Therapeutic advances in COVID-19

Naoka Murakami, Robert Hayden, Thomas Hills, Hanny Al-Samkari, Jonathan Casey, Lorenzo Del Sorbo, Patrick R Lawler, Meghan E Sise, David E Leaf, Naoka Murakami, Robert Hayden, Thomas Hills, Hanny Al-Samkari, Jonathan Casey, Lorenzo Del Sorbo, Patrick R Lawler, Meghan E Sise, David E Leaf

Abstract

Over 2 years have passed since the start of the COVID-19 pandemic, which has claimed millions of lives. Unlike the early days of the pandemic, when management decisions were based on extrapolations from in vitro data, case reports and case series, clinicians are now equipped with an armamentarium of therapies based on high-quality evidence. These treatments are spread across seven main therapeutic categories: anti-inflammatory agents, antivirals, antithrombotics, therapies for acute hypoxaemic respiratory failure, anti-SARS-CoV-2 (neutralizing) antibody therapies, modulators of the renin-angiotensin-aldosterone system and vitamins. For each of these treatments, the patient population characteristics and clinical settings in which they were studied are important considerations. Although few direct comparisons have been performed, the evidence base and magnitude of benefit for anti-inflammatory and antiviral agents clearly outweigh those of other therapeutic approaches such as vitamins. The emergence of novel variants has further complicated the interpretation of much of the available evidence, particularly for antibody therapies. Importantly, patients with acute and chronic kidney disease were under-represented in many of the COVID-19 clinical trials, and outcomes in this population might differ from those reported in the general population. Here, we examine the clinical evidence for these therapies through a kidney medicine lens.

Conflict of interest statement

H.A.-S. reports no disclosures relevant to the manuscript; universal disclosures include consultancy (Agios, Dova/Sobi, argenx, Rigel, Novartis, Forma, Moderna) and research funding (Agios, Dova/Sobi, Amgen). P.R.L. is an investigator in the REMAP-CAP ACE2 RAS Domain, which is investigating renin–angiotensin–system-modulating treatments for COVID-19, is supported by a Heart and Stroke Foundation of Canada National New Investigator Award, and has received unrelated consulting honoraria from Novartis, CorEvitas, and Brigham and Women’s Hospital (Boston, MA, USA), as well as unrelated royalties from McGraw-Hill Publishing. L.D.S. is inventor of a patent licensed to SQI Diagnostic and has received unrelated research funding from the Canadian Institutes of Health Research. M.E.S. has received research funding from Gilead Sciences awarded to her institution; additional, unrelated disclosures include research funding from AbbVie, Merck, EMD-Serono, Angion and serving as a scientific advisory board member for Travere and Mallinckrodt. The other authors declare no competing interests.

© 2022. Springer Nature Limited.

Figures

Fig. 1. Classes of therapies for COVID-19.
Fig. 1. Classes of therapies for COVID-19.
Therapies for COVID-19 can be broadly categorized as targeting the host response to infection (including inflammation, thrombosis, acute respiratory distress syndrome (ARDS) and renin–angiotensin–aldosterone system (RAAS) activation) or targeting the virus directly (including direct antivirals and antibody-based therapies). SARS-CoV-2 infection can lead to hyperinflammation characterized by abundant circulating levels of pro-inflammatory cytokines such as IL-6. Therapies targeting inflammation include immunosuppressive drugs such as glucocorticoids (for example, dexamethasone) and anti-IL-6 receptor antibodies (for example, tocilizumab). Several antithrombotic therapies have also been trialled to address the haemostatic and thrombotic complications associated with COVID-19, whereas different methods of oxygen delivery and intubation can be employed to treat patients with ARDS. COVID-19 can also disrupt RAAS homeostasis and drugs such as angiotensin-converting enzyme (ACE) inhibitors or angiotensin II receptor blockers are being investigated as potential therapies. Finally, therapies targeting SARS-CoV-2 directly include antivirals that disrupt viral replication and neutralizing antibody therapies that prevent virus entry into host cells.
Fig. 2. Timeline of publication of pivotal…
Fig. 2. Timeline of publication of pivotal phase III randomized clinical trials of COVID-19 therapies.
Timeline of publication and key features of pivotal phase III randomized clinical trials of COVID-19 therapies. These trials are categorized into six treatment categories: anti-inflammatory agents, antivirals, renin–angiotensin–aldosterone system (RAAS) modification, antithrombotic agents, convalescent plasma and monoclonal antibody therapies. AC, anticoagulation; ACEi, angiotensin-converting enzyme inhibitor; ARB, angiotensin II receptor blocker; d, days; ECMO, extracorporeal membrane oxygenation; ICU, intensive care unit; IMV, invasive mechanical ventilation; NIV, noninvasive ventilation; outpts, outpatients; pts, patients. This timeline reflects published data as of 29 May 2022.
Fig. 3. Anti-inflammatory and antiviral agents for…
Fig. 3. Anti-inflammatory and antiviral agents for COVID-19 including dose adjustment for kidney function impairment.
The immunosuppressive therapies dexamethasone and tocilizumab can be used without dose adjustments in patients with kidney disease, including those with kidney failure. However, the anti-inflammatory agent baricitinib must be administered at reduced doses in patients with estimated glomerular filtration rate (eGFR) 2. In the case of antivirals used to treat SARS-CoV-2 infection, molnupiravir can be used without dose adjustments and remdesivir, although currently not recommended for use in patients with eGFR < 30 ml/min/1.73 m2, has been reportedly used in patients across the spectrum of kidney dysfunction, including in patients with kidney failure who require kidney replacement therapy (KRT). By contrast, nirmatrelvir–ritonavir is contraindicated in patients with eGFR <30 ml/min/1.73 m2 and, given its potential to increase exposure to calcineurin inhibitors (CNIs) and mammalian target of rapamycin (mTOR) inhibitors, must be used with extreme caution in recipients of solid organ transplants, and only if CNI and/or mTOR levels can be monitored closely. The asterisk indicates that in patients with eGFR 30–59 ml/min/1.73 m2, the dose of nirmatrelvir is reduced by 50% but the ritonavir dose remains unchanged.

References

    1. Karim SSA, Karim QA. Omicron SARS-CoV-2 variant: a new chapter in the COVID-19 pandemic. Lancet. 2021;398:2126–2128. doi: 10.1016/S0140-6736(21)02758-6.
    1. Flythe JE, et al. Characteristics and outcomes of individuals with pre-existing kidney disease and COVID-19 admitted to intensive care units in the United States. Am. J. Kidney Dis. 2021;77:190–203.e1. doi: 10.1053/j.ajkd.2020.09.003.
    1. Massie AB, et al. Quantifying excess deaths among solid organ transplant recipients in the COVID-19 era. Am. J. Transplant. 2022;22:2077–2082. doi: 10.1111/ajt.17036.
    1. Chan L, et al. AKI in hospitalized patients with COVID-19. J. Am. Soc. Nephrol. 2021;32:151–160. doi: 10.1681/ASN.2020050615.
    1. Gupta S, et al. AKI treated with renal replacement therapy in critically ill patients with COVID-19. J. Am. Soc. Nephrol. 2021;32:161–176. doi: 10.1681/ASN.2020060897.
    1. Mehta P, et al. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033–1034. doi: 10.1016/S0140-6736(20)30628-0.
    1. Gupta S, et al. Factors associated with death in critically ill patients with coronavirus disease 2019 in the US. JAMA Intern. Med. 2020;180:1436–1447. doi: 10.1001/jamainternmed.2020.3596.
    1. Del Valle DM, et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat. Med. 2020;26:1636–1643. doi: 10.1038/s41591-020-1051-9.
    1. Short SAP, et al. d-dimer and death in critically ill patients with coronavirus disease 2019. Crit. Care Med. 2021;49:e500–e511. doi: 10.1097/CCM.0000000000004917.
    1. Wilson JG, et al. Cytokine profile in plasma of severe COVID-19 does not differ from ARDS and sepsis. JCI Insight. 2020;5:140289. doi: 10.1172/jci.insight.140289.
    1. RECOVERY Collaborative Group Dexamethasone in hospitalized patients with Covid-19. N. Engl. J. Med. 2021;384:693–704. doi: 10.1056/NEJMoa2021436.
    1. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-analysis. JAMA. 2020;324:1330–1341. doi: 10.1001/jama.2020.17023.
    1. Gupta S, et al. Association between early treatment with tocilizumab and mortality among critically ill patients with COVID-19. JAMA Intern. Med. 2021;181:41–51. doi: 10.1001/jamainternmed.2020.6252.
    1. Guaraldi G, et al. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. Lancet Rheumatol. 2020;2:e474–e484. doi: 10.1016/S2665-9913(20)30173-9.
    1. Stone JH, et al. Efficacy of tocilizumab in patients hospitalized with Covid-19. N. Engl. J. Med. 2020;383:2333–2344. doi: 10.1056/NEJMoa2028836.
    1. Salvarani C, et al. Effect of tocilizumab vs standard care on clinical worsening in patients hospitalized with COVID-19 pneumonia: a randomized clinical trial. JAMA Intern. Med. 2021;181:24–31. doi: 10.1001/jamainternmed.2020.6615.
    1. Leaf DE, Gupta S, Wang W. Tocilizumab in covid-19. N. Engl. J. Med. 2021;384:86–87. doi: 10.1056/NEJMc2032911.
    1. REMAP-CAP Investigators Interleukin-6 receptor antagonists in critically ill patients with covid-19. N. Engl. J. Med. 2021;384:1491–1502. doi: 10.1056/NEJMoa2100433.
    1. RECOVERY Collaborative Group. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397:1637–1645. doi: 10.1016/S0140-6736(21)00676-0.
    1. Normand SLT. The RECOVERY platform. N. Engl. J. Med. 2021;384:757–758. doi: 10.1056/NEJMe2025674.
    1. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group Association between administration of IL-6 antagonists and mortality among patients hospitalized for COVID-19: a meta-analysis. JAMA. 2021;326:499–518. doi: 10.1001/jama.2021.11330.
    1. Kalil AC, et al. Baricitinib plus remdesivir for hospitalized adults with Covid-19. N. Engl. J. Med. 2021;384:795–807. doi: 10.1056/NEJMoa2031994.
    1. Marconi VC, 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;9:1407–1418. doi: 10.1016/S2213-2600(21)00331-3.
    1. Ely EW, et al. Efficacy and safety of baricitinib plus standard of care for the treatment of critically ill hospitalised adults with COVID-19 on invasive mechanical ventilation or extracorporeal membrane oxygenation: an exploratory, randomised, placebo-controlled trial. Lancet Respir. Med. 2022;10:327–336. doi: 10.1016/S2213-2600(22)00006-6.
    1. RECOVERY Collaborative Group. Baricitinib in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial and updated meta-analysis. Lancet. 2022;400:359–368. doi: 10.1016/S0140-6736(22)01109-6.
    1. Axfors C, et al. Mortality outcomes with hydroxychloroquine and chloroquine in COVID-19 from an international collaborative meta-analysis of randomized trials. Nat. Commun. 2021;12:2349. doi: 10.1038/s41467-021-22446-z.
    1. Arabi YM, et al. Lopinavir-ritonavir and hydroxychloroquine for critically ill patients with COVID-19: REMAP-CAP randomized controlled trial. Intensive Care Med. 2021;47:867–886. doi: 10.1007/s00134-021-06448-5.
    1. Reis G, et al. Effect of early treatment with ivermectin among patients with Covid-19. N. Engl. J. Med. 2022;386:1721–1731. doi: 10.1056/NEJMoa2115869.
    1. Sheahan TP, et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci. Transl Med. 2017;9:eaal3653. doi: 10.1126/scitranslmed.aal3653.
    1. Wang M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271. doi: 10.1038/s41422-020-0282-0.
    1. Gottlieb RL, et al. Early remdesivir to prevent progression to severe Covid-19 in outpatients. N. Engl. J. Med. 2022;386:305–315. doi: 10.1056/NEJMoa2116846.
    1. Beigel JH, et al. Remdesivir for the treatment of Covid-19 — final report. N. Engl. J. Med. 2020;383:1813–1826. doi: 10.1056/NEJMoa2007764.
    1. WHO Solidarity Trial Consortium. Remdesivir and three other drugs for hospitalised patients with COVID-19: final results of the WHO Solidarity randomised trial and updated meta-analyses. Lancet. 2022;399:1941–1953. doi: 10.1016/S0140-6736(22)00519-0.
    1. Owen DR, et al. An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science. 2021;374:1586–1593. doi: 10.1126/science.abl4784.
    1. Hammond J, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with Covid-19. N. Engl. J. Med. 2022;386:1397–1408. doi: 10.1056/NEJMoa2118542.
    1. Sheahan TP, et al. An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci. Transl Med. 2020;12:eabb5883. doi: 10.1126/scitranslmed.abb5883.
    1. Jayk Bernal A, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N. Engl. J. Med. 2022;386:509–520. doi: 10.1056/NEJMoa2116044.
    1. Hoover RK, et al. Clinical pharmacokinetics of sulfobutylether-β-cyclodextrin in patients with varying degrees of renal impairment. J. Clin. Pharmacol. 2018;58:814–822. doi: 10.1002/jcph.1077.
    1. Estiverne C, et al. Remdesivir in patients with estimated GFR <30 ml/min per 1.73 m2 or on renal replacement therapy. Kidney Int. Rep. 2021;6:835–838. doi: 10.1016/j.ekir.2020.11.025.
    1. Thakare S, et al. Safety of remdesivir in patients with acute kidney injury or CKD. Kidney Int. Rep. 2021;6:206–210. doi: 10.1016/j.ekir.2020.10.005.
    1. Pettit NN, et al. Remdesivir use in the setting of severe renal impairment: a theoretical concern or real risk? Clin. Infect. Dis. 2021;73:e3990–e3995. doi: 10.1093/cid/ciaa1851.
    1. Sukeishi A, et al. Population pharmacokinetic modeling of GS-441524, the active metabolite of remdesivir, in Japanese COVID-19 patients with renal dysfunction. CPT Pharmacomet. Syst. Pharmacol. 2022;11:94–103. doi: 10.1002/psp4.12736.
    1. Seethapathy R, Zhao S, Long JD, Strohbehn IA, Sise ME. A propensity score-matched observational study of remdesivir in patients with COVID-19 and severe kidney disease. Kidney360. 2022;3:269–278. doi: 10.34067/KID.0006152021.
    1. US National Library of Medicine. (2022).
    1. US Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization for paxlovid. FDA (2022).
    1. Salerno DM, et al. Early clinical experience with nirmatrelvir/ritonavir for the treatment of COVID-19 in solid organ transplant recipients. Am. J. Transplant. 2022;22:2083–2088. doi: 10.1111/ajt.17027.
    1. Swanstrom R, Schinazi RF. Lethal mutagenesis as an antiviral strategy. Science. 2022;375:497–498. doi: 10.1126/science.abn0048.
    1. Al-Samkari H, et al. Thrombosis, bleeding, and the observational effect of early therapeutic anticoagulation on survival in critically ill patients with COVID-19. Ann. Intern. Med. 2021;174:622–632. doi: 10.7326/M20-6739.
    1. Al-Samkari H, et al. COVID-19 and coagulation: bleeding and thrombotic manifestations of SARS-CoV-2 infection. Blood. 2020;136:489–500. doi: 10.1182/blood.2020006520.
    1. Iba T, Connors JM, Levy JH. The coagulopathy, endotheliopathy, and vasculitis of COVID-19. Inflamm. Res. 2020;69:1181–1189. doi: 10.1007/s00011-020-01401-6.
    1. Goshua G, et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol. 2020;7:e575–e582. doi: 10.1016/S2352-3026(20)30216-7.
    1. Al-Samkari H, Song F, Van Cott EM, Kuter DJ, Rosovsky R. Evaluation of the prothrombin fragment 1.2 in patients with coronavirus disease 2019 (COVID-19) Am. J. Hematol. 2020;95:1479–1485. doi: 10.1002/ajh.25962.
    1. ATTACC Investigators, ACTIV-4a Investigators & REMAP-CAP Investigators Therapeutic anticoagulation with heparin in noncritically ill patients with Covid-19. N. Engl. J. Med. 2021;385:790–802. doi: 10.1056/NEJMoa2105911.
    1. REMAP-CAP Investigators, ACTIV-4a Investigators & ATTACC Investigators Therapeutic anticoagulation with heparin in critically ill patients with Covid-19. N. Engl. J. Med. 2021;385:777–789. doi: 10.1056/NEJMoa2103417.
    1. Al-Samkari H. Finding the optimal thromboprophylaxis dose in patients with COVID-19. JAMA. 2021;325:1613–1615. doi: 10.1001/jama.2021.4295.
    1. Cuker A, et al. American Society of Hematology 2021 guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19. Blood Adv. 2021;5:872–888. doi: 10.1182/bloodadvances.2020003763.
    1. National Institutes of Health. COVID-19 treatment guidelines: antithrombotic therapy in patients with COVID-19. NIH (2022).
    1. American Society of Hematology. ASH clinical practice guidelines on venous thromboembolism. ASH (2022).
    1. Spyropoulos AC, et al. Efficacy and safety of therapeutic-dose heparin vs standard prophylactic or intermediate-dose heparins for thromboprophylaxis in high-risk hospitalized patients with COVID-19: the HEP-COVID randomized clinical trial. JAMA Intern. Med. 2021;181:1612–1620. doi: 10.1001/jamainternmed.2021.6203.
    1. Sholzberg M, et al. Effectiveness of therapeutic heparin versus prophylactic heparin on death, mechanical ventilation, or intensive care unit admission in moderately ill patients with covid-19 admitted to hospital: RAPID randomised clinical trial. BMJ. 2021;375:n2400. doi: 10.1136/bmj.n2400.
    1. Lopes RD, et al. Therapeutic versus prophylactic anticoagulation for patients admitted to hospital with COVID-19 and elevated D-dimer concentration (ACTION): an open-label, multicentre, randomised, controlled trial. Lancet. 2021;397:2253–2263. doi: 10.1016/S0140-6736(21)01203-4.
    1. Giannis D, et al. Postdischarge thromboembolic outcomes and mortality of hospitalized patients with COVID-19: the CORE-19 registry. Blood. 2021;137:2838–2847. doi: 10.1182/blood.2020010529.
    1. Ramacciotti E, et al. Rivaroxaban versus no anticoagulation for post-discharge thromboprophylaxis after hospitalisation for COVID-19 (MICHELLE): an open-label, multicentre, randomised, controlled trial. Lancet. 2022;399:50–59. doi: 10.1016/S0140-6736(21)02392-8.
    1. Spyropoulos AC, et al. Post-discharge prophylaxis with rivaroxaban reduces fatal and major thromboembolic events in medically ill patients. J. Am. Coll. Cardiol. 2020;75:3140–3147. doi: 10.1016/j.jacc.2020.04.071.
    1. RECOVERY Collaborative Group. Aspirin in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2022;399:143–151. doi: 10.1016/S0140-6736(21)01825-0.
    1. Connors JM, et al. Effect of antithrombotic therapy on clinical outcomes in outpatients with clinically stable symptomatic COVID-19: the ACTIV-4B randomized clinical trial. JAMA. 2021;326:1703–1712. doi: 10.1001/jama.2021.17272.
    1. REMAP-CAP Writing Committee for the REMAP-CAP Investigators Effect of antiplatelet therapy on survival and organ support-free days in critically ill patients with COVID-19: a randomized clinical trial. JAMA. 2022;327:1247–1259. doi: 10.1001/jama.2022.2910.
    1. US National Library of Medicine. (2022).
    1. Kaw D, Malhotra D. Platelet dysfunction and end-stage renal disease. Semin. Dial. 2006;19:317–322. doi: 10.1111/j.1525-139X.2006.00179.x.
    1. Wattanakit K, Cushman M, Stehman-Breen C, Heckbert SR, Folsom AR. Chronic kidney disease increases risk for venous thromboembolism. J. Am. Soc. Nephrol. 2008;19:135–140. doi: 10.1681/ASN.2007030308.
    1. Martel N, Lee J, Wells PS. Risk for heparin-induced thrombocytopenia with unfractionated and low-molecular-weight heparin thromboprophylaxis: a meta-analysis. Blood. 2005;106:2710–2715. doi: 10.1182/blood-2005-04-1546.
    1. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141:e24S–e43S. doi: 10.1378/chest.11-2291.
    1. Endres P, et al. Filter clotting with continuous renal replacement therapy in COVID-19. J. Thromb. Thrombolysis. 2021;51:966–970. doi: 10.1007/s11239-020-02301-6.
    1. Arnold F, et al. Comparison of different anticoagulation strategies for renal replacement therapy in critically ill patients with COVID-19: a cohort study. BMC Nephrol. 2020;21:486. doi: 10.1186/s12882-020-02150-8.
    1. Seshadri M, Ahamed J, Laurence J. Intervention in COVID-19 linked hypercoagulable states characterized by circuit thrombosis utilizing a direct thrombin inhibitor. Thrombosis Update. 2020;1:100009. doi: 10.1016/j.tru.2020.100009.
    1. Rahbar E, Cotton BA, Wade CE, Cardenas JC. Acquired antithrombin deficiency is a risk factor for venous thromboembolism after major trauma. Thromb. Res. 2021;204:9–12. doi: 10.1016/j.thromres.2021.05.015.
    1. Legrand M, et al. Pathophysiology of COVID-19-associated acute kidney injury. Nat. Rev. Nephrol. 2021;17:751–764. doi: 10.1038/s41581-021-00452-0.
    1. Rapkiewicz AV, et al. Megakaryocytes and platelet-fibrin thrombi characterize multi-organ thrombosis at autopsy in COVID-19: a case series. EClinicalMedicine. 2020;24:100434. doi: 10.1016/j.eclinm.2020.100434.
    1. Hisama N, et al. Anticoagulant pretreatment attenuates production of cytokine-induced neutrophil chemoattractant following ischemia-reperfusion of rat liver. Dig. Dis. Sci. 1996;41:1481–1486. doi: 10.1007/BF02088576.
    1. Marini JJ, Gattinoni L. Management of COVID-19 respiratory distress. JAMA. 2020;323:2329–2330. doi: 10.1001/jama.2020.6825.
    1. Papoutsi E, Giannakoulis VG, Xourgia E, Routsi C, Kotanidou A. Siempos II. Effect of timing of intubation on clinical outcomes of critically ill patients with COVID-19: a systematic review and meta-analysis of non-randomized cohort studies. Crit. Care. 2021;25:121. doi: 10.1186/s13054-021-03540-6.
    1. Ziehr DR, et al. Respiratory pathophysiology of mechanically ventilated patients with COVID-19: a cohort study. Am. J. Respir. Crit. Care Med. 2020;201:1560–1564. doi: 10.1164/rccm.202004-1163LE.
    1. Anesi GL, et al. Characteristics, outcomes, and trends of patients with COVID-19-related critical illness at a learning health system in the United States. Ann. Intern. Med. 2021;174:613–621. doi: 10.7326/M20-5327.
    1. Gaeckle NT, et al. Aerosol generation from the respiratory tract with various modes of oxygen delivery. Am. J. Respir. Crit. Care Med. 2020;202:1115–1124. doi: 10.1164/rccm.202006-2309OC.
    1. Perkins GD, et al. Effect of noninvasive respiratory strategies on intubation or mortality among patients with acute hypoxemic respiratory failure and COVID-19: the RECOVERY-RS randomized clinical trial. JAMA. 2022;327:546–558. doi: 10.1001/jama.2022.0028.
    1. Guérin C, et al. Prone positioning in severe acute respiratory distress syndrome. N. Engl. J. Med. 2013;368:2159–2168. doi: 10.1056/NEJMoa1214103.
    1. Mathews KS, et al. Prone positioning and survival in mechanically ventilated patients with coronavirus disease 2019-related respiratory failure. Crit. Care Med. 2021;49:1026–1037.
    1. Ehrmann S, et al. Awake prone positioning for COVID-19 acute hypoxaemic respiratory failure: a randomised, controlled, multinational, open-label meta-trial. Lancet Respir. Med. 2021;9:1387–1395. doi: 10.1016/S2213-2600(21)00356-8.
    1. Alhazzani AW, et al. Effect of awake prone positioning on endotracheal intubation in patients with COVID-19 and acute respiratory failure: a randomized clinical trial. JAMA. 2022;327:2104–2113. doi: 10.1001/jama.2022.7993.
    1. Qian ET, et al. Assessment of awake prone positioning in hospitalized adults with COVID-19: a nonrandomized controlled trial. JAMA Intern. Med. 2022;182:612–621. doi: 10.1001/jamainternmed.2022.1070.
    1. National Heart, Lung, and Blood Institute PETAL Clinical Trials Network. Moss M, et al. Early neuromuscular blockade in the acute respiratory distress syndrome. N. Engl. J. Med. 2019;380:1997–2008. doi: 10.1056/NEJMoa1901686.
    1. Shaefi S, et al. Extracorporeal membrane oxygenation in patients with severe respiratory failure from COVID-19. Intensive Care Med. 2021;47:208–221. doi: 10.1007/s00134-020-06331-9.
    1. Urner M, et al. Venovenous extracorporeal membrane oxygenation in patients with acute covid-19 associated respiratory failure: comparative effectiveness study. BMJ. 2022;377:e068723. doi: 10.1136/bmj-2021-068723.
    1. Goligher EC, et al. Extracorporeal membrane oxygenation for severe acute respiratory distress syndrome and posterior probability of mortality benefit in a post hoc Bayesian analysis of a randomized clinical trial. JAMA. 2018;320:2251–2259. doi: 10.1001/jama.2018.14276.
    1. Gannon WD, et al. Association between availability of ECMO and mortality in COVID-19 patients eligible for ECMO: a natural experiment. Am. J. Respir. Crit. Care Med. 2022;205:1354–1357. doi: 10.1164/rccm.202110-2399LE.
    1. Darmon M, et al. Acute respiratory distress syndrome and risk of AKI among critically ill patients. Clin. J. Am. Soc. Nephrol. 2014;9:1347–1353. doi: 10.2215/CJN.08300813.
    1. Teixeira JP, Ambruso S, Griffin BR, Faubel S. Pulmonary consequences of acute kidney injury. Semin. Nephrol. 2019;39:3–16. doi: 10.1016/j.semnephrol.2018.10.001.
    1. Pantaleo G, Correia B, Fenwick C, Joo VS, Perez L. Antibodies to combat viral infections: development strategies and progress. Nat. Rev. Drug Discov. 2022;21:676–696. doi: 10.1038/s41573-022-00495-3.
    1. Janiaud P, et al. Association of convalescent plasma treatment with clinical outcomes in patients with COVID-19: a systematic review and meta-analysis. JAMA. 2021;325:1185–1195. doi: 10.1001/jama.2021.2747.
    1. Bégin P, et al. Convalescent plasma for hospitalized patients with COVID-19: an open-label, randomized controlled trial. Nat. Med. 2021;27:2012–2024. doi: 10.1038/s41591-021-01488-2.
    1. RECOVERY Collaborative Group. Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised controlled, open-label, platform trial. Lancet. 2021;397:2049–2059. doi: 10.1016/S0140-6736(21)00897-7.
    1. Writing Committee for the REMAP-CAP Investigators Effect of convalescent plasma on organ support-free days in critically ill patients with COVID-19: a randomized clinical trial. JAMA. 2021;326:1690–1702. doi: 10.1001/jama.2021.18178.
    1. Sullivan DJ, et al. Early outpatient treatment for Covid-19 with convalescent plasma. N. Engl. J. Med. 2022;386:1700–1711. doi: 10.1056/NEJMoa2119657.
    1. Libster R, et al. Early high-titer plasma therapy to prevent severe Covid-19 in older adults. N. Engl. J. Med. 2021;384:610–618. doi: 10.1056/NEJMoa2033700.
    1. Alemany A, et al. High-titre methylene blue-treated convalescent plasma as an early treatment for outpatients with COVID-19: a randomised, placebo-controlled trial. Lancet Respir. Med. 2022;10:278–288. doi: 10.1016/S2213-2600(21)00545-2.
    1. Korley FK, et al. Early convalescent plasma for high-risk outpatients with Covid-19. N. Engl. J. Med. 2021;385:1951–1960. doi: 10.1056/NEJMoa2103784.
    1. Millat-Martinez P, et al. Prospective individual patient data meta-analysis of two randomized trials on convalescent plasma for COVID-19 outpatients. Nat. Commun. 2022;13:2583. doi: 10.1038/s41467-022-29911-3.
    1. National Institutes of Health. Anti-SARS-CoV-2 antibody products summary recommendations: COVID-19 treatment guidelines. NIH (2022).
    1. Taylor PC, et al. Neutralizing monoclonal antibodies for treatment of COVID-19. Nat. Rev. Immunol. 2021;21:382–393. doi: 10.1038/s41577-021-00542-x.
    1. RECOVERY Collaborative Group. Casirivimab and imdevimab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2022;399:665–676. doi: 10.1016/S0140-6736(22)00163-5.
    1. Dougan M, et al. A randomized, placebo-controlled clinical trial of bamlanivimab and etesevimab together in high-risk ambulatory patients with COVID-19 and validation of the prognostic value of persistently high viral load. Clin. Infect. Dis. 2021;75:e440–e449. doi: 10.1093/cid/ciab912.
    1. Gupta A, et al. Effect of sotrovimab on hospitalization or death among high-risk patients with mild to moderate COVID-19: a randomized clinical trial. JAMA. 2022;327:1236–1246. doi: 10.1001/jama.2022.2832.
    1. Weinreich DM, et al. REGEN-COV antibody combination and outcomes in outpatients with Covid-19. N. Engl. J. Med. 2021;385:e81. doi: 10.1056/NEJMoa2108163.
    1. Takashita E, et al. Efficacy of antibodies and antiviral drugs against Covid-19 Omicron variant. N. Engl. J. Med. 2022;386:995–998. doi: 10.1056/NEJMc2119407.
    1. Levin MJ, et al. Intramuscular AZD7442 (tixagevimab-cilgavimab) for prevention of Covid-19. N. Engl. J. Med. 2022;386:2188–2200. doi: 10.1056/NEJMoa2116620.
    1. Molnar MZ, et al. Outcomes of critically ill solid organ transplant patients with COVID-19 in the United States. Am. J. Transpl. 2020;20:3061–3071. doi: 10.1111/ajt.16280.
    1. Cristelli MP, et al. Efficacy of convalescent plasma to treat mild to moderate COVID-19 in kidney transplant patients: a propensity score matching analysis. Transplantation. 2022;106:e92–e94. doi: 10.1097/TP.0000000000003962.
    1. Kluger MA, et al. Convalescent plasma treatment for early post-kidney transplant acquired COVID-19. Transpl. Infect. Dis. 2021;23:e13685. doi: 10.1111/tid.13685.
    1. Rodionov RN, et al. Potential benefit of convalescent plasma transfusions in immunocompromised patients with COVID-19. Lancet Microbe. 2021;2:e138. doi: 10.1016/S2666-5247(21)00030-6.
    1. Thompson MA, et al. Association of convalescent plasma therapy with survival in patients with hematologic cancers and COVID-19. JAMA Oncol. 2021;7:1167–1175. doi: 10.1001/jamaoncol.2021.1799.
    1. Arikawa S, et al. Effectiveness of neutralizing antibody cocktail in hemodialysis patients: a case series of 20 patients treated with or without REGN-COV2. Clin. Exp. Nephrol. 2022;26:476–485. doi: 10.1007/s10157-021-02151-3.
    1. Gueguen J, et al. Early administration of anti-SARS-CoV-2 monoclonal antibodies prevents severe Covid-19 in kidney transplant patients. Kidney Int. Rep. 2022;7:1241–1247. doi: 10.1016/j.ekir.2022.03.020.
    1. Bertrand D, et al. Efficacy of anti-SARS-CoV-2 monoclonal antibody prophylaxis and vaccination on the Omicron variant of COVID-19 in kidney transplant recipients. Kidney Int. 2022;102:440–442. doi: 10.1016/j.kint.2022.05.007.
    1. Al Jurdi A, et al. Tixagevimab/cilgavimab pre-exposure prophylaxis is associated with lower breakthrough infection risk in vaccinated solid organ transplant recipients during the omicron wave. Am. J. Transplant. 2022 doi: 10.1111/ajt.17128.
    1. US Food and Drug Administration. Fact sheet for health care providers emergency use authorization (EUA) of bamlanivimab and etesevimab. FDA (2020).
    1. US Food and Drug Administration. Fact sheet for health care providers emergency use authorization (EUA) of REGEN-COV. FDA (2020).
    1. US Food and Drug Administration. Fact sheet for healthcare providers emergency use authorization (EUA) of sotrovimab. FDA (2020).
    1. Vaduganathan M, et al. Renin-angiotensin-aldosterone system inhibitors in patients with Covid-19. N. Engl. J. Med. 2020;382:1653–1659. doi: 10.1056/NEJMsr2005760.
    1. Sfera A, et al. Intoxication with endogenous angiotensin II: a COVID-19 hypothesis. Front. Immunol. 2020;11:1472. doi: 10.3389/fimmu.2020.01472.
    1. Hoffmann M, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280.e8. doi: 10.1016/j.cell.2020.02.052.
    1. Hamming I, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004;203:631–637. doi: 10.1002/path.1570.
    1. Jiang F, et al. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat. Rev. Cardiol. 2014;11:413–426. doi: 10.1038/nrcardio.2014.59.
    1. Imai Y, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–116. doi: 10.1038/nature03712.
    1. Zhang H, Penninger JM, Li Y, Zhong N, Slutsky AS. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46:586–590. doi: 10.1007/s00134-020-05985-9.
    1. Rysz S, et al. COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin-aldosterone system. Nat. Commun. 2021;12:2417. doi: 10.1038/s41467-021-22713-z.
    1. Lawler PR, Derde LPG, Mc Verry B, Russell JA, van de Veerdonk F. The renin-angiotensin system in acute lung injury. Crit. Care Med. 2022;50:1411–1415. doi: 10.1097/CCM.0000000000005567.
    1. Caldeira D, Alarcão J, Vaz-Carneiro A, Costa J. Risk of pneumonia associated with use of angiotensin converting enzyme inhibitors and angiotensin receptor blockers: systematic review and meta-analysis. BMJ. 2012;345:e4260. doi: 10.1136/bmj.e4260.
    1. Chung SC, Providencia R, Sofat R. Association between angiotensin blockade and incidence of influenza in the United Kingdom. N. Engl. J. Med. 2020;383:397–400. doi: 10.1056/NEJMc2005396.
    1. Kim J, et al. Effect of antihypertensive medications on sepsis-related outcomes: a population-based cohort study. Crit. Care Med. 2019;47:e386–e393. doi: 10.1097/CCM.0000000000003654.
    1. Baral R, et al. Association between renin-angiotensin-aldosterone system inhibitors and clinical outcomes in patients with COVID-19: a systematic review and meta-analysis. JAMA Netw. Open. 2021;4:e213594. doi: 10.1001/jamanetworkopen.2021.3594.
    1. Lopes RD, et al. Effect of discontinuing vs continuing angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers on days alive and out of the hospital in patients admitted with COVID-19: a randomized clinical trial. JAMA. 2021;325:254–264. doi: 10.1001/jama.2020.25864.
    1. Duarte M, et al. Telmisartan for treatment of Covid-19 patients: an open multicenter randomized clinical trial. EClinicalMedicine. 2021;37:100962. doi: 10.1016/j.eclinm.2021.100962.
    1. Puskarich MA, et al. A multi-center phase II randomized clinical trial of losartan on symptomatic outpatients with COVID-19. EClinicalMedicine. 2021;37:100957. doi: 10.1016/j.eclinm.2021.100957.
    1. Ramagopalan SV, et al. A ChIP-seq defined genome-wide map of vitamin D receptor binding: associations with disease and evolution. Genome Res. 2010;20:1352–1360. doi: 10.1101/gr.107920.110.
    1. Liu PT, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311:1770–1773. doi: 10.1126/science.1123933.
    1. National Heart, Lung, and Blood Institute PETAL Clinical Trials Network. Ginde AA, et al. Early high-dose vitamin D3 for critically ill, vitamin D-deficient patients. N. Engl. J. Med. 2019;381:2529–2540. doi: 10.1056/NEJMoa1911124.
    1. Amrein K, et al. Effect of high-dose vitamin D3 on hospital length of stay in critically ill patients with vitamin D deficiency: the VITdAL-ICU randomized clinical trial. JAMA. 2014;312:1520–1530. doi: 10.1001/jama.2014.13204.
    1. Murai IH, et al. Effect of a single high dose of vitamin D3 on hospital length of stay in patients with moderate to severe COVID-19: a randomized clinical trial. JAMA. 2021;325:1053–1060. doi: 10.1001/jama.2020.26848.
    1. Leaf DE, Ginde AA. Vitamin D3 to treat COVID-19: different disease, same answer. JAMA. 2021;325:1047–1048. doi: 10.1001/jama.2020.26850.
    1. US National Library of Medicine. (2022).
    1. Tran MT, et al. PGC1α drives NAD biosynthesis linking oxidative metabolism to renal protection. Nature. 2016;531:528–532. doi: 10.1038/nature17184.
    1. Poyan Mehr A, et al. De novo NAD+ biosynthetic impairment in acute kidney injury in humans. Nat. Med. 2018;24:1351–1359. doi: 10.1038/s41591-018-0138-z.
    1. US National Library of Medicine. (2022).
    1. US National Library of Medicine. (2021).
    1. Raines NH, et al. Niacinamide may be associated with improved outcomes in COVID-19-related acute kidney injury: an observational study. Kidney360. 2021;2:33–41. doi: 10.34067/KID.0006452020.
    1. US National Library of Medicine. (2021).
    1. US National Library of Medicine. (2022).
    1. Sato R, et al. Effect of IV high-dose vitamin c on mortality in patients with sepsis: a systematic review and meta-analysis of randomized controlled trials. Crit. Care Med. 2021;49:2121–2130.
    1. Lamontagne F, et al. Intravenous vitamin C in adults with sepsis in the intensive care unit. N. Engl. J. Med. 2022;386:2387–2398. doi: 10.1056/NEJMoa2200644.
    1. Thomas S, et al. Effect of high-dose zinc and ascorbic acid supplementation vs usual care on symptom length and reduction among ambulatory patients with SARS-CoV-2 infection: the COVID A to Z randomized clinical trial. JAMA Netw. Open. 2021;4:e210369. doi: 10.1001/jamanetworkopen.2021.0369.
    1. Zhang J, et al. Pilot trial of high-dose vitamin C in critically ill COVID-19 patients. Ann. Intensive Care. 2021;11:5. doi: 10.1186/s13613-020-00792-3.
    1. Johns Hopkins University & Medicine. Coronavirus Resource Center. JHU (2022).
    1. INSPIRATION Investigators Effect of intermediate-dose vs standard-dose prophylactic anticoagulation on thrombotic events, extracorporeal membrane oxygenation treatment, or mortality among patients with COVID-19 admitted to the intensive care unit: the INSPIRATION randomized clinical trial. JAMA. 2021;325:1620–1630. doi: 10.1001/jama.2021.4152.
    1. ACTIV-3/TICO LY-CoV555 Study Group A neutralizing monoclonal antibody for hospitalized patients with Covid-19. N. Engl. J. Med. 2021;384:905–914. doi: 10.1056/NEJMoa2033130.
    1. Dougan M, et al. Bamlanivimab plus etesevimab in mild or moderate Covid-19. N. Engl. J. Med. 2021;385:1382–1392. doi: 10.1056/NEJMoa2102685.

Source: PubMed

3
Abonnieren