly and hemodialysis patients 13,14 . Either in monotherapy or in combination with IFN-α, Tα1 has been approved in 30 countries for treatment of chronic viral infections, including chronic hepatitis, chronic hepatitis C, and HIV 13,14 . During the 2009 pandemic outbreak of H1N1 influenza, Tα1 provided an earlier and greater response to the vaccine in a clinical study with Focetria™ MF59-adjuvanted monovalent H1N1 vaccine 15 . Interestingly, the use of Tα1 has also been claimed in SARS 16,17 . By reducing the mortality rate and decreasing the incidence of secondary infections, Tα1 has shown a promising activity also in sepsis 18 . Not surprisingly, Tα1 has been included in a recent randomized, open, controlled trial in combination with darunavir/cobicistat or lopinavir/ritonavir in the treatment of COVID-19 (registration number ChiCTR2000029541) 1 . Owing to its ability to maintain immune homeostasis by activating the tolerogenic pathway of tryptophan catabolism via the immunoregulatory enzyme indoleamine 2,3-dioxygenase 1, Tα1 specifically potentiated immune tolerance in diseased lungs, breaking the vicious circle linking chronic lung inflammation and infections 19 and restoring ciliary beating function 20 . Thus Tα1 holds promise for representing an ideal candidate drug that, as suggested 21 , will boost immunity at an early stage of SARS-CoV-2 infection and promote immune tolerance and tissue homeostasis in the severe respiratory stage. Fig. 1 The figure illustrates the dynamics of the antiviral innate and adaptive immune response and its dysregulation potentially leading to the progression of COVID-19 infection. As explained in the text, by virtue of its multifaceted activity, thymosin α1 may boost immunity at an early and late stages of SARS-CoV-2 infection and, like cytokine-specific antagonists, may adversely affect the cytokine storm by promoting immune tolerance and tissue homeostasis in the severe respiratory stage. Even though virtual screening makes it possible to discover molecules relatively quickly, these compounds still need to be experimentally tested before clinical use. Drug repurposing, with a breadth of a dual activity against the virus and the host, may thus rightly come into play with safety and low cost. Note added in proof After the submission of the present comment, the following papers (shown in References) have been published: 1. Pontali, E. et al. Safety and efficacy of early high-dose IV anakinra in severe COVID-19 lung disease. J. Allergy Clin. Immunol. S0091–6749, 30634–30635 (2020). 10.1016/j.jaci.2020.05.002. 2. Filocamo, G. et al. Use of anakinra in severe COVID-19: a case report. Int. J. Infect. Dis. S1201–9712, 30333–30337 (2020). 10.1016/j.ijid.2020.05.026. 3. Day, J. W., Fox, T. A., Halsey, R., Carpenter, B., & Kottaridis, P. D. Br J. IL-1 blockade with anakinra in acute leukaemia patients with severe COVID-19 pneumonia appears safe and may result in clinical improvement. Haematology. 10.1111/bjh.16873 (2020). 4. Dimopoulos, G. et al. Favorable anakinra responses in severe COVID-19 patients with secondary hemophagocytic lymphohistiocytosis. Cell Host Microbe. 10.1016/j.chom.2020.05.007 (2020). 5. Liu, Y. et al. Thymosin Alpha 1 (Tα1) reduces the mortality of severe COVID-19 by restoration of lymphocytopenia and reversion of exhausted T cells. Clin. Infect Dis. 10.1093/cid/ciaa630 (2020).

          Related collections

          Most cited references13

          • 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

            Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury

            The 2019 novel coronavirus (2019-nCoV) outbreak is a major challenge for clinicians. The clinical course of patients remains to be fully characterised, little data are available that describe the disease pathogenesis, and no pharmacological therapies of proven efficacy yet exist. Corticosteroids were widely used during the outbreaks of severe acute respiratory syndrome (SARS)-CoV 1 and Middle East respiratory syndrome (MERS)-CoV, 2 and are being used in patients with 2019-nCoV in addition to other therapeutics. 3 However, current interim guidance from WHO on clinical management of severe acute respiratory infection when novel coronavirus (2019-nCoV) infection is suspected (released Jan 28, 2020) advises against the use of corticosteroids unless indicated for another reason. 4 Understanding the evidence for harm or benefit from corticosteroids in 2019-nCoV is of immediate clinical importance. Here we discuss the clinical outcomes of corticosteroid use in coronavirus and similar outbreaks (table ). Table Summary of clinical evidence to date Outcomes of corticosteroid therapy * Comment MERS-CoV Delayed clearance of viral RNA from respiratory tract 2 Adjusted hazard ratio 0·4 (95% CI 0·2–0·7) SARS-CoV Delayed clearance of viral RNA from blood 5 Significant difference but effect size not quantified SARS-CoV Complication: psychosis 6 Associated with higher cumulative dose, 10 975 mg vs 6780 mg hydrocortisone equivalent SARS-CoV Complication: diabetes 7 33 (35%) of 95 patients treated with corticosteroid developed corticosteroid-induced diabetes SARS-CoV Complication: avascular necrosis in survivors 8 Among 40 patients who survived after corticosteroid treatment, 12 (30%) had avascular necrosis and 30 (75%) had osteoporosis Influenza Increased mortality 9 Risk ratio for mortality 1·75 (95% CI 1·3–2·4) in a meta-analysis of 6548 patients from ten studies RSV No clinical benefit in children10, 11 No effect in largest randomised controlled trial of 600 children, of whom 305 (51%) had been treated with corticosteroids CoV=coronavirus. MERS=Middle East respiratory syndrome. RSV=respiratory syncytial virus. SARS=severe acute respiratory syndrome. * Hydrocortisone, methylprednisolone, dexamethasone, and prednisolone. Acute lung injury and acute respiratory distress syndrome are partly caused by host immune responses. Corticosteroids suppress lung inflammation but also inhibit immune responses and pathogen clearance. In SARS-CoV infection, as with influenza, systemic inflammation is associated with adverse outcomes. 12 In SARS, inflammation persists after viral clearance.13, 14 Pulmonary histology in both SARS and MERS infections reveals inflammation and diffuse alveolar damage, 15 with one report suggesting haemophagocytosis. 16 Theoretically, corticosteroid treatment could have a role to suppress lung inflammation. In a retrospective observational study reporting on 309 adults who were critically ill with MERS, 2 almost half of patients (151 [49%]) were given corticosteroids (median hydrocortisone equivalent dose [ie, methylprednisolone 1:5, dexamethasone 1:25, prednisolone 1:4] of 300 mg/day). Patients who were given corticosteroids were more likely to require mechanical ventilation, vasopressors, and renal replacement therapy. After statistical adjustment for immortal time and indication biases, the authors concluded that administration of corticosteroids was not associated with a difference in 90-day mortality (adjusted odds ratio 0·8, 95% CI 0·5–1·1; p=0·12) but was associated with delayed clearance of viral RNA from respiratory tract secretions (adjusted hazard ratio 0·4, 95% CI 0·2–0·7; p=0·0005). However, these effect estimates have a high risk of error due to the probable presence of unmeasured confounders. In a meta-analysis of corticosteroid use in patients with SARS, only four studies provided conclusive data, all indicating harm. 1 The first was a case-control study of SARS patients with (n=15) and without (n=30) SARS-related psychosis; all were given corticosteroid treatment, but those who developed psychosis were given a higher cumulative dose than those who did not (10 975 mg hydrocortisone equivalent vs 6780 mg; p=0·017). 6 The second was a randomised controlled trial of 16 patients with SARS who were not critically ill; the nine patients who were given hydrocortisone (mean 4·8 days [95% CI 4·1–5·5] since fever onset) had greater viraemia in the second and third weeks after infection than those who were given 0·9% saline control. 5 The remaining two studies reported diabetes and avascular necrosis as complications associated with corticosteroid treatment.7, 8 A 2019 systematic review and meta-analysis 9 identified ten observational studies in influenza, with a total of 6548 patients. The investigators found increased mortality in patients who were given corticosteroids (risk ratio [RR] 1·75, 95% CI 1·3–2·4; p=0·0002). Among other outcomes, length of stay in an intensive care unit was increased (mean difference 2·1, 95% CI 1·2–3·1; p<0·0001), as was the rate of secondary bacterial or fungal infection (RR 2·0, 95% CI 1·0–3·8; p=0·04). Corticosteroids have been investigated for respiratory syncytial virus (RSV) in clinical trials in children, with no conclusive evidence of benefit and are therefore not recommended. 10 An observational study of 50 adults with RSV infection, in which 33 (66%) were given corticosteroids, suggested impaired antibody responses at 28 days in those given corticosteroids. 17 Life-threatening acute respiratory distress syndrome occurs in 2019-nCoV infection. 18 However, generalising evidence from acute respiratory distress syndrome studies to viral lung injury is problematic because these trials typically include a majority of patients with acute respiratory distress syndrome of non-pulmonary or sterile cause. A review of treatments for acute respiratory distress syndrome of any cause, based on six studies with a total of 574 patients, 19 concluded that insufficient evidence exists to recommend corticosteroid treatment. 20 Septic shock has been reported in seven (5%) of 140 patients with 2019-nCoV included in published reports as of Jan 29, 2020.3, 18 Corticosteroids are widely used in septic shock despite uncertainty over their efficacy. Most patients in septic shock trials have bacterial infection, leading to vasoplegic shock and myocardial insufficiency.21, 22 In this group, there is potential that net benefit might be derived from steroid treatment in severe shock.21, 22 However, shock in severe hypoxaemic respiratory failure is often a consequence of increased intrathoracic pressure (during invasive ventilation) impeding cardiac filling, and not vasoplegia. 23 In this context, steroid treatment is unlikely to provide a benefit. No clinical data exist to indicate that net benefit is derived from corticosteroids in the treatment of respiratory infection due to RSV, influenza, SARS-CoV, or MERS-CoV. The available observational data suggest increased mortality and secondary infection rates in influenza, impaired clearance of SARS-CoV and MERS-CoV, and complications of corticosteroid therapy in survivors. If it is present, the effect of steroids on mortality in those with septic shock is small, and is unlikely to be generalisable to shock in the context of severe respiratory failure due to 2019-nCoV. Overall, no unique reason exists to expect that patients with 2019-nCoV infection will benefit from corticosteroids, and they might be more likely to be harmed with such treatment. We conclude that corticosteroid treatment should not be used for the treatment of 2019-nCoV-induced lung injury or shock outside of a clinical trial.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed

              With more than 81 000 deaths worldwide from coronavirus disease 2019 (COVID-19) by April 8, 2020, 1 it is incumbent on researchers to accelerate clinical trials of any readily available and potentially acceptably safe therapies that could reduce the rising death toll. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gains access to host cells via angiotensin-converting enzyme 2, which is expressed in the type II surfactant-secreting alveolar cells of the lungs. 2 Severe COVID-19 is associated with a major immune inflammatory response with abundant neutrophils, lymphocytes, macrophages, and immune mediators. Which mediators are most important in driving the immune pathology remains to be elucidated. Deaths from COVID-19 are chiefly due to diffuse alveolar damage with pulmonary oedema, hyaline membrane formation, and interstitial mononuclear inflammatory infiltrate compatible with early-phase adult respiratory distress syndrome (ARDS). 3 Prevention of ARDS and death in patients with COVID-19 is a pressing health emergency. Anti-tumour necrosis factor (TNF) antibodies have been used for more than 20 years in severe cases of autoimmune inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, or ankylosing spondylitis. There are ten (as reported on Sept 29, 2019) US Food and Drug Administration approved and four off-label indications for anti-TNF therapy, 4 indicating that TNF is a valid target in many inflammatory diseases. TNF is present in blood and disease tissues of patients with COVID-19 5 and TNF is important in nearly all acute inflammatory reactions, acting as an amplifier of inflammation. We propose that anti-TNF therapy should be evaluated in patients with COVID-19 on hospital admission to prevent progression to needing intensive care support. There is evidence of an inflammatory excess in patients with COVID-19. Lung pathology in COVID-19 is characterised by capillary leakage of fluid and recruitment of immune-inflammatory lymphocytes, neutrophils, and macrophages, 6 implying a role for adhesion molecules, chemokines, and cytokines targeting vascular endothelium. Cytokine upregulation is documented in COVID-19. In patients with COVID-19, there is upregulation of pro-inflammatory cytokines in the blood, including interleukin (IL)-1, IL-6, TNF, and interferon γ,7, 8 and patients in intensive care units have increased concentrations of many cytokines. Preliminary data from Salford Royal Hospital and the University of Manchester in the UK document the presence of proliferating excess monocytes expressing TNF by intracellular staining in patients with COVID-19 in intensive care (Hussell T, Grainger J, Menon M, Mann E, University of Manchester, Manchester, UK, personal communication). Available cytokine data on immunology and inflammation in COVID-19 are summarised in the appendix. Initial reports comprising a trial of 21 severe and critical COVID-19 patients in China (ChiCTR2000029765) and a case study from France 9 of clinical benefit with the anti-IL6 receptor antibody 10 tocilizumab in COVID-19 suggest that cytokines are of importance in the “cytokine storm” and further controlled clinical trials are in progress. Although there are many potential drug candidates for reducing inflammation in COVID-19, only a few drugs such as the anti-TNF antibodies infliximab or adalimumab are potentially effective, widely available, and have a well established safety profile. The potential role of anti-TNF therapy thus warrants consideration. Preclinical studies suggest that the response to severe respiratory syncytial virus (RSV) and influenza in mice is ameliorated by anti-TNF therapy, which reduces weight loss, disease duration, and cell and fluid infiltrate. 11 This research suggests a potential rationale for use of anti-TNF therapy in viral pneumonia, especially given the known mechanism of action of TNF and the reversal of TNF-induced immunopathology by TNF blockade in multiple diseases. It is known TNF is produced in most types of inflammation, especially in the acute phase, and is important in the coordination and development of the inflammatory response. However, too much production of TNF for too long becomes immune suppressive. 12 Blockade of TNF alone is clinically effective in many circumstances and diseases, despite the presence of many other pro-inflammatory cytokines and mediators. There is evidence of a “TNF dependent cytokine cascade” in rheumatoid arthritis tissue and upon bacterial challenge in baboons.13, 14 Thus, if TNF is blocked, there is a rapid (ie, <12 h) decrease of IL-6 and IL-1 concentrations in patients with active rheumatoid arthritis 15 and, importantly, a reduction of adhesion molecules and vascular endothelial growth factor, which is also known as vascular permeability factor, denoting its importance in capillary leak.15, 16, 17, 18, 19 Furthermore, a reduction in leucocyte trafficking occurs in inflamed tissues of joints due to reduction in adhesion molecules and chemokines 20 with reduction in cell content and exudate. Finally, after anti-TNF infusion tissue TNF is reduced as it passes into the blood bound to the anti-TNF antibody. Blood concentrations of immunoreactive, but biologically inactive, TNF increase more than ten times after infusion. 15 For these reasons it is possible that a single infusion of anti-TNF antibody might reduce some of the processes that occur during COVID-19 lung inflammation, reducing TNF and other inflammatory mediators, cellularity, and exudate. © 2020 Marco Mantovani/Getty Images 2020 Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active. What would be the best time for intervention with anti-TNF therapy in patients with COVID-19? We postulate that the earlier the better after hospital admission might be the answer because patients will already have initiated anti-viral immunity for several days. There is a balance to be struck between stage of intervention and ensuring patients are at sufficient risk of a poor outcome and can be appropriately monitored. We propose that initial assessment of anti-TNF therapy in clinical trials should be in patients with moderate disease admitted to hospital and who require oxygen support but not intensive care. If this treatment approach proved beneficial with a good safety profile, treatment in the community for people identified as being at high risk of progressing to hospital admission might be considered. The range of available formulations and administration routes of anti-TNF products could facilitate this treatment approach. Is there a trade-off between immunity and virus clearance? The use of powerful anti-inflammatory drugs in acute viral diseases has to be approached with caution because of the risk of increasing viral replication or bacterial infections. For lung viral infections, the higher the infectious dose, the greater the tissue damage from viral replication and the ensuing immune response. In animal models that resemble lung viral infection in humans, the immune response to the virus is so great that even a moderate reduction in inflammation is beneficial—eg, mice with severe pneumonia from RSV or influenza benefit from anti-TNF treatment without compromising viral clearance 11 because more of the lung architecture is preserved. However, concerns about safety are important when considering new therapy. Would anti-TNF therapy increase the risk of bacterial or fungal super-infections? After respiratory viral infection, superinfections with other organisms occur at the most severe end of the disease spectrum. Many research groups have elucidated the mechanisms responsible 21 and anecdotal evidence suggests that bacteria might have a role in in COVID-19,5, 22 although this remains to be confirmed. Bacteria gain a foothold faster in a lung that is damaged. Experimental studies suggest that if the duration of inflammation is limited, with its associated collateral lung damage, then bacterial superinfection is reduced. 23 There is concern that anti-TNF therapy might increase the risk of bacterial infection. 24 Yet two randomised studies in critically unwell patients with septic shock25, 26 showed that monoclonal anti-TNF therapy had good safety data with no evidence of increased secondary bacterial infections in the anti-TNF treated group. In an observational trial in rheumatoid arthritis patients with serious infections, the risk of sepsis and death was reduced in patients on TNF inhibitors compared with those on synthetic disease-modifying anti-rheumatic drugs (DMARDS). 27 46 (11%) of 399 patients on TNF inhibitors developed sepsis after serious infection, of whom 20 (43%) died, compared with 74 (17%) of 444 patients on DMARDS who developed sepsis, of whom 54 (74%) died. 27 Paradoxically, another class of TNF inhibitor, a TNF-R2 Ig-Fc fusion protein, etanercept, was associated with moderately increased mortality in a randomised trial of this treatment for sepsis, 28 possibly due to its faster off-rate for TNF potentially resulting in some redistribution and bioavailability of pathogenic TNF rather than its clearance. There has been interest as to whether the safety of anti-TNF therapy in patients with COVID-19 might be gleaned from analysis of the course of COVID-19 in patients with inflammatory bowel disease (IBD) or rheumatoid arthritis who are already on anti-TNF treatment. As of April 6, 2020, on SECURE-IBD, a coronavirus and IBD reporting database with a register of outcomes of IBD patients with COVID-19, there were 116 patients on anti-TNF therapy alone, 99 of whom recovered without hospitalisation and one patient died. By contrast, about half of 71 patients on sulfasalazine/mesalamine recovered without hospital admission and six patients died. Thus IBD patients with COVID-19 on anti-TNF therapy do not fare worse than those treated with other drugs, but there are insufficient data to make conclusions about a better outcome. We believe there is sufficient evidence to support clinical trials of anti-TNF therapy in patients with COVID-19. With an average of 2 days between hospital admission and ARDS, 7 we propose anti-TNF therapy should be initiated as early as is practicable. If there is preliminary evidence of benefit and safety of anti-TNF therapy in hospitalised patients, we suggest consideration should be given to out of hospital treatment for patients with COVID-19 at high risk, such as older people and those with pre-existing conditions, and who can be monitored appropriately.
                Bookmark

                Author and article information

                Contributors
                luigina.romani@unipg.it
                garenco@icloud.com
                Journal
                Cell Death Discov
                Cell Death Discov
                Cell Death Discovery
                Nature Publishing Group UK (London )
                2058-7716
                12 June 2020
                12 June 2020
                2020
                : 6
                : 49
                Affiliations
                [1 ]ISNI 0000 0004 1757 3630, GRID grid.9027.c, Department of Experimental Medicine, , University of Perugia, ; Perugia, Italy
                [2 ]University San Raffaele and IRCCS San Raffaele, 00166 Rome, Italy
                Author information
                http://orcid.org/0000-0002-1356-525X
                http://orcid.org/0000-0001-6674-2128
                Article
                283
                10.1038/s41420-020-0283-2
                7290072
                32547788
                2755592d-969e-4945-8bf9-078a14796e40
                © The Author(s) 2020

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 20 April 2020
                : 5 May 2020
                : 12 May 2020
                Funding
                Funded by: FundRef https://doi.org/10.13039/501100008385, Fondazione per la Ricerca sulla Fibrosi Cistica (Fondazione FFC);
                Award ID: FFC#24/2018
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100000780, European Commission (EC);
                Award ID: 847507 - HDM-FUN
                Award Recipient :
                Categories
                Comment
                Custom metadata
                © The Author(s) 2020

                infectious diseases,medical research
                infectious diseases, medical research

                Comments

                Comment on this article