Antirheumatic Drugs against COVID-19 from the Perspective of Rheumatologists
Abstract
:1. Introduction
2. Mechanisms of SARS-CoV-2 Infection
Drug | Mechanism of Action | Dosage Used in Some Clinical Trials | Potential Efficacy Reported in Clinical Trials | Recommendations |
---|---|---|---|---|
Chloroquine Hydroxychloroquine | Inhibit viral entry, endocytosis, and production of cytokines | 800 to 1600 mg p.o. 1 to 3 divided doses on the first day, and then 200 to 800 mg in 1 to 2 divided doses daily for 5 to 21 days | None | WHO recommend against administering chloroquine or hydroxychloroquine, except in a clinical trial |
Colchicine | Inhibit endocytosis | 0.5 mg p.o. twice daily for 3 days, then once daily for 27 days | Administration as soon as the diagnosis reduces the risk of disease aggravation and the resulting number of hospitalization Improve points on a 7-grade clinical status scale | |
Cyclosporine A | Suppress the T cell response Inhibit the pro-inflammatory cytokines | 1–2 mg/kg/day p.o. divided into two doses for 7 days | Improve outcomes and reduce mortality, mainly in patients with moderate to severe disease | |
Tacrolimus | Suppress the T cell response | Dose to achieve plasma levels of 8–10 ng/mL p.o. | Not reported | |
Glucocorticoids | Suppress inflammation and excessive immune response | dexamethasone (6 mg daily for up to 10 days) | Reduce 28-day mortality | The Panel recommends using dexamethasone in hospitalized patients with COVID-19 who require supplemental oxygen |
Tocilizumab | Suppresses cytokine storms by IL-6 inhibition | 8 mg/kg i.v. (maximum dose 800 mg) in combination with dexamethasone | Improve 90-day survival Reduce organ support-free days Reduce time to discharge | The Panel recommends concomitant use of dexamethasone in recently hospitalized patients who are exhibiting rapid respiratory decompensation |
Sarilumab | 400 mg i.v. | The Panel recommends its use only when tocilizumab is not available or is not feasible to use | ||
Siltuximab | 11 mg/kg i.v. | Not yet reported | ||
Anakinra | Suppress the cytokine storm, reduce endothelial dysfunction and microvascular alteration by inhibiting IL-1 | 200 mg i.v. twice daily for 3 days, 100 mg i.v. twice daily on day 4, and 100 mg i.v. on day 5 | Reduce mortality at day 28 and the need for mechanical ventilation | |
Canakinumab | 450 mg for body weight 40–59 kg, 600 mg for 60–80 kg or 750 mg for >80 kg i.v. | Improve oxygenation Decrease serum CRP levels | ||
Baricitinib | Reduce viral entry Inhibit proinflammatory intracellular signals of some cytokines by inhibiting JAK1 and JAK2 | 4 mg p.o. per day for 14 days or until hospital discharge | Reduce a median time to recovery by 1 day Accelerate improvement in clinical status Reduce 28-day mortality | The Panel recommends concomitant use with dexamethasesone in recently hospitalized patients receiving high-flow oxygen therapy or non-invasive ventilation |
Tofacitinib | Inhibit proinflammatory intracellular signals of some cytokines by inhibiting JAK1 and JAK3 | 10 mg p.o. twice daily on day 1, followed by 5 mg twice daily on day 2–5 | Not yet reported | |
Infliximab | Suppress inflammation by inhibiting TNF-α | 5 mg/kg i.v. | Reduced mortality | |
Abatacept | Inhibit T cell activation and suppress the production of inflammatory cytokines | 10 mg/kg i.v. (maximum dose 1000 mg) | Not yet reported | |
Apremilast | Prevent the overproduction of inflammatory cytokines by inhibiting PDE4 | 30 mg p.o. twice daily for 14 days | Antipyretic effects Improve oxygenation |
3. Pharmacotherapy
3.1. Chloroquine (CQ) and Hydroxychloroquine (HCQ)
3.2. Colchicine
3.3. Calcineurin Inhibitors
3.4. Glucocorticoids
3.5. IL-6 Inhibitors
3.6. IL-1 Inhibitors
3.7. Janus Kinase (JAK) Inhibitors
3.8. TNF Inhibitors
3.9. Cytotoxic T Lymphocyte-Associated Antigen 4 (CTLA-4)-Ig
3.10. Phosphodiesterase 4 (PDE4) Inhibitors
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review. JAMA 2020, 324, 782–793. [Google Scholar] [CrossRef] [PubMed]
- Epidemiology Working Group for NCIP Epidemic Response, Chinese Center for Disease Control and Prevention. [The epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) in China]. Zhonghua Liu Xing Bing Xue Za Zhi 2020, 41, 145–151. [Google Scholar]
- Wu, C.; Chen, X.; Cai, Y.; Xia, J.; Zhou, X.; Xu, S.; Huang, H.; Zhang, L.; Zhou, X.; Du, C.; et al. Risk Factors Associated with Acute Respiratory Distress Syndrome and Death in Patients with Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern. Med. 2020, 180, 934–943. [Google Scholar] [CrossRef] [Green Version]
- Siddiqi, H.K.; Mehra, M.R. COVID-19 illness in native and immunosuppressed states: A clinical–therapeutic staging proposal. J. Heart Lung Transplant. 2020, 39, 405–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGonagle, D.; Sharif, K.; O’Regan, A.; Bridgewood, C. The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun. Rev. 2020, 19, 102537. [Google Scholar] [CrossRef]
- Del Valle, D.M.; Kim-Schulze, S.; Huang, H.H.; Beckmann, N.D.; Nirenberg, S.; Wang, B.; Lavin, Y.; Swartz, T.H.; Madduri, D.; Stock, A.; et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med. 2020, 26, 1636–1643. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
- Behrens, E.M.; Koretzky, G.A. Review: Cytokine Storm Syndrome: Looking Toward the Precision Medicine Era. Arthritis Rheumatol. 2017, 69, 1135–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages. Nat. Rev. Immunol. 2020, 20, 355–362. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Zhu, L.; Chan, T.; Lu, X.; Shen, W.; Madigan, M.C.; Gillies, M.C.; Zhou, F. Chloroquine and Hydroxychloroquine Are Novel Inhibitors of Human Organic Anion Transporting Polypeptide 1A. J. Pharm. Sci. 2016, 105, 884–890. [Google Scholar] [CrossRef] [PubMed]
- Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
- Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vincent, M.J.; Bergeron, E.; Benjannet, S.; Erickson, B.R.; Rollin, P.E.; Ksiazek, T.G.; Seidah, N.G.; Nichol, S.T. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol. J. 2005, 2, 69. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Cao, R.; Xu, M.; Wang, X.; Zhang, H.; Hu, H.; Li, Y.; Hu, Z.; Zhong, W.; Wang, M. Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov. 2020, 6, 16. [Google Scholar] [CrossRef] [Green Version]
- Van den Borne, B.E.; Dijkmans, B.A.; de Rooij, H.H.; le Cessie, S.; Verweij, C.L. Chloroquine and hydroxychloroquine equally affect tumor necrosis factor-alpha, interleukin 6, and interferon-gamma production by peripheral blood mononuclear cells. J. Rheumatol. 1997, 24, 55–60. [Google Scholar] [PubMed]
- Coronavirus (COVID-19) Update: FDA Revokes Emergency Use Authorization for Chloroquine and Hydroxychloroquine. Available online: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-revokes-emergency-use-authorization-chloroquine-and (accessed on 29 October 2021).
- Pan, H.; Peto, R.; Henao-Restrepo, A.M.; Preziosi, M.P.; Sathiyamoorthy, V.; Abdool Karim, Q.; Alejandria, M.M.; Hernández García, C.; Kieny, M.-P.; Malekzadeh, R.; et al. Repurposed Antiviral Drugs for COVID-19—Interim WHO Solidarity Trial Results. N. Engl. J. Med. 2021, 384, 497–511. [Google Scholar]
- RECOVERY Collaborative Group; Horby, P.; Mafham, M.; Linsell, L.; Bell, J.L.; Staplin, N.; Emberson, J.R.; Wiselka, M.; Ustianowski, A.; Elmahi, E.; et al. Effect of Hydroxychloroquine in Hospitalized Patients with COVID-19. N. Eng. J. Med. 2020, 383, 2030–2040. [Google Scholar]
- Furtado, R.H.M.; Berwanger, O.; Fonseca, H.A.; Corrêa, T.D.; Ferraz, L.R.; Lapa, M.G.; Zampieri, F.G.; Veiga, V.C.; Azevedo, L.C.P.; Rosa, R.G.; et al. Azithromycin in addition to standard of care versus standard of care alone in the treatment of patients admitted to the hospital with severe COVID-19 in Brazil (COALITION II): A randomised clinical trial. Lancet 2020, 396, 959–967. [Google Scholar] [CrossRef]
- Cavalcanti, A.B.; Zampieri, F.G.; Rosa, R.G.; Azevedo, L.C.; Veiga, V.C.; Avezum, A.; Damiani, L.P.; Marcadenti, A.; Kawano-Dourado, L.; Lisboa, T.; et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate COVID-19. N. Engl. J. Med. 2020, 383, 2041–2052. [Google Scholar] [CrossRef]
- Kim, M.S.; An, M.H.; Kim, W.J.; Hwang, T.-H. Comparative efficacy and safety of pharmacological interventions for the treatment of COVID-19: A systematic review and network meta-analysis. PLoS Med. 2020, 17, e1003501. [Google Scholar] [CrossRef]
- Rochwerg, B.; Siemieniuk, R.A.; Agoritsas, T.; Lamontagne, F.; Askie, L.; Lytvyn, L.; Leo, Y.-S.; Macdonald, H.; Zeng, L.; Amin, W.; et al. A living WHO guideline on drugs for COVID-19. BMJ 2020, 370, m3379. [Google Scholar] [CrossRef]
- ZITHROMAX® Pfizer, New York, NY. Available online: http://labeling.pfizer.com/ShowLabeling.aspx?format=PDF&id=511 (accessed on 29 October 2021).
- PLAQUENIL® Sanofi-aventis, Quebec, Laval. Available online: https://pdf.hres.ca/dpd_pm/00052851.PDF (accessed on 29 October 2021).
- Damle, B.; Vourvahis, M.; Wang, E.; Leaney, J.; Corrigan, B. Clinical Pharmacology Perspectives on the Antiviral Activity of Azithromycin and Use in COVID-19. Clin. Pharmacol. Ther. 2020, 108, 201–211. [Google Scholar] [CrossRef] [PubMed]
- Abella, B.S.; Jolkovsky, E.L.; Biney, B.T.; Uspal, J.E.; Hyman, M.C.; Frank, I.; Hensley, S.E.; Gill, S.; Vogl, D.T.; Maillard, I.; et al. Efficacy and Safety of Hydroxychloroquine vs Placebo for Pre-exposure SARS-CoV-2 Prophylaxis Among Health Care Workers: A Randomized Clinical Trial. JAMA Intern. Med. 2021, 181, 195–202. [Google Scholar] [CrossRef] [PubMed]
- Boulware, D.R.; Pullen, M.; Bangdiwala, A.S.; Pastick, K.A.; Lofgren, S.M.; Okafor, E.C.; Skipper, C.P.; Nascene, A.A.; Nicol, M.R.; Abassi, M.; et al. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for COVID-19. N. Engl. J. Med. 2020, 383, 517–525. [Google Scholar] [CrossRef] [PubMed]
- Bartoszko, J.J.; Siemieniuk, R.A.C.; Kum, E.; Qasim, A.; Zeraatkar, D.; Ge, L.; Han, M.A.; Sadeghirad, B.; Agarwal, A.; Agoritsas, T.; et al. Prophylaxis against COVID-19: Living systematic review and network meta-analysis. medRxiv 2021. [Google Scholar] [CrossRef] [PubMed]
- Lamontagne, F.; Agoritsas, T.; Siemieniuk, R.; Rochwerg, B.; Bartoszko, J.; Askie, L.; Macdonald, H.; Amin, W.; Bausch, F.J.; Burhan, E.; et al. A living WHO guideline on drugs to prevent COVID-19. BMJ 2021, 372, n526. [Google Scholar] [CrossRef] [PubMed]
- Members of the Consensus Conference on Treatment of Skin and Mucosal Lesions (Committee of Guideline for the Diagnosis and Treatment of Mucocutaneous Lesions of Behçet’s disease); Nakamura, K.; Iwata, Y.; Asai, J.; Kawakami, T.; Tsunemi, Y.; Takeuchi, M.; Mizuki, N.; Kaneko, F. Guidelines for the treatment of skin and mucosal lesions in Behçet’s disease: A secondary publication. J. Dermatol. 2020, 47, 223–235. [Google Scholar] [CrossRef]
- Colcrys® AR Scientifc, Inc., Philadelphia, PA. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/022351MedGuide.pdf (accessed on 29 October 2021).
- Leung, Y.Y.; Yao Hui, L.L.; Kraus, V.B. Colchicine—Update on mechanisms of action and therapeutic uses. Semin. Arthritis Rheum. 2015, 45, 341–350. [Google Scholar] [CrossRef] [Green Version]
- Chen, I.-Y.; Moriyama, M.; Chang, M.-F.; Ichinohe, T. Severe Acute Respiratory Syndrome Coronavirus Viroporin 3a Activates the NLRP3 Inflammasome. Front. Microbiol. 2019, 10, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siu, K.-L.; Yuen, K.-S.; Castano-Rodriguez, C.; Ye, Z.-W.; Yeung, M.-L.; Fung, S.-Y.; Yuan, S.; Chan, C.-P.; Yuen, K.-Y.; Enjuanes, L.; et al. Severe acute respiratory syndrome Coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. 2019, 33, 8865–8877. [Google Scholar] [CrossRef] [PubMed]
- Shi, C.-S.; Nabar, N.R.; Huang, N.-N.; Kehrl, J.H. SARS-Coronavirus Open Reading Frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Discov. 2019, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Grailer, J.J.; Canning, B.A.; Kalbitz, M.; Haggadone, M.D.; Dhond, R.M.; Andjelkovic, A.V.; Zetoune, F.S.; Ward, P.A. Critical Role for the NLRP3 Inflammasome during Acute Lung Injury. J. Immunol. 2014, 192, 5974–5983. [Google Scholar] [CrossRef] [PubMed]
- Kaksonen, M.; Roux, A. Mechanisms of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 2018, 19, 313–326. [Google Scholar] [CrossRef]
- Stebbing, J.; Phelan, A.; Griffin, I.; Tucker, C.; Oechsle, O.; Smith, D.; Richardson, P. COVID-19: Combining antiviral and anti-inflammatory treatments. Lancet Infect. Dis. 2020, 20, 400–402. [Google Scholar] [CrossRef]
- Deftereos, S.G.; Giannopoulos, G.; Vrachatis, D.A.; Siasos, G.D.; Giotaki, S.G.; Gargalianos, P.; Metallidis, S.; Sianos, G.; Baltagiannis, S.; Pnagopoulos, S.; et al. Effect of Colchicine vs Standard Care on Cardiac and Inflammatory Biomarkers and Clinical Outcomes in Patients Hospitalized with Coronavirus Disease 2019: The GRECCO-19 Randomized Clinical Trial. JAMA Netw. Open. 2020, 3, e2013136. [Google Scholar] [CrossRef]
- Tardif, J.-C.; Bouabdallaoui, N.; L’Allier, P.L.; Gaudet, D.; Shah, B.; Pillinger, M.H.; Lopez-Sendon, J.; da Luz, P.; Verret, L.; Audet, S.; et al. Efficacy of Colchicine in Non-Hospitalized Patients with COVID-19. medRxiv 2021. [Google Scholar] [CrossRef]
- Novartis. NEORAL® Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/050715s027,050716s028lbl.pdf (accessed on 29 October 2021).
- Tacrolimus® Pfizer, Tokyo. Available online: https://www.info.pmda.go.jp/go/pack/3999014M1030_1_16 (accessed on 29 October 2021).
- Liu, J.; Farmer, J.D.; Lane, W.S.; Friedman, J.; Weissman, I.; Schreiber, S.L. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell 1991, 66, 807–815. [Google Scholar] [CrossRef]
- Liddicoat, A.M.; Lavelle, E.C. Modulation of innate immunity by cyclosporine A. Biochem. Pharmacol. 2019, 163, 472–480. [Google Scholar] [CrossRef]
- De Wilde, A.H.; Zevenhoven-Dobbe, J.C.; van der Meer, Y.; Thiel, V.; Narayanan, K.; Makino, S.; Snijder, E.J.; van Hemert, M.J. Cyclosporin A inhibits the replication of diverse coronaviruses. J. Gen. Virol. 2011, 92 Pt 11, 2542–2548. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, Y.; Sato, Y.; Sasaki, T. Suppression of Coronavirus Replication by Cyclophilin Inhibitors. Viruses 2013, 5, 1250–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.S.; Kuok, D.I.T.; Cheung, M.C.; Ng, M.M.T.; Ng, K.C.; Hui, K.P.Y.; Peiris, J.S.M.; Chan, M.C.W.; Nicholls, J.M. Effect of interferon alpha and cyclosporine treatment separately and in combination on Middle East Respiratory Syndrome Coronavirus (MERS-CoV) replication in a human in-vitro and ex-vivo culture model. Antiviral Res. 2018, 155, 89–96. [Google Scholar] [CrossRef]
- Carbajo-Lozoya, J.; Müller, M.A.; Kallies, S.; Thiel, V.; Drosten, C.; von Brunn, A. Replication of human coronaviruses SARS-CoV, HCoV-NL63 and HCoV-229E is inhibited by the drug FK506. Virus Res. 2012, 165, 112–117. [Google Scholar] [CrossRef] [PubMed]
- Galvez-Romero, J.L.; Palmeros-Rojas, O.; Real-Ramírez, F.A.; Sánchez-Romero, S.; Tome-Maxil, R.; Ramírez-Sandoval, M.P.; Olivos-Rodríguez, R.; Flores-Encarnación, S.E.; Cabrera-Estrada, A.A.; Ávila-Morales, J.; et al. Cyclosporine A plus low-dose steroid treatment in COVID-19 improves clinical outcomes in patients with moderate to severe disease: A pilot study. J. Intern. Med. 2021, 289, 906–920. [Google Scholar] [CrossRef] [PubMed]
- Belli, L.S.; Fondevila, C.; Cortesi, P.A.; Conti, S.; Karam, V.; Adam, R.; Coilly, A.; Ericzon, B.G.; Loinaz, C.; Cuervas-Mons, V.; et al. Protective Role of Tacrolimus, Deleterious Role of Age and Comorbidities in Liver Transplant Recipients with COVID-19: Results From the ELITA/ELTR Multi-center European Study. Gastroenterology 2021, 160, 1151–1163. [Google Scholar] [CrossRef] [PubMed]
- Solanich, X.; Antolí, A.; Padullés, N.; Fanlo-Maresma, M.; Iriarte, A.; Mitjavila, F.; Capdevila, O.; Molina, M.; Sabater, J.; Bas, J.; et al. Pragmatic, open-label, single-center, randomized, phase II clinical trial to evaluate the efficacy and safety of methylprednisolone pulses and tacrolimus in patients with severe pneumonia secondary to COVID-19: The TACROVID trial protocol. Contemp. Clin. Trials Commun. 2021, 21, 100716. [Google Scholar] [CrossRef] [PubMed]
- Hirano, Y.; Madokoro, S.; Kondo, Y.; Okamoto, K.; Tanaka, H. Corticosteroid treatment for early acute respiratory distress syndrome: A systematic review and meta-analysis of randomized trials. J. Intensive Care 2020, 8, 1–8. [Google Scholar] [CrossRef]
- Sterne, J.A.C.; Murthy, S.; Diaz, J.V.; Slutsky, A.S.; Villar, J.; Angus, D.C.; Annane, D.; Pontes Azavedo, L.C.; Berwanger, O.; Cavalcanti, A.B.; et al. Association Between Administration of Systemic Corticosteroids and Mortality Among Critically Ill Patients With COVID-19: A Meta-analysis. JAMA 2020, 324, 1330–1341. [Google Scholar] [PubMed]
- Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; Elmahi, E.; et al. Dexamethasone in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar]
- National Institutes of Health. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines; Therapeutic Management of Hospitalized Adults with COVID-19. Available online: https://www.covid19treatmentguidelines.nih.gov/management/clinical-management/hospitalized-adults--therapeutic-management (accessed on 29 October 2021).
- Jeronimo, C.M.P.; Farias, M.E.L.; Val, F.F.A.; Sampaio, V.S.; Alexandre, M.A.A.; Melo, G.C.; Safe, I.P.; Borba, M.G.S.; Netto, R.L.A.; Maciel, A.B.S.; et al. Methylprednisolone as Adjunctive Therapy for Patients Hospitalized with Coronavirus Disease 2019 (COVID-19; Metcovid): A Randomized, Double-blind, Phase IIb, Placebo-controlled Trial. Clin. Infect. Dis. 2021, 72, e373–e381. [Google Scholar] [CrossRef] [PubMed]
- Tomazini, B.M.; Maia, I.S.; Cavalcanti, A.B.; Berwanger, O.; Rosa, R.G.; Veiga, V.C.; Avezum, A.; Lopes, R.D.; Bueno, F.R.; Silva, M.V.A.O.; et al. Effect of Dexamethasone on Days Alive and Ventilator-Free in Patients with Moderate or Severe Acute Respiratory Distress Syndrome and COVID-19: The CoDEX Randomized Clinical Trial. JAMA 2020, 324, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
- Dequin, P.F.; Heming, N.; Meziani, F.; Plantefève, G.; Voiriot, G.; Badié, J.; Francois, B.; Aubron, C.; Ricard, J.-D.; Ehrmann, S.; et al. Effect of Hydrocortisone on 21-Day Mortality or Respiratory Support Among Critically Ill Patients With COVID-19: A Randomized Clinical Trial. JAMA 2020, 324, 1298–1306. [Google Scholar] [CrossRef] [PubMed]
- Angus, D.C.; Derde, L.; Al-Beidh, F.; Annane, D.; Arabi, Y.; Beane, A.; van Bentum-Puijk, W.; Berry, L.; Bhimani, Z.; Bonten, M.; et al. Effect of Hydrocortisone on Mortality and Organ Support in Patients with Severe COVID-19: The REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA 2020, 324, 1317–1329. [Google Scholar] [PubMed]
- Edalatifard, M.; Akhtari, M.; Salehi, M.; Naderi, Z.; Jamshidi, A.; Mostafaei, S.; Najafizadeh, S.R.; Farhadi, E.; Jalili, N.; Esfahani, M.; et al. Intravenous methylprednisolone pulse as a treatment for hospitalised severe COVID-19 patients: Results from a randomised controlled clinical trial. Eur. Respir. J. 2020, 56, 2002808. [Google Scholar] [CrossRef] [PubMed]
- Arabi, Y.M.; Mandourah, Y.; Al-Hameed, F.; Sindi, A.A.; Almekhlafi, G.A.; Hussein, M.A.; Jose, J.; Pinto, R.; Al-Omari, A.; Kharaba, A.; et al. Corticosteroid Therapy for Critically Ill Patients with Middle East Respiratory Syndrome. Am. J. Respir. Crit. Care Med. 2018, 197, 757–767. [Google Scholar] [CrossRef] [PubMed]
- Stockman, L.J.; Bellamy, R.; Garner, P. SARS: Systematic Review of Treatment Effects. PLoS Med. 2006, 3, e343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodrigo, C.; Leonardi-Bee, J.; Nguyen-Van-Tam, J.; Lim, W.S. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst. Rev. 2013, 3, Cd010406. [Google Scholar] [CrossRef] [Green Version]
- Spagnuolo, V.; Guffanti, M.; Galli, L.; Poli, A.; Querini, P.R.; Ripa, M.; Clementi, M.; Scarpellini, P.; Lazzarin, A.; Tresoldi, M.; et al. Viral clearance after early corticosteroid treatment in patients with moderate or severe COVID-19. Sci. Rep. 2020, 10, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Wölfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Müller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-19. Nature 2020, 581, 465–469. [Google Scholar] [CrossRef] [Green Version]
- He, X.; Lau, E.H.Y.; Wu, P.; Deng, X.; Wang, J.; Hao, X.; Lau, Y.C.; Wong, J.Y.; Guan, Y.; Tan, X.; et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat. Med. 2020, 26, 672–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corticosteroids. Available online: https://www.covid19treatmentguidelines.nih.gov/immunomodulators/corticosteroids (accessed on 29 October 2021).
- Liu, J.; Wang, T.; Cai, Q.; Sun, L.; Huang, D.; Zhou, G.; He, Q.; Wang, F.; Liu, L.; Chen, J. Longitudinal changes of liver function and hepatitis B reactivation in COVID-19 patients with pre-existing chronic hepatitis B virus infection. Hepatol. Res. 2020, 50, 1211–1221. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-Tajes, S.; Miralpeix, A.; Costa, J.; López-Suñé, E.; Laguno, M.; Pocurull, A.; Lens, S.; Mariño, Z.; Forns, X. Low risk of hepatitis B reactivation in patients with severe COVID-19 who receive immunosuppressive therapy. J. Viral Hepat. 2021, 28, 89–94. [Google Scholar] [CrossRef] [PubMed]
- Mulchandani, R.; Lyngdoh, T.; Kakkar, A.K. Deciphering the COVID-19 cytokine storm: Systematic review and meta-analysis. Eur. J. Clin. Investig. 2021, 51, e13429. [Google Scholar] [CrossRef] [PubMed]
- Coomes, E.A.; Haghbayan, H. Interleukin-6 in COVID-19: A systematic review and meta-analysis. Rev. Med. Virol. 2020, 30, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Leisman, D.E.; Ronner, L.; Pinotti, R.; Taylor, M.D.; Sinha, P.; Calfee, C.S.; Hirayama, A.V.; Mastroiani, F.; Turtle, C.J.; Harhay, M.O.; 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, 8, 1233–1244. [Google Scholar] [CrossRef]
- Bohn, M.K.; Hall, A.; Sepiashvili, L.; Jung, B.; Steele, S.; Adeli, K. Pathophysiology of COVID-19: Mechanisms Underlying Disease Severity and Progression. Physiology 2020, 35, 288–301. [Google Scholar] [CrossRef] [PubMed]
- ACTEMRA® Chugai Pharmaceutical Co., Ltd. Tokyo, Japan. Available online: https://www.info.pmda.go.jp/go/pack/6399421A1020_1_18 (accessed on 29 October 2021).
- ACTEMRA® Genentech, Inc. South San Francisco, CA. Available online: https://www.gene.com/download/pdf/actemra_prescribing.pdf (accessed on 29 October 2021).
- EUSA Pharma. SYLVANT® (Siltuximab). 2020. Available online: https://www.sylvant.com/en-gb (accessed on 29 October 2021).
- Hermine, O.; Mariette, X.; Tharaux, P.L.; Resche-Rigon, M.; Porcher, R.; Ravaud, P. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern. Med. 2021, 181, 32–40. [Google Scholar] [CrossRef] [PubMed]
- Rosas, I.O.; Bräu, N.; Waters, M.; Go, R.C.; Hunter, B.D.; Bhagani, S.; Skiest, D.; Aziz, M.S.; Cooper, N.; Douglas, I.S.; et al. Tocilizumab in Hospitalized Patients with Severe COVID-19 Pneumonia. N. Engl. J. Med. 2021, 384, 1503–1516. [Google Scholar] [CrossRef] [PubMed]
- WHO R & D Blueprint. Novel Coronavirus. COVID-19 Therapeutic Trial Synopsis. Available online: https://www.who.int/blueprint/priority-diseases/key-action/COVID-19_Treatment_Trial_Design_Master_Protocol_synopsis_Final_18022020.pdf (accessed on 29 October 2021).
- Roche. Roche’s Phase III EMPACTA Study Showed Actemra/RoActemra Reduced the Likelihood of Needing Mechanical Ventilation in Hospitalised Patients with COVID-19 Associated Pneumonia. Available online: https://www.roche.com/media/releases/med-cor-2020-09-18.htm (accessed on 29 October 2021).
- The REMAP-CAP Investigators VOPCG; Gordon, A.C.; Mouncey, P.R.; Al-Beidh, F.; Kathryn, M.; Alistair, R.; Nichol, D.; Yaseen, M.A.; Annane, D.; Beane, A.; et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with COVID-19—Preliminary report. medRxiv 2021. [Google Scholar] [CrossRef]
- A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect. Dis. 2020, 20, e192–e197. [CrossRef]
- Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. Lancet 2021, 397, 1637–1645. [CrossRef]
- Sanofi and Regeneron Provide Update on Kevzara® (Sarilumab) Phase 3 U.S. Trial in COVID-19 Patients. Available online: https://newsroom.regeneron.com/news-releases/news-release-details/regeneron-and-sanofi-provide-update-kevzarar-sarilumab-phase-3 (accessed on 29 October 2021).
- Lescure, F.-X.; Honda, H.; Fowler, R.A.; Lazar, J.S.; Shi, G.; Wung, P.; Patel, N.; Hagino, O.; Bazzalo, I.J.; Casas, M.M.; et al. Sarilumab in patients admitted to hospital with severe or critical COVID-19: A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 2021, 9, 522–532. [Google Scholar] [CrossRef]
- Interim Clinical Commissioning Policy: Tocilizumab for Hospitalised Patients with COVID-19 Pneumonia (Adults). Available online: https://www.england.nhs.uk/coronavirus/wp-content/uploads/sites/52/2021/02/C1143-interim-clinical-commissioning-policy-tocilizumab-rps-v2.pdf (accessed on 29 October 2021).
- National Institutes of Health. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines; Interleukin-6 Inhibitors. Available online: https://www.covid19treatmentguidelines.nih.gov/therapies/immunomodulators/interleukin-6-inhibitors (accessed on 29 October 2021).
- Roche Provides Update on the Phase III REMDACTA Trial of Actemra/RoActemra Plus Veklury in Patients with Severe COVID-19 Pneumonia. Available online: https://www.roche.com/media/releases/med-cor-2021-03-11.htm (accessed on 29 October 2021).
- Gritti, G.; Raimondi, F.; Ripamonti, D.; Riva, I.; Landi, F.; Alborghetti, L.; Frigeni, M.; Damiani, M.; Micò, C.; Fagiuoli, S.; et al. IL-6 signalling pathway inactivation with siltuximab in patients with COVID-19 respiratory failure: An observational cohort study. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Stone, J.H.; Frigault, M.J.; Serling-Boyd, N.J.; Fernandes, A.D.; Harvey, L.; Foulkes, A.S.; Horick, N.K.; Healy, B.C.; Shah, R.; Bensaci, A.M.; et al. Efficacy of Tocilizumab in Patients Hospitalized with COVID-19. N. Engl. J. Med. 2020, 383, 2333–2344. [Google Scholar] [CrossRef] [PubMed]
- Food and Drug Administration. Anakinra® (Kineret) Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/103950s5136lbl.pdf (accessed on 29 October 2021).
- Otsuka, R.; Seino, K.I. Macrophage activation syndrome and COVID-19. Inflamm. Regen. 2020, 40, 19. [Google Scholar] [CrossRef] [PubMed]
- Eloseily, E.M.; Weiser, P.; Crayne, C.B.; Haines, H.; Mannion, M.L.; Stoll, M.L.; Beukelman, T.; Atkinson, T.P.; Cron, R.Q. Benefit of Anakinra in Treating Pediatric Secondary Hemophagocytic Lymphohistiocytosis. Arthritis Rheumatol. 2020, 72, 326–334. [Google Scholar] [CrossRef] [PubMed]
- Fearon, W.F.; Fearon, D.T. Inflammation and cardiovascular disease: Role of the interleukin-1 receptor antagonist. Circulation 2008, 117, 2577–2579. [Google Scholar] [CrossRef] [Green Version]
- Guzik, T.J.; Mohiddin, S.A.; DiMarco, A.; Patel, V.; Savvatis, K.; Marelli-Berg, F.M.; Madhur, M.S.; Tomaszewski, M.; Maffia, P.; D’Acquisto, F.; et al. COVID-19 and the cardiovascular system: Implications for risk assessment, diagnosis, and treatment options. Cardiovasc. Res. 2020, 116, 1666–1687. [Google Scholar] [CrossRef] [PubMed]
- Novartis Pharmaceuticals Corporation. ILARIS® HIGHLIGHTS OF PRESCRIBING INFORMATION. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/BLA125319_858687lbl.pdf (accessed on 29 October 2021).
- Cavalli, G.; Larcher, A.; Tomelleri, A.; Campochiaro, C.; Della-Torre, E.; De Luca, G.; Farina, N.; Boffini, N.; Ruggeri, A.; Poli, A.; et al. Interleukin-1 and interleukin-6 inhibition compared with standard management in patients with COVID-19 and hyperinflammation: A cohort study. Lancet Rheumatol. 2021, 3, e253–e261. [Google Scholar] [CrossRef]
- Balkhair, A.; Al-Zakwani, I.; Al Busaidi, M.; Al-Khirbash, A.; Al Mubaihsi, S.; BaTaher, H.; Al Aghbari, J.; Al Kindi, M.; Baawain, S.; Al Alawi, A.; et al. Anakinra in hospitalized patients with severe COVID-19 pneumonia requiring oxygen therapy: Results of a prospective, open-label, interventional study. Int. J. Infect. Dis. 2021, 103, 288–296. [Google Scholar] [CrossRef]
- CORIMUNO-19 Collaborative Group. Effect of anakinra versus usual care in adults in hospital with COVID-19 and mild-to-moderate pneumonia (CORIMUNO-ANA-1): A randomised controlled trial. Lancet Respir. Med. 2021, 9, 295–304. [Google Scholar] [CrossRef]
- The REMAP-CAP Investigators; Derde, L.P.G.; Gordon, A.C.; Mouncey, P.R.; Al-Beidh, F.; Rowan, K.M.; Nichol, A.D.; Arabi, Y.M.; Annane, D.; Beane, A.; et al. Effectiveness of Tocilizumab, Sarilumab, and Anakinra for critically ill patients with COVID-19 The REMAP-CAP COVID-19 Immune Modulation Therapy Domain Randomized Clinical Trial. medRxiv 2021. [Google Scholar] [CrossRef]
- Caricchio, R.; Abbate, A.; Gordeev, I.; Meng, J.; Hsue, P.Y.; Neogi, T.; Ardulino, R.; Fomina, D.; Bogdanov, R.; Stepanenko, T.; et al. Effect of Canakinumab vs Placebo on Survival Without Invasive Mechanical Ventilation in Patients Hospitalized with Severe COVID-19: A Randomized Clinical Trial. JAMA 2021, 326, 230–239. [Google Scholar] [CrossRef] [PubMed]
- O’Shea, J.J.; Pesu, M.; Borie, D.C.; Changelian, P.S. A new modality for immunosuppression: Targeting the JAK/STAT pathway. Nat. Rev. Drug Discov. 2004, 3, 555–564. [Google Scholar] [CrossRef] [PubMed]
- Leonard, W.J.; O’Shea, J.J. JAKS AND STATS: Biological Implications. Annu. Rev. Immunol. 1998, 16, 293–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghoreschi, K.; Laurence, A.; O’Shea, J.J. Janus kinases in immune cell signaling. Immunol. Rev. 2009, 228, 273–287. [Google Scholar] [CrossRef] [Green Version]
- Winthrop, K.L. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat. Rev. Rheumatol. 2017, 13, 234–243. [Google Scholar] [CrossRef]
- O’Shea, J.J.; Laurence, A.; McInnes, I. Back to the future: Oral targeted therapy for RA and other autoimmune diseases. Nat. Rev. Rheumatol. 2013, 9, 173–182. [Google Scholar] [CrossRef] [PubMed]
- Angelini, J.; Talotta, R.; Roncato, R.; Fornasier, G.; Barbiero, G.; Dal Cin, L.; Brancati, S.; Scaglione, F. JAK-Inhibitors for the Treatment of Rheumatoid Arthritis: A Focus on the Present and an Outlook on the Future. Biomolecules 2020, 10, 1002. [Google Scholar] [CrossRef] [PubMed]
- Feldmann, M.; Maini, R.N.; Woody, J.N.; Holgate, S.T.; Winter, G.; Rowland, M.; Richards, D.; Hussell, T. Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed. Lancet 2020, 395, 1407–1409. [Google Scholar] [CrossRef]
- Luo, W.; Li, Y.-X.; Jiang, L.-J.; Chen, Q.; Wang, T.; Ye, D.-W. Targeting JAK-STAT Signaling to Control Cytokine Release Syndrome in COVID-19. Trends Pharmacol. Sci. 2020, 41, 531–543. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Zhang, Y.; Qiao, W.; Zhang, J.; Qi, Z. Baricitinib, a drug with potential effect to prevent SARS-COV-2 from entering target cells and control cytokine storm induced by COVID-19. Int. Immunopharmacol. 2020, 86, 106749. [Google Scholar] [CrossRef]
- Mesev, E.V.; Ledesma, R.A.; Ploss, A. Decoding type I and III interferon signalling during viral infection. Nat. Microbiol. 2019, 4, 914–924. [Google Scholar] [CrossRef] [PubMed]
- Kang, S.; Brown, H.M.; Hwang, S. Direct Antiviral Mechanisms of Interferon-Gamma. Immune Netw. 2018, 18, e33. [Google Scholar] [CrossRef] [PubMed]
- Ziegler, C.G.K.; Allon, S.J.; Nyquist, S.K.; Mbano, I.M.; Miao, V.N.; Tzouanas, C.N.; Cao, Y.; Yousif, A.S.; Bals, J.; Hauser, B.M.; et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues. Cell 2020, 181, 1016–1035.e19. [Google Scholar] [CrossRef]
- Verden, A.; Dimbil, M.; Kyle, R.; Overstreet, B.; Hoffman, K.B. Analysis of Spontaneous Postmarket Case Reports Submitted to the FDA Regarding Thromboembolic Adverse Events and JAK Inhibitors. Drug Saf. 2017, 41, 357–361. [Google Scholar] [CrossRef] [PubMed]
- Teuwen, L.A.; Geldhof, V.; Pasut, A.; Carmeliet, P. COVID-19: The vasculature unleashed. Nat. Rev. Immunol. 2020, 20, 389–391. [Google Scholar] [CrossRef] [PubMed]
- Marietta, M.; Coluccio, V.; Luppi, M. COVID-19, coagulopathy and venous thromboembolism: More questions than answers. Intern. Emerg. Med. 2020, 15, 1375–1387. [Google Scholar] [CrossRef] [PubMed]
- Ozolina, A.; Sarkele, M.; Sabelnikovs, O.; Skesters, A.; Jaunalksne, I.; Serova, J.; Ievins, T.; Bjertnaes, L.J.; Vanags, I. Activation of Coagulation and Fibrinolysis in Acute Respiratory Distress Syndrome: A Prospective Pilot Study. Front. Med. 2016, 3, 64. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Yan, X.; Fan, Q.; Liu, H.; Liu, X.; Liu, Z.; Zhang, Z. D-dimer levels on admission to predict in-hospital mortality in patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1324–1329. [Google Scholar] [CrossRef] [PubMed]
- Obi, A.T.; Barnes, G.D.; Napolitano, L.M.; Henke, P.K.; Wakefield, T.W. Venous thrombosis epidemiology, pathophysiology, and anticoagulant therapies and trials in severe acute respiratory syndrome coronavirus 2 infection. J. Vasc. Surgery Venous Lymphat. Disord. 2021, 9, 23–35. [Google Scholar] [CrossRef]
- Rodriguez-Garcia, J.L.; Sanchez-Nievas, G.; Arevalo-Serrano, J.; Garcia-Gomez, C.; Jimenez-Vizuete, J.M.; Martinez-Alfaro, E. Baricitinib improves respiratory function in patients treated with corticosteroids for SARS-CoV-2 pneumonia: An observational cohort study. Rheumatology 2021, 60, 399–407. [Google Scholar] [CrossRef]
- Richardson, P.; Griffin, I.; Tucker, C.; Smith, D.; Oechsle, O.; Phelan, A.; Rawling, M.; Savory, E.; Stebbing, J. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet 2020, 395, e30–e31. [Google Scholar] [CrossRef] [Green Version]
- Kalil, A.C.; Patterson, T.F.; Mehta, A.K.; Tomashek, K.M.; Wolfe, C.R.; Ghazaryan, V.; Marconi, V.C.; Ruiz-Palacios, G.M.; Hsieh, L.; Kline, S.; et al. Baricitinib plus Remdesivir for Hospitalized Adults with COVID-19. N. Engl. J. Med. 2021, 384, 795–807. [Google Scholar] [CrossRef]
- Marconi, V.C.; Ramanan, A.V.; de Bono, S.; Kartman, C.E.; Krishnan, V.; Liao, R.; Piruzeli, M.L.B.; Goldman, J.D.; Alatorre-Alexander, J.; Pellegrini, R.D.C.; et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): A randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir. Med. 2021. [Google Scholar] [CrossRef]
- Robinson, P.C.; Liew, D.F.; Liew, J.W.; Monaco, C.; Richards, D.; Shivakumar, S.; Tanner, H.L.; Feldmann, M. The Potential for Repurposing Anti-TNF as a Therapy for the Treatment of COVID-19. Med 2020, 1, 90–102. [Google Scholar] [CrossRef]
- Oikonomou, N.; Harokopos, V.; Zalevsky, J.; Valavanis, C.; Kotanidou, A.; Szymkowski, D.; Kollias, G.; Aidinis, V. Soluble TNF Mediates the Transition from Pulmonary Inflammation to Fibrosis. PLoS ONE 2006, 1, e108. [Google Scholar] [CrossRef]
- Li, G.; Fan, Y.; Lai, Y.; Han, T.; Li, Z.; Zhou, P.; Pan, P.; Wang, W.; Hu, D.; Liu, X.; et al. Coronavirus infections and immune responses. J. Med. Virol. 2020, 92, 424–432. [Google Scholar] [CrossRef] [PubMed]
- Brito, C.A.; Paiva, J.G.; Pimentel, F.N.; Guimarães, R.S.; Moreira, M.R. COVID-19 in patients with rheumatological diseases treated with anti-TNF. Ann. Rheum. Dis. 2021, 80, e62. [Google Scholar] [CrossRef]
- Ingegnoli, F.; Fantini, F.; Favalli, E.G.; Soldi, A.; Griffini, S.; Galbiati, V.; Meroni, P.L.; Cugno, M. Inflammatory and prothrombotic biomarkers in patients with rheumatoid arthritis: Effects of tumor necrosis factor-α blockade. J. Autoimmun. 2008, 31, 175–179. [Google Scholar] [CrossRef]
- McDermott, J.E.; Mitchell, H.D.; Gralinski, L.E.; Eisfeld, A.J.; Josset, L.; Bankhead, A., 3rd; Neumann, G.; Tilton, S.C.; Schafer, A.; Li, C.; et al. The effect of inhibition of PP1 and TNFα signaling on pathogenesis of SARS coronavirus. BMC Syst. Biol. 2016, 10, 93. [Google Scholar] [CrossRef] [Green Version]
- Gianfrancesco, M.; Hyrich, K.L.; Al-Adely, S.; Carmona, L.; Danila, M.I.; Gossec, L.; Izadi, Z.; Jacobsohn, L.; Katz, P.; Lawson-Tovey, S.; et al. Characteristics associated with hospitalisation for COVID-19 in people with rheumatic disease: Data from the COVID-19 Global Rheumatology Alliance physician-reported registry. Ann. Rheum. Dis. 2020, 79, 859–866. [Google Scholar] [CrossRef]
- Stallmach, A.; Kortgen, A.; Gonnert, F.; Coldewey, S.M.; Reuken, P.; Bauer, M. Infliximab against severe COVID-19-induced cytokine storm syndrome with organ failure—a cautionary case series. Crit. Care 2020, 24, 1–3. [Google Scholar] [CrossRef]
- Bristol-Myers Squibb Company. ORENCIA® (Abatacept) HIGHLIGHTS OF PRESCRIBING INFORMATION. Available online: https://packageinserts.bms.com/pi/pi_orencia.pdf (accessed on 29 October 2021).
- Stephen-Victor, E.; Das, M.; Karnam, A.; Pitard, B.; Gautier, J.-F.; Bayry, J. Potential of regulatory T-cell-based therapies in the management of severe COVID-19. Eur. Respir. J. 2020, 56, 2002182. [Google Scholar] [CrossRef]
- Li, H.; Zuo, J.; Tang, W. Phosphodiesterase-4 Inhibitors for the Treatment of Inflammatory Diseases. Front. Pharmacol. 2018, 9, 1048. [Google Scholar] [CrossRef] [Green Version]
- Schafer, P.; Parton, A.; Gandhi, A.; Capone, L.; Adams, M.; Wu, L.; Bartlett, J.; Loveland, M.; Gilhar, A.; Cheung, Y.-F.; et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br. J. Pharmacol. 2010, 159, 842–855. [Google Scholar] [CrossRef] [Green Version]
- Dalamaga, M.; Karampela, I.; Mantzoros, C.S. Commentary: Phosphodiesterase 4 inhibitors as potential adjunct treatment targeting the cytokine storm in COVID-19. Metabolism 2020, 109, 154282. [Google Scholar] [CrossRef]
- Santaniello, A.; Vigone, B.; Beretta, L. Letter to the editor: Immunomodulation by phosphodiesterase-4 inhibitor in COVID-19 patients. Metabolism 2020, 110, 154300. [Google Scholar] [CrossRef]
- Implementation of the Randomized Embedded Multifactorial Adaptive Platform for COVID-19 (REMAP-COVID) trial in a US health system-lessons learned and recommendations. Trials 2021, 22, 100. [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kawazoe, M.; Kihara, M.; Nanki, T. Antirheumatic Drugs against COVID-19 from the Perspective of Rheumatologists. Pharmaceuticals 2021, 14, 1256. https://doi.org/10.3390/ph14121256
Kawazoe M, Kihara M, Nanki T. Antirheumatic Drugs against COVID-19 from the Perspective of Rheumatologists. Pharmaceuticals. 2021; 14(12):1256. https://doi.org/10.3390/ph14121256
Chicago/Turabian StyleKawazoe, Mai, Mari Kihara, and Toshihiro Nanki. 2021. "Antirheumatic Drugs against COVID-19 from the Perspective of Rheumatologists" Pharmaceuticals 14, no. 12: 1256. https://doi.org/10.3390/ph14121256
APA StyleKawazoe, M., Kihara, M., & Nanki, T. (2021). Antirheumatic Drugs against COVID-19 from the Perspective of Rheumatologists. Pharmaceuticals, 14(12), 1256. https://doi.org/10.3390/ph14121256