1
|
Gambari R, Papi C, Gasparello J, Agostinelli E, Finotti A. Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 2025; 29:85. [PMID: 40084194 PMCID: PMC11904878 DOI: 10.3892/etm.2025.12835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences and to develop novel therapeutic approaches. Interleukin-6 (IL-6) and IL-8 are considered significant therapeutic targets for COVID-19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19-25 nucleotides in length, non-coding RNAs that regulate gene expression at the post-transcriptional level through the sequence-selective recognition of the 3'-untranslated region (3'-UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID-19 'cytokine storm', miR-93-5p can inhibit IL-8 gene expression by directly targeting the 3'-UTR of IL-8 mRNA. In addition, miR-93-5p can regulate Toll-like receptor-4 (TLR4) and interleukin-1 receptor-associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor-κB (NF-κB) pathway and the expression of NF-κB-regulated genes, such as IL-6, IL-1β and other hyper-expressed genes during the COVID-19 'cytokine storm'. In the present study, the results provided preliminary evidence suggesting that the miR-93-5p-based miRNA therapeutics could be combined with the anti-inflammatory aged garlic extract (AGE) to more effectively inhibit IL-8 gene expression. The human bronchial epithelial IB3-1 cell line was employed as experimental model system. IB3-1 cells were stimulated with the BNT162b2 COVID-19 vaccine and transfected with pre-miR-93-5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2-induced expression of the IL-8 gene. The accumulation of IL-8 mRNA was assessed by RT-qPCR; the release of IL-8 protein was determined by Bio-Plex assay. In addition, the possible applications of TLR4/NF-κB inhibitory agents (such as miR-93-5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID-19, cystic fibrosis and other respiratory diseases, were summarized.
Collapse
Affiliation(s)
- Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
2
|
Dargahi-Malamir M, Hosseinzadeh M, Khedri R, Hosseininejad H, Behmanesh B, Hatami A, Oskouei SY, Mehraban Z, Kouti W, Pouladzadeh M. A Pilot and Feasibility Study to Evaluate the Effectiveness of Tofacitinib Add-On Therapy to Remdesivir in Severely Ill COVID-19 Patients. MAEDICA 2024; 19:322-329. [PMID: 39188838 PMCID: PMC11345074 DOI: 10.26574/maedica.2024.19.2.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
AIM This placebo-controlled and randomized pilot study aimed to assess the feasibility and impact of tofacitinib/remdesivir combination therapy compared to remdesivir alone on clinical and laboratory findings of severely ill COVID-19 patients for future large-scale studies. METHOD Fifty patients were included in this study. They were randomly allocated into two groups of 25 subjects each. Patients in the treatment group received a five-day course of tofacitinib (10 mg twice daily) in addition to a five-day course of remdesivir, whereas the control group received a 10-day course of remdesivir with a placebo. RESULTS There was no significant difference in rates of need for intubation (oxygen saturation level), intensive care unit (ICU) admission, death and length of hospitalization between the two groups (P>0.05). Nevertheless, the PRIEST severity score was significantly reduced in the treatment group compared to the control group (P=0.03, effect size [95% CI]: -0.616 [0.0233-1.1723]). Moreover, the mean level of C-reactive protein after 10 days was significantly decreased in the treatment group but increased in the control group (P=0.006). CONCLUSION Tofacitinib adopted in this pilot study modulate the inflammation and reduce the PRIEST score in severe COVID-19 patients. So, it is feasible and could be applied in future larger-scale trials to precisely determine its effects on coronavirus infections.
Collapse
Affiliation(s)
- Mehrdad Dargahi-Malamir
- Department of Pulmonology, Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mofid Hosseinzadeh
- Emergency Medicine Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Khedri
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hooshmand Hosseininejad
- Department of Pulmonology, Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Babak Behmanesh
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Hatami
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Yashar Oskouei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Mehraban
- Department of Pulmonology, Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Wesam Kouti
- Lung Diseases & Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mandana Pouladzadeh
- Emergency Medicine Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
3
|
Becker AP, Mang S, Rixecker T, Lepper PM. [COVID-19 in the intensive care unit]. Pneumologie 2024; 78:330-345. [PMID: 38759701 DOI: 10.1055/a-1854-2693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
The acute respiratory failure as well as ARDS (acute respiratory distress syndrome) have challenged clinicians since the initial description over 50 years ago. Various causes can lead to ARDS and therapeutic approaches for ARDS/ARF are limited to the support or replacement of organ functions and the prevention of therapy-induced consequences. In recent years, triggered by the SARS-CoV-2 pathogen, numerous cases of acute lung failure (C-ARDS) have emerged. The pathophysiological processes of classical ARDS and C-ARDS are essentially similar. In their final stages of inflammation, both lead to a disruption of the blood-air barrier. Treatment strategies for C-ARDS, like classical ARDS, focus on supporting or replacing organ functions and preventing consequential damage. This article summarizes the treatment strategies in the intensive care unit.
Collapse
|
4
|
Amante EJB, David-Wang AS, Tee ML, Punzalan FER, Añonuevo JC, Fernandez LC, Albay AB, Malabad JCM, Climacosa FMM, Pajes ANNI, Cuaño PMGM, Alejandría MM. Intravenous Tocilizumab versus Standard of Care in the Treatment of Severe and Critical COVID-19-related Pneumonia: A Single-center, Double-blind, Placebo-controlled, Phase 3 Trial. ACTA MEDICA PHILIPPINA 2024; 58:7-13. [PMID: 38846161 PMCID: PMC11151130 DOI: 10.47895/amp.vi0.6175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Background Severe and critical COVID-19 disease is characterized by hyperinflammation involving pro-inflammatory cytokines, particularly IL-6. Tocilizumab is a monoclonal antibody that blocks IL-6 receptors. Objectives This study evaluated the efficacy of tocilizumab in Filipino patients with severe to critical COVID-19 disease. Methods This phase 3 randomized double-blind trial, included patients hospitalized for severe or critical COVID-19 in a 1:1 ratio to receive either tocilizumab plus local standard of care or placebo plus standard of care. Patients were eligible for a repeat IV infusion within 24-48 hours if they deteriorated or did not improve. Treatment success or clinical improvement was defined as at least two categories of improvement from baseline in the WHO 7-point Ordinal Scale of patient status, in an intention-to-treat manner. Results Forty-nine (49) patients were randomized in the tocilizumab arm and 49 in the placebo arm. There was no significant difference in age, comorbidities, COVID-19 severity, need for mechanical ventilation, presence of acute respiratory distress syndrome, or biomarker levels between groups. Use of adjunctive therapy was similar between groups, with corticosteroid used in 91.8% in tocilizumab group and 81.6% in the placebo group, while remdesivir was used in 98% of participants in both groups.There was no significant difference between groups in terms of treatment success in both the intention-to-treat analysis (relative risk=1.05, 95% CI: 0.85-1.30) and per-protocol analysis (relative risk=0.98, 95% CI: 0.80 to 1.21). There was no significant difference in time to improvement of at least two categories relative to baseline on the 7-point Ordinal Scale of clinical status. Conclusion The use of tocilizumab on top of standard of care in the management of patients with severe to critical COVID-19 did not result in significant improvement as defined by the WHO 7-point Ordinal Scale of patient status, nor in significant improvement in incidence of mechanical ventilation, incidence of ICU admission, length of ICU stay, and mortality rate.
Collapse
Affiliation(s)
- Eric Jason B. Amante
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Aileen S. David-Wang
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Michael L. Tee
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Felix Eduardo R. Punzalan
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - John C. Añonuevo
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Lenora C. Fernandez
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Albert B. Albay
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | | | | | - A. Nico Nahar I. Pajes
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Patricia Maria Gregoria M. Cuaño
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| | - Marissa M. Alejandría
- Department of Medicine, Philippine General Hospital, University of the Philippines Manila
- College of Medicine, University of the Philippines Manila
| |
Collapse
|
5
|
Krupka S, Hoffmann A, Jasaszwili M, Dietrich A, Guiu-Jurado E, Klöting N, Blüher M. Consequences of COVID-19 on Adipose Tissue Signatures. Int J Mol Sci 2024; 25:2908. [PMID: 38474155 DOI: 10.3390/ijms25052908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Since the emergence of coronavirus disease-19 (COVID-19) in 2019, it has been crucial to investigate the causes of severe cases, particularly the higher rates of hospitalization and mortality in individuals with obesity. Previous findings suggest that adipocytes may play a role in adverse COVID-19 outcomes in people with obesity. The impact of COVID-19 vaccination and infection on adipose tissue (AT) is currently unclear. We therefore analyzed 27 paired biopsies of visceral and subcutaneous AT from donors of the Leipzig Obesity BioBank that have been categorized into three groups (1: no infection/no vaccination; 2: no infection but vaccinated; 3: infected and vaccinated) based on COVID-19 antibodies to spike (indicating vaccination) and/or nucleocapsid proteins. We provide additional insights into the impact of COVID-19 on AT biology through a comprehensive histological transcriptome and serum proteome analysis. This study demonstrates that COVID-19 infection is associated with smaller average adipocyte size. The impact of infection on gene expression was significantly more pronounced in subcutaneous than in visceral AT and mainly due to immune system-related processes. Serum proteome analysis revealed the effects of the infection on circulating adiponectin, interleukin 6 (IL-6), and carbonic anhydrase 5A (CA5A), which are all related to obesity and blood glucose abnormalities.
Collapse
Affiliation(s)
- Sontje Krupka
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Mariami Jasaszwili
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Arne Dietrich
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Esther Guiu-Jurado
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| |
Collapse
|
6
|
Aiges M, Ramana KV. Significance of Vitamin Supplementation in Reducing the Severity of COVID-19. Mini Rev Med Chem 2024; 24:254-264. [PMID: 36967461 DOI: 10.2174/1389557523666230324081713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/24/2022] [Accepted: 01/11/2023] [Indexed: 03/29/2023]
Abstract
Coronavirus disease-19 (COVID-19), a serious pandemic due to the SARS-CoV-2 virus infection, caused significant lockdowns, healthcare shortages, and deaths worldwide. The infection leads to an uncontrolled systemic inflammatory response causing severe respiratory distress and multiple-organ failure. Quick development of several vaccines efficiently controlled the spread of COVID-19. However, the rise of various new subvariants of COVID-19 demonstrated some concerns over the efficacy of existing vaccines. Currently, better vaccines to control these variants are still under development as several new subvariants of COVID-19, such as omicron BA-4, BA-5, and BF-7 are still impacting the world. Few antiviral treatments have been shown to control COVID-19 symptoms. Further, control of COVID-19 symptoms has been explored with many natural and synthetic adjuvant compounds in hopes of treating the deadly and contagious disease. Vitamins have been shown to modulate the immune system, function as antioxidants, and reduce the inflammatory response. Recent studies have investigated the potential role of vitamins, specifically vitamins A, B, C, D, and E, in reducing the immune and inflammatory responses and severity of the complication. In this brief article, we discussed our current understanding of the role of vitamins in controlling COVID-19 symptoms and their potential use as adjuvant therapy.
Collapse
Affiliation(s)
- Myia Aiges
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - Kota V Ramana
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| |
Collapse
|
7
|
Abdel-Salam Elgohary M, Ali A, J Alarfaj S, Shahin H, Ibrahim Zaki A, Medhat Hasan E, Emam Mohamed M, Mahmoud Elkholy A, El-Masry TA, Samir Kamal J, Ali AbdelRahim M, Wageh Saber A, Seadawy MG, Elshishtawy MHM, El-Bouseary MM. Even one dose of tocilizumab could hinder bad prognosis of cytokines storm in COVID-19 patients. Cytokine 2024; 173:156433. [PMID: 37972479 DOI: 10.1016/j.cyto.2023.156433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/04/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Severe COVID-19 pneumonia is a principal cause of death due to cascade of hyper inflammatory condition that leading to lung damage. Therefore, an effective therapy to countercurrent the surge of uncontrolled inflammation is mandatory to propose. Anti-interlukin-6 receptor antagonist monoclonal therapy, tocilizumab (TCZ) showed potential results in COVID-19 patients. This study aimed to emphasize the factors associated with mortality in COVID-19 patients that treated with tocilizumab and may influence the level of serum IL-6. A retrospective cohort study included all patients with clinical parameters that pointed to presence of cytokines storm and treated with one or more doses of TCZ beside the regular protocol of COVID-19 pneumonia. The factors that influence the mortality in addition to the level of serum IL-6 were analyzed. A total of 377 patients were included, 69.5 % of them received only one dose of TCZ which started mainly at the third day of admission. The mortality rate was 29.44 %. Regardless the time of starting TCZ, just one dose was fair enough to prevent bad consequence; OR = 0.04, P = 0.001.However, in spite of protective action of TCZ, older age and female sex were significant risk factors for mortality, P = 0.001 and 0.01 respectively, as well heart disease. Moreover, increasing the level of neutrophil, AST and IL-6 were associated with bad prognosis. In the same line, treatment with ivermectin, chloroquine and remdesivir inversely affect the level of IL-6. Early treatments of COVID-19 pneumonia with at least one dose of tocilizumab minimized the fatality rate.
Collapse
Affiliation(s)
| | - Asmaa Ali
- Department of Pulmonary Medicine, Abbassia Chest Hospital, MOH, Cairo, Egypt; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| | - Sumaiah J Alarfaj
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University. P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hesham Shahin
- Chest Consultant, Manager of Military Chest Hospital, Cairo, Egypt.
| | | | | | | | | | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt.
| | | | | | - Ashgan Wageh Saber
- Chemistry Laboratory Department, Central Military Laboratories, Cairo, Egypt.
| | - Mohamed G Seadawy
- Biological Prevention Department, Ministry of Defense, Cairo, Egypt.
| | | | - Maisra M El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| |
Collapse
|
8
|
Aljuhani O, Al Sulaiman K, B Korayem G, Alharbi A, Altebainawi AF, Aldkheel SA, Alotaibi SG, Vishwakarma R, Alshareef H, Alsohimi S, AlFaifi M, Al Shaya A, Alhaidal HA, Alsubaie RM, Alrashidi H, Albarqi KJ, Alangari DT, Alanazi RM, Altaher NM, Al-Dorzi HM. The use of Tocilizumab in COVID-19 critically ill patients with renal impairment: a multicenter, cohort study. Ren Fail 2023; 45:2268213. [PMID: 37870869 PMCID: PMC11001317 DOI: 10.1080/0886022x.2023.2268213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023] Open
Abstract
Tocilizumab (TCZ) is recommended in patients with COVID-19 who require oxygen therapy or ventilatory support. Despite the wide use of TCZ, little is known about its safety and effectiveness in patients with COVID-19 and renal impairment. Therefore, this study evaluated the safety and effectiveness of TCZ in critically ill patients with COVID-19 and renal impairment. A multicenter retrospective cohort study included all adult COVID-19 patients with renal impairment (eGFR˂60 mL/min) admitted to the ICUs between March 2020 and July 2021. Patients were categorized into two groups based on TCZ use (Control vs. TCZ). The primary endpoint was the development of acute kidney injury (AKI) during ICU stay. We screened 1599 patients for eligibility; 394 patients were eligible, and 225 patients were included after PS matching (1:2 ratio); there were 75 TCZ-treated subjects and 150 controls. The rate of AKI was higher in the TCZ group compared with the control group (72.2% versus 57.4%; p = 0.03; OR: 1.83; 95% CI: 1.01, 3.34; p = 0.04). Additionally, the ICU length of stay was significantly longer in patients who received TCZ (17.5 days versus 12.5 days; p = 0.006, Beta coefficient: 0.30 days, 95% CI: 0.09, 0.50; p = 0.005). On the other hand, the 30-day and in-hospital mortality were lower in patients who received TCZ compared to the control group (HR: 0.45, 95% CI: 0.27, 0.73; p = 0.01 and HR: 0.63, 95% CI: 0.41, 0.96; p = 0.03, respectively). The use of TCZ in this population was associated with a statistically significantly higher rate of AKI while improving the overall survival on the other hand. Further research is needed to assess the risks and benefits of TCZ treatment in critically ill COVID-19 patients with renal impairment.
Collapse
Affiliation(s)
- Ohoud Aljuhani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid Al Sulaiman
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Saudi Critical Care Pharmacy Research (SCAPE) Platform, Riyadh, Saudi Arabia
| | - Ghazwa B Korayem
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Aisha Alharbi
- Pharmaceutical Care Department, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Ali F. Altebainawi
- Pharmaceutical Care Services, King Salman Specialist Hospital, Hail Health Cluster, Ministry of Health, Hail, Saudi Arabia
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Shatha A. Aldkheel
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sarah G. Alotaibi
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Hanan Alshareef
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Samiah Alsohimi
- Pharmaceutical Care Services, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
- Pharmaceutical Care Services, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Mashael AlFaifi
- Pharmaceutical Care Department, King Saud Medical City, Riyadh, Saudi Arabia
| | - Abdulrahman Al Shaya
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haifa A. Alhaidal
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Raghad M. Alsubaie
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hessah Alrashidi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Khalid J. Albarqi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Dalal T. Alangari
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Reem M. Alanazi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Noora M. Altaher
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hasan M. Al-Dorzi
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Intensive Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Alrajhi NN. Post-COVID-19 pulmonary fibrosis: An ongoing concern. Ann Thorac Med 2023; 18:173-181. [PMID: 38058790 PMCID: PMC10697304 DOI: 10.4103/atm.atm_7_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/05/2023] [Accepted: 07/10/2023] [Indexed: 12/08/2023] Open
Abstract
Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 rapidly spread across the globe causing over 6 million deaths and major compromization of health facilities. The vast majority of survivors post-COVID-19 are left with variable degrees of health sequelae including pulmonary, neurological, psychological, and cardiovascular complications. Post-COVID-19 pulmonary fibrosis is one of the major concerns arising after the recovery from this pandemic. Risk factors for post-COVID-19 pulmonary fibrosis include age, male sex, and the severity of COVID-19 disease. High-resolution computed tomography provides diagnostic utility to diagnose pulmonary fibrosis as it provides more details regarding the pattern and the extent of pulmonary fibrosis. Emerging data showing similarities between post-COVID-19 pulmonary fibrosis and idiopathic pulmonary fibrosis, finding that needs further exploration. The management of post-COVID-19 pulmonary fibrosis depends on many factors but largely relies on excluding other causes of pulmonary fibrosis, the extent of fibrosis, and physiological impairment. Treatment includes immunosuppressants versus antifibrotics or both.
Collapse
Affiliation(s)
- Nuha Nasser Alrajhi
- Department of Medicine, Pulmonary Unit, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Ciabatti M, Zocchi C, Olivotto I, Bolognese L, Pieroni M. Myocarditis and COVID-19 related issues. Glob Cardiol Sci Pract 2023; 2023:e202328. [PMID: 38404624 PMCID: PMC10886760 DOI: 10.21542/gcsp.2023.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/12/2023] [Indexed: 02/27/2024] Open
Abstract
The recent COVID-19 (Coronavirus Disease 2019) pandemic by SARS-CoV2 infection has caused millions of deaths and hospitalizations across the globe. In the early pandemic phases, the infection had been initially considered a primary pulmonary disease. However, increasing evidence has demonstrated a wide range of possible cardiac involvement. Most of systemic and cardiac damage is likely sustained by a complex interplay between inflammatory, immune-related and thrombotic mechanisms. Biventricular failure and myocardial damage with elevation of cardiac biomarkers have been reported in COVID-19 patients, although histological demonstration of acute myocarditis has been rarely documented. Indeed while cardiac magnetic resonance findings include different patterns of myocardial involvement in terms of late gadolinium enhancement, histological data from necropsy and endomyocardial biopsy showed peculiar inflammatory patterns, mostly composed by macrophages. On the other hand COVID-19 vaccines based on mRN technology have been also associated with increased risk of myocarditis. COVID-19 and mRNA vaccine-related myocarditis present different clinical and imaging presentations and recent data suggest the presence of distinctive immunological mechanisms involved.
Collapse
Affiliation(s)
| | - Chiara Zocchi
- Cardiovascular Department, San Donato Hospital, Arezzo, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Meyer Children Hospital, Florence, Italy
| | | | | |
Collapse
|
11
|
Heron L, Buitrago-Garcia D, Ipekci AM, Baumann R, Imeri H, Salanti G, Counotte MJ, Low N. How to update a living systematic review and keep it alive during a pandemic: a practical guide. Syst Rev 2023; 12:156. [PMID: 37660117 PMCID: PMC10474670 DOI: 10.1186/s13643-023-02325-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 08/17/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND The covid-19 pandemic has highlighted the role of living systematic reviews. The speed of evidence generated during the covid-19 pandemic accentuated the challenges of managing high volumes of research literature. METHODS In this article, we summarise the characteristics of ongoing living systematic reviews on covid-19, and we follow a life cycle approach to describe key steps in a living systematic review. RESULTS We identified 97 living systematic reviews on covid-19, published up to 7th November 2022, which focused mostly on the effects of pharmacological interventions (n = 46, 47%) or the prevalence of associated conditions or risk factors (n = 30, 31%). The scopes of several reviews overlapped considerably. Most living systematic reviews included both observational and randomised study designs (n = 45, 46%). Only one-third of the reviews has been updated at least once (n = 34, 35%). We address practical aspects of living systematic reviews including how to judge whether to start a living systematic review, methods for study identification and selection, data extraction and evaluation, and give recommendations at each step, drawing from our own experience. We also discuss when it is time to stop and how to publish updates. CONCLUSIONS Methods to improve the efficiency of searching, study selection, and data extraction using machine learning technologies are being developed, their performance and applicability, particularly for reviews based on observational study designs should improve, and ways of publishing living systematic reviews and their updates will continue to evolve. Finally, knowing when to end a living systematic review is as important as knowing when to start.
Collapse
Affiliation(s)
- Leonie Heron
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Diana Buitrago-Garcia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School of Health Sciences, University of Bern, Bern, Switzerland
| | - Aziz Mert Ipekci
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Rico Baumann
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Hira Imeri
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Jacobs Center for Productive Youth Development, University of Zurich, Zürich, Switzerland
| | - Georgia Salanti
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Michel Jacques Counotte
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.
| |
Collapse
|
12
|
Luo J, Chen Z, Liu D, Li H, He S, Zeng L, Yang M, Liu Z, Xiao X, Zhang L. Methodological quality and reporting quality of COVID-19 living systematic review: a cross-sectional study. BMC Med Res Methodol 2023; 23:175. [PMID: 37525117 PMCID: PMC10388517 DOI: 10.1186/s12874-023-01980-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/18/2023] [Indexed: 08/02/2023] Open
Abstract
OBJECTIVES The main objective of this study is to evaluate the methodological quality and reporting quality of living systematic reviews (LSRs) on Coronavirus disease 2019 (COVID-19), while the secondary objective is to investigate potential factors that may influence the overall quality of COVID-19 LSRs. METHODS Six representative databases, including Medline, Excerpta Medica Database (Embase), Cochrane Library, China national knowledge infrastructure (CNKI), Wanfang Database, and China Science, Technology Journal Database (VIP) were systematically searched for COVID-19 LSRs. Two authors independently screened articles, extracted data, and then assessed the methodological and reporting quality of COVID-19 LSRs using the "A Measurement Tool to Assess systematic Reviews-2" (AMSTAR-2) tool and "Preferred Reporting Items for Systematic reviews and Meta-Analyses" (PRISMA) 2020 statement, respectively. Univariate linear regression and multivariate linear regression were used to explore eight potential factors that might affect the methodological quality and reporting quality of COVID-19 LSRs. RESULTS A total of 64 COVID-19 LSRs were included. The AMSTAR-2 evaluation results revealed that the number of "yes" responses for each COVID-19 LSR was 13 ± 2.68 (mean ± standard deviation). Among them, 21.9% COVID-19 LSRs were rated as "high", 4.7% as "moderate", 23.4% as "low", and 50% as "critically low". The evaluation results of the PRISMA 2020 statement showed that the sections with poor adherence were methods, results and other information. The number of "yes" responses for each COVID-19 LSR was 21 ± 4.18 (mean ± standard deviation). The number of included studies and registration are associated with better methodological quality; the number of included studies and funding are associated with better reporting quality. CONCLUSIONS Improvement is needed in the methodological and reporting quality of COVID-19 LSRs. Researchers conducting COVID-19 LSRs should take note of the quality-related factors identified in this study to generate evidence-based evidence of higher quality.
Collapse
Affiliation(s)
- Jiefeng Luo
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhe Chen
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dan Liu
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hailong Li
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Siyi He
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Linan Zeng
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Mengting Yang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zheng Liu
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xue Xiao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Lingli Zhang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China.
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China.
- NMPA Key Laboratory for Technical Research On Drug Products In Vitro and In Vivo Correlation, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Fischer AL, Messer S, Riera R, Martimbianco ALC, Stegemann M, Estcourt LJ, Weibel S, Monsef I, Andreas M, Pacheco RL, Skoetz N. Antiplatelet agents for the treatment of adults with COVID-19. Cochrane Database Syst Rev 2023; 7:CD015078. [PMID: 37489818 PMCID: PMC10368416 DOI: 10.1002/14651858.cd015078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
BACKGROUND Severe coronavirus disease 2019 (COVID-19) can cause thrombotic events that lead to severe complications or death. Antiplatelet agents, such as acetylsalicylic acid, have been shown to effectively reduce thrombotic events in other diseases: they could influence the course of COVID-19 in general. OBJECTIVES To assess the efficacy and safety of antiplatelets given with standard care compared to no treatment or standard care (with/without placebo) for adults with COVID-19. SEARCH METHODS We searched the Cochrane COVID-19 Study Register (which comprises MEDLINE (PubMed), Embase, ClinicalTrials.gov, WHO ICTRP, medRxiv, CENTRAL), Web of Science, WHO COVID-19 Global literature on coronavirus disease and the Epistemonikos COVID-19 L*OVE Platform to identify completed and ongoing studies without language restrictions to December 2022. SELECTION CRITERIA We followed standard Cochrane methodology. We included randomised controlled trials (RCTs) evaluating antiplatelet agents for the treatment of COVID-19 in adults with COVID-19, irrespective of disease severity, gender or ethnicity. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies, we used the Cochrane risk of bias tool (RoB 2) for RCTs. We rated the certainty of evidence using the GRADE approach for the outcomes. MAIN RESULTS Antiplatelets plus standard care versus standard care (with/without placebo) Adults with a confirmed diagnosis of moderate to severe COVID-19 We included four studies (17,541 participants) that recruited hospitalised people with a confirmed diagnosis of moderate to severe COVID-19. A total of 8964 participants were analysed in the antiplatelet arm (either with cyclooxygenase inhibitors or P2Y12 inhibitors) and 8577 participants in the control arm. Most people were older than 50 years and had comorbidities such as hypertension, lung disease or diabetes. The studies were conducted in high- to lower middle-income countries prior to wide-scale vaccination programmes. Antiplatelets compared to standard care: - probably result in little to no difference in 28-day mortality (risk ratio (RR) 0.95, 95% confidence interval (CI) 0.85 to 1.05; 3 studies, 17,249 participants; moderate-certainty evidence). In absolute terms, this means that for every 177 deaths per 1000 people not receiving antiplatelets, there were 168 deaths per 1000 people who did receive the intervention (95% CI 151 to 186 per 1000 people); - probably result in little to no difference in worsening (new need for invasive mechanical ventilation or death up to day 28) (RR 0.95, 95% CI 0.90 to 1.01; 2 studies, 15,266 participants; moderate-certainty evidence); - probably result in little to no difference in improvement (participants discharged alive up to day 28) (RR 1.00, 95% CI 0.96 to 1.04; 2 studies, 15,454 participants; moderate-certainty evidence); - probably result in a slight reduction of thrombotic events at longest follow-up (RR 0.90, 95% CI 0.80 to 1.02; 4 studies, 17,518 participants; moderate-certainty evidence); - may result in a slight increase in serious adverse events at longest follow-up (Peto odds ratio (OR) 1.57, 95% CI 0.48 to 5.14; 1 study, 1815 participants; low-certainty evidence), but non-serious adverse events during study treatment were not reported; - probably increase the occurrence of major bleeding events at longest follow-up (Peto OR 1.68, 95% CI 1.29 to 2.19; 4 studies, 17,527 participants; moderate-certainty evidence). Adults with a confirmed diagnosis of asymptomatic SARS-CoV-2 infection or mild COVID-19 We included two RCTs allocating participants, of whom 4209 had confirmed mild COVID-19 and were not hospitalised. A total of 2109 participants were analysed in the antiplatelet arm (treated with acetylsalicylic acid) and 2100 participants in the control arm. No study included people with asymptomatic SARS-CoV-2 infection. Antiplatelets compared to standard care: - may result in little to no difference in all-cause mortality at day 45 (Peto OR 1.00, 95% CI 0.45 to 2.22; 2 studies, 4209 participants; low-certainty evidence); - may slightly decrease the incidence of new thrombotic events up to day 45 (Peto OR 0.37, 95% CI 0.09 to 1.46; 2 studies, 4209 participants; low-certainty evidence); - may make little or no difference to the incidence of serious adverse events up to day 45 (Peto OR 1.00, 95% CI 0.60 to 1.64; 1 study, 3881 participants; low-certainty evidence), but non-serious adverse events were not reported. The evidence is very uncertain about the effect of antiplatelets on the following outcomes (compared to standard care plus placebo): - admission to hospital or death up to day 45 (Peto OR 0.79, 95% CI 0.57 to 1.10; 2 studies, 4209 participants; very low-certainty evidence); - major bleeding events up to longest follow-up (no event occurred in 328 participants; very low-certainty evidence). Quality of life and adverse events during study treatment were not reported. AUTHORS' CONCLUSIONS In people with confirmed or suspected COVID-19 and moderate to severe disease, we found moderate-certainty evidence that antiplatelets probably result in little to no difference in 28-day mortality, clinical worsening or improvement, but probably result in a slight reduction in thrombotic events. They probably increase the occurrence of major bleeding events. Low-certainty evidence suggests that antiplatelets may result in a slight increase in serious adverse events. In people with confirmed COVID-19 and mild symptoms, we found low-certainty evidence that antiplatelets may result in little to no difference in 45-day mortality and serious adverse events, and may slightly reduce thrombotic events. The effects on the combined outcome admission to hospital or death up to day 45 and major bleeding events are very uncertain. Quality of life was not reported. Included studies were conducted in high- to lower middle-income settings using antiplatelets prior to vaccination roll-outs. We identified a lack of evidence concerning quality of life assessments, adverse events and people with asymptomatic infection. The 14 ongoing and three completed, unpublished RCTs that we identified in trial registries address similar settings and research questions as in the current body of evidence. We expect to incorporate the findings of these studies in future versions of this review.
Collapse
Affiliation(s)
- Anna-Lena Fischer
- Department of Anaesthesia and Intensive care, Universitätsklinikum Leipzig, 04103 Leipzig, Germany
| | - Sarah Messer
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Rachel Riera
- Cochrane Brazil Rio de Janeiro, Cochrane, Petrópolis, Brazil
- Center of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Núcleo de Ensino e Pesquisa em Saúde Baseada em Evidências, Avaliação Tecnológica em Saúde e Eduação em Saúde (NEP-Sbeats), Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
| | - Ana Luiza C Martimbianco
- Cochrane Brazil Rio de Janeiro, Cochrane, Petrópolis, Brazil
- Center of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Universidade Metropolitana de Santos (UNIMES), Santos, Brazil
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Stephanie Weibel
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marike Andreas
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Rafael L Pacheco
- Center of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Núcleo de Ensino e Pesquisa em Saúde Baseada em Evidências, Avaliação Tecnológica em Saúde e Eduação em Saúde (NEP-Sbeats), Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
- Centro de Pesquisa Médica, Centro Universitário São Camilo (CUSC), São Paulo, Brazil
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Wang K, Fenton BT, Deng Y, Anthony SE, Dao VX, Schindler E, Lipton RB, Guirguis A, Skanderson M, Seng EK, Sico JJ. Calcitonin Gene-Related Peptide Monoclonal Antibodies and Risk of SARS-CoV-2 Infection and Severe COVID-19 Outcomes Among Veterans With Migraine Disorder. JAMA Netw Open 2023; 6:e2326371. [PMID: 37523183 PMCID: PMC10391301 DOI: 10.1001/jamanetworkopen.2023.26371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/15/2023] [Indexed: 08/01/2023] Open
Abstract
Importance Calcitonin gene-related peptide (CGRP), a neuropeptide involved in migraine pathophysiology, is also a key neuroimmune modulator. CGRP antagonists may help mitigate the hyperinflammatory response observed in patients with COVID-19; however, findings from the literature are contradictory, and to date, no study has investigated the safety and effectiveness of CGRP antagonists against COVID-19. Objective To evaluate the association between CGRP monoclonal antibody (mAb) treatment and risk of SARS-CoV-2 infection and sequela hospitalization, requiring supplemental oxygen, use of mechanical ventilation, or death. Design, Setting, and Participants This retrospective cohort study analyzed the electronic health records of US veterans aged 18 to 65 years who were diagnosed with migraine disorder and were at risk of COVID-19 between January 20, 2020, and May 19, 2022. Exposure Initiation of CGRP mAbs. Main Outcomes and Measures The main outcome was cumulative incidence of SARS-CoV-2 infection. Odds of 30-day hospitalization, requiring supplemental oxygen, use of mechanical ventilation, or death were secondary outcomes. Results Among 8 178 652 eligible person-trials (354 294 veterans), 9992 (mean [SD] age, 46.0 [9.5] years; 53.9% male) initiated CGRP mAbs and 8 168 660 (mean [SD] age, 46.6 [10.2] years; 65.7% male) did not initiate CGRP mAbs. Over a 28-month follow-up period, 1247 initiators (12.5%) and 780 575 noninitiators (9.6%) tested positive for SARS-CoV-2. After censoring persons who deviated from treatment, the incidence was 7.4 cases per 1000 person-months among initiators and 6.9 per 1000 person-months among noninitiators. The inverse probability-weighted observational analogs of intention-to-treat and per-protocol hazard ratios were 0.95 (95% CI, 0.89-1.01) and 0.93 (95% CI, 0.86-1.02), respectively. No significant differences in the likelihood of hospitalization (odds ratio [OR], 0.93; 95% CI, 0.62-1.41), requiring supplemental oxygen (OR, 0.77; 95% CI, 0.45-1.30), use of mechanical ventilation (OR, 0.85; 95% CI, 0.26-2.84), or death (OR, 0.67; 95% CI, 0.09-5.23) were observed between CGRP mAb initiators and noninitiators who tested positive for SARS-CoV-2. Conclusions and Relevance In this cohort study, CGRP mAb treatment was not associated with positive SARS-CoV-2 test results or risk of severe COVID-19 outcomes, suggesting that CGRP mAbs may be used for migraine prevention during the COVID-19 pandemic. Given the few events of requiring supplemental oxygen, use of mechanical ventilation, and death, replication analysis in a larger sample of patients later in the course of disease is warranted.
Collapse
Affiliation(s)
- Kaicheng Wang
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
- Yale Center for Analytic Sciences, Yale School of Public Health, New Haven, Connecticut
| | - Brenda T. Fenton
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
- Pain Research, Informatics, Multi-morbidities, and Education Center, VA Connecticut Healthcare System, West Haven
| | - Yanhong Deng
- Yale Center for Analytic Sciences, Yale School of Public Health, New Haven, Connecticut
| | - Sarah E. Anthony
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Vinh X. Dao
- Headache Center of Excellence, VA Minneapolis Health Care System, Minneapolis, Minnesota
| | - Emmanuelle Schindler
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
- Headache Center of Excellence, VA Connecticut Healthcare System, West Haven
| | - Richard B. Lipton
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
| | - Alexander Guirguis
- Headache Center of Excellence, VA Connecticut Healthcare System, West Haven
| | - Melissa Skanderson
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
| | - Elizabeth K. Seng
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
- Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, New York
| | - Jason J. Sico
- Research, Education, Evaluation and Engagement Activities Center for Headache, Headache Centers of Excellence, US Department of Veterans Affairs, Orange, Connecticut
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
- Headache Center of Excellence, VA Connecticut Healthcare System, West Haven
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
15
|
Ghosn L, Assi R, Evrenoglou T, Buckley BS, Henschke N, Probyn K, Riveros C, Davidson M, Graña C, Bonnet H, Jarde A, Ávila C, Nejstgaard CH, Menon S, Ferrand G, Kapp P, Breuer C, Schmucker C, Sguassero Y, Nguyen TV, Devane D, Meerpohl JJ, Rada G, Hróbjartsson A, Grasselli G, Tovey D, Ravaud P, Chaimani A, Boutron I. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev 2023; 6:CD013881. [PMID: 37260086 PMCID: PMC10237088 DOI: 10.1002/14651858.cd013881.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
BACKGROUND It has been reported that people with COVID-19 and pre-existing autoantibodies against type I interferons are likely to develop an inflammatory cytokine storm responsible for severe respiratory symptoms. Since interleukin 6 (IL-6) is one of the cytokines released during this inflammatory process, IL-6 blocking agents have been used for treating people with severe COVID-19. OBJECTIVES To update the evidence on the effectiveness and safety of IL-6 blocking agents compared to standard care alone or to a placebo for people with COVID-19. SEARCH METHODS We searched the World Health Organization (WHO) International Clinical Trials Registry Platform, the Living OVerview of Evidence (L·OVE) platform, and the Cochrane COVID-19 Study Register to identify studies on 7 June 2022. SELECTION CRITERIA We included randomized controlled trials (RCTs) evaluating IL-6 blocking agents compared to standard care alone or to placebo for people with COVID-19, regardless of disease severity. DATA COLLECTION AND ANALYSIS Pairs of researchers independently conducted study selection, extracted data and assessed risk of bias. We assessed the certainty of evidence using the GRADE approach for all critical and important outcomes. In this update we amended our protocol to update the methods used for grading evidence by establishing minimal important differences for the critical outcomes. MAIN RESULTS This update includes 22 additional trials, for a total of 32 trials including 12,160 randomized participants all hospitalized for COVID-19 disease. We identified a further 17 registered RCTs evaluating IL-6 blocking agents without results available as of 7 June 2022. The mean age range varied from 56 to 75 years; 66.2% (8051/12,160) of enrolled participants were men. One-third (11/32) of included trials were placebo-controlled. Twenty-two were published in peer-reviewed journals, three were reported as preprints, two trials had results posted only on registries, and results from five trials were retrieved from another meta-analysis. Eight were funded by pharmaceutical companies. Twenty-six included studies were multicenter trials; four were multinational and 22 took place in single countries. Recruitment of participants occurred between February 2020 and June 2021, with a mean enrollment duration of 21 weeks (range 1 to 54 weeks). Nineteen trials (60%) had a follow-up of 60 days or more. Disease severity ranged from mild to critical disease. The proportion of participants who were intubated at study inclusion also varied from 5% to 95%. Only six trials reported vaccination status; there were no vaccinated participants included in these trials, and 17 trials were conducted before vaccination was rolled out. We assessed a total of six treatments, each compared to placebo or standard care. Twenty trials assessed tocilizumab, nine assessed sarilumab, and two assessed clazakizumab. Only one trial was included for each of the other IL-6 blocking agents (siltuximab, olokizumab, and levilimab). Two trials assessed more than one treatment. Efficacy and safety of tocilizumab and sarilumab compared to standard care or placebo for treating COVID-19 At day (D) 28, tocilizumab and sarilumab probably result in little or no increase in clinical improvement (tocilizumab: risk ratio (RR) 1.05, 95% confidence interval (CI) 1.00 to 1.11; 15 RCTs, 6116 participants; moderate-certainty evidence; sarilumab: RR 0.99, 95% CI 0.94 to 1.05; 7 RCTs, 2425 participants; moderate-certainty evidence). For clinical improvement at ≥ D60, the certainty of evidence is very low for both tocilizumab (RR 1.10, 95% CI 0.81 to 1.48; 1 RCT, 97 participants; very low-certainty evidence) and sarilumab (RR 1.22, 95% CI 0.91 to 1.63; 2 RCTs, 239 participants; very low-certainty evidence). The effect of tocilizumab on the proportion of participants with a WHO Clinical Progression Score (WHO-CPS) of level 7 or above remains uncertain at D28 (RR 0.90, 95% CI 0.72 to 1.12; 13 RCTs, 2117 participants; low-certainty evidence) and that for sarilumab very uncertain (RR 1.10, 95% CI 0.90 to 1.33; 5 RCTs, 886 participants; very low-certainty evidence). Tocilizumab reduces all cause-mortality at D28 compared to standard care/placebo (RR 0.88, 95% CI 0.81 to 0.94; 18 RCTs, 7428 participants; high-certainty evidence). The evidence about the effect of sarilumab on this outcome is very uncertain (RR 1.06, 95% CI 0.86 to 1.30; 9 RCTs, 3305 participants; very low-certainty evidence). The evidence is uncertain for all cause-mortality at ≥ D60 for tocilizumab (RR 0.91, 95% CI 0.80 to 1.04; 9 RCTs, 2775 participants; low-certainty evidence) and very uncertain for sarilumab (RR 0.95, 95% CI 0.84 to 1.07; 6 RCTs, 3379 participants; very low-certainty evidence). Tocilizumab probably results in little to no difference in the risk of adverse events (RR 1.03, 95% CI 0.95 to 1.12; 9 RCTs, 1811 participants; moderate-certainty evidence). The evidence about adverse events for sarilumab is uncertain (RR 1.12, 95% CI 0.97 to 1.28; 4 RCT, 860 participants; low-certainty evidence). The evidence about serious adverse events is very uncertain for tocilizumab (RR 0.93, 95% CI 0.81 to 1.07; 16 RCTs; 2974 participants; very low-certainty evidence) and uncertain for sarilumab (RR 1.09, 95% CI 0.97 to 1.21; 6 RCTs; 2936 participants; low-certainty evidence). Efficacy and safety of clazakizumab, olokizumab, siltuximab and levilimab compared to standard care or placebo for treating COVID-19 The evidence about the effects of clazakizumab, olokizumab, siltuximab, and levilimab comes from only one or two studies for each blocking agent, and is uncertain or very uncertain. AUTHORS' CONCLUSIONS In hospitalized people with COVID-19, results show a beneficial effect of tocilizumab on all-cause mortality in the short term and probably little or no difference in the risk of adverse events compared to standard care alone or placebo. Nevertheless, both tocilizumab and sarilumab probably result in little or no increase in clinical improvement at D28. Evidence for an effect of sarilumab and the other IL-6 blocking agents on critical outcomes is uncertain or very uncertain. Most of the trials included in our review were done before the waves of different variants of concern and before vaccination was rolled out on a large scale. An additional 17 RCTs of IL-6 blocking agents are currently registered with no results yet reported. The number of pending studies and the number of participants planned is low. Consequently, we will not publish further updates of this review.
Collapse
Affiliation(s)
- Lina Ghosn
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Rouba Assi
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Theodoros Evrenoglou
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | | | | | | | - Carolina Riveros
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Mauricia Davidson
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Carolina Graña
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Hillary Bonnet
- Cochrane France, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Alexander Jarde
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | | | - Camilla Hansen Nejstgaard
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | | | | | - Philipp Kapp
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Breuer
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Christine Schmucker
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | | | | | - Declan Devane
- Evidence Synthesis Ireland, Galway, Ireland
- Cochrane Ireland and HRB-Trials Methodology Research Network, Galway, Ireland
- University of Galway, Galway, Ireland
| | - Joerg J Meerpohl
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Gabriel Rada
- Epistemonikos Foundation, Santiago, Chile
- UC Evidence Center, Cochrane Chile Associated Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asbjørn Hróbjartsson
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Giacomo Grasselli
- Department of Anesthesia, Intensive Care and Emergency Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | | | - Philippe Ravaud
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Anna Chaimani
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Isabelle Boutron
- Cochrane France, Paris, France
- Centre d'Epidémiologie Clinique, AP-HP, Hôpital Hôtel Dieu, F-75004, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| |
Collapse
|
16
|
Liu Q, Ma F, Zhong Y, Wang G, Hu L, Zhang Y, Xie J. Efficacy and safety of human umbilical cord-derived mesenchymal stem cells for COVID-19 pneumonia: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2023; 14:118. [PMID: 37143167 PMCID: PMC10159228 DOI: 10.1186/s13287-023-03286-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Elevated levels of inflammatory factors are associated with poor prognosis in coronavirus disease-19 (COVID-19). However, mesenchymal stem cells (MSCs) have immunomodulatory functions. Accordingly, this meta-analysis aimed to determine the efficacy and safety of MSC-based therapy in patients with COVID-19 pneumonia. METHODS Online global databases were used to find relevant studies. Two independent researchers then selected and evaluated the studies for suitability while the Cochrane risk of bias tool determined the quality of all articles and Cochran's Q test and I2 index assessed the degree of heterogeneity in the principal studies. Statistical analysis was performed using Review Manager software, and the effect of each study on the overall estimate was evaluated by sensitivity analysis. RESULTS Seven studies were included in the meta-analysis, and all MSCs used in the trials were acquired from the umbilical cord. The results of these studies (n = 328) indicated that patients with COVID-19 pneumonia who received MSCs had a 0.58 risk of death compared with controls (95% CI = 0.38, 0.87; P = 0.53; I2 = 0%). In terms of inflammatory biomarkers, MSCs reduced the levels of C-reactive protein (n = 88; MD = - 32.49; 95% CI = - 48.43, - 16.56; P = 0.46; I2 = 0%) and interferon-gamma (n = 44; SMD = - 1.23; 95% CI = - 1.89, - 0.57; P = 0.37; I2 = 0%) in severe COVID-19 patients but had no significant effect on interleukin-6 (n = 185; MD = - 0.75; 95% CI = - 7.76, 6.27; P = 0.57; I2 = 0%). A summary of the data revealed no significant differences in adverse events (n = 287) or serious adverse events (n = 229) between the MSC and control groups. CONCLUSIONS Infusion of umbilical cord-derived MSCs is an effective strategy for treating patients with COVID-19 pneumonia, with no noticeable adverse effects.
Collapse
Affiliation(s)
- Qinxue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China
| | - Fengjie Ma
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China
| | - Li Hu
- Department of Anesthesiology, Second Affiliated Hospital of Jiaxing University, No.1518 North Huancheng Road, Nanhu District, Jiaxing, 314000, China
| | - Yaping Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 East Qingchun Road, Jianggan District, Hangzhou, 310016, China.
| |
Collapse
|
17
|
Evrenoglou T, Boutron I, Seitidis G, Ghosn L, Chaimani A. metaCOVID: A web-application for living meta-analyses of COVID-19 trials. Res Synth Methods 2023; 14:479-488. [PMID: 36772980 DOI: 10.1002/jrsm.1627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/07/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Outputs from living evidence syntheses projects have been used widely during the pandemic by guideline developers to form evidence-based recommendations. However, the needs of different stakeholders cannot be accommodated by solely providing pre-defined non amendable numerical summaries. Stakeholders also need to understand the data and perform their own exploratory analyses. This requires resources, time, statistical expertise, software knowledge as well as relevant clinical expertise to avoid spurious conclusions. To assist them, we created the metaCOVID application which, based on automation processes, facilitates the fast exploration of the data and the conduct of sub-analyses tailored to end-users needs. metaCOVID has been created in R and is freely available as an R-Shiny application. Based on the COVID-NMA platform (https://covid-nma.com/) the application conducts living meta-analyses of randomized controlled trials related to COVID-19 treatments and vaccines for several outcomes. Several options are available for subgroup and sensitivity analyses. The results are presented in downloadable forest plots. We illustrate metaCOVID through three examples involving well-known treatments and vaccines for COVID-19. The application is freely available from https://covid-nma.com/metacovid/.
Collapse
Affiliation(s)
- Theodoros Evrenoglou
- Centre of Research in Epidemiology and Statistics (CRESS-U1153), Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Paris, France
| | - Isabelle Boutron
- Centre of Research in Epidemiology and Statistics (CRESS-U1153), Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Paris, France.,Centre d'Épidémiologie Clinique, AP-HP, Hôpital Hôtel-Dieu, Paris, France
| | - Georgios Seitidis
- Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece
| | - Lina Ghosn
- Centre of Research in Epidemiology and Statistics (CRESS-U1153), Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Paris, France.,Centre d'Épidémiologie Clinique, AP-HP, Hôpital Hôtel-Dieu, Paris, France
| | - Anna Chaimani
- Centre of Research in Epidemiology and Statistics (CRESS-U1153), Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Paris, France
| |
Collapse
|
18
|
Zhang JJ, Dong X, Liu GH, Gao YD. Risk and Protective Factors for COVID-19 Morbidity, Severity, and Mortality. Clin Rev Allergy Immunol 2023; 64:90-107. [PMID: 35044620 PMCID: PMC8767775 DOI: 10.1007/s12016-022-08921-5] [Citation(s) in RCA: 326] [Impact Index Per Article: 163.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
The outbreak of the coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become an evolving global health crisis. Currently, a number of risk factors have been identified to have a potential impact on increasing the morbidity of COVID-19 in adults, including old age, male sex, pre-existing comorbidities, and racial/ethnic disparities. In addition to these factors, changes in laboratory indices and pro-inflammatory cytokines, as well as possible complications, could indicate the progression of COVID-19 into a severe and critical stage. Children predominantly suffer from mild illnesses due to COVID-19. Similar to adults, the main risk factors in pediatric patients include age and pre-existing comorbidities. In contrast, supplementation with a healthy diet and sufficient nutrition, COVID-19 vaccination, and atopic conditions may act as protective factors against the infection of SARS-CoV-2. COVID-19 vaccination not only protects vulnerable individuals from SARS-CoV-2 infection, more importantly, it may also reduce the development of severe disease and death due to COVID-19. Currently used therapies for COVID-19 are off-label and empiric, and their impacts on the severity and mortality of COVID-19 are still unclear. The interaction between asthma and COVID-19 may be bidirectional and needs to be clarified in more studies. In this review, we highlight the clinical evidence supporting the rationale for the risk and protective factors for the morbidity, severity, and mortality of COVID-19.
Collapse
Affiliation(s)
- Jin-Jin Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiang Dong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, Hubei, China
| | - Guang-Hui Liu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, Hubei, China
| | - Ya-Dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China.
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
19
|
Ferrarini A, Vacca A, Solimando AG, Tavio M, Acquaviva R, Rocchi M, Nitti C, Salvi A, Menditto V, Luchetti Gentiloni MM, Russo A, Moretti M, Pavani M, Giacometti A, Bonifazi M, Zuccatosta L, Romani L, Racanelli V, Moroncini G, Gabrielli A, Pomponio G. Early administration of tofacitinib in COVID-19 pneumonitis: An open randomised controlled trial. Eur J Clin Invest 2023; 53:e13898. [PMID: 36380693 DOI: 10.1111/eci.13898] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Controversies on sub-populations most sensitive to therapy and the best timing of starting the treatment still surround the use of immunomodulatory drugs in COVID-19. OBJECTIVES We designed a multicentre open-label randomised controlled trial to test the effect of prompt adding of tofacitinib to standard therapy for hospitalised patients affected by mild/moderate COVID-19 pneumonitis. METHODS Patients admitted to three Italian hospitals affected by COVID-19 pneumonitis not requiring mechanical ventilation were randomised to receive standard treatment alone or tofacitinib (10 mg/bid) for 2 weeks, starting within the first 24 h from admission. RESULTS A total of 116 patients were randomised; 49 in the experimental arm completed the 14-day treatment period, 9 discontinued tofacitinib as the disease worsened and were included in the analysis, and 1 died of respiratory failure. All 58 control patients completed the study. Clinical and demographic characteristics were similar between the study groups. In the tofacitinib group, 9/58 (15.5%) patients progressed to noninvasive ventilation (CPAP) to maintain SO2 > 93%, invasive mechanical ventilation or death by day 14 was 15.5%, significantly less than in the control group (20/58, 34.4%, RR 0,45, RRR -55%, NNT 5; p = .018). No differences in severe adverse effect incidence had been observed across the groups. CONCLUSION High-dose tofacitinib therapy in patients with COVID pneumonitis is safe and may prevent deterioration to respiratory failure.
Collapse
Affiliation(s)
- Alessia Ferrarini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Gastroenterologia ed Endoscopia Digestiva, Ospedali Riuniti Marche Nord, Fano, Italy
| | - Angelo Vacca
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Antonio Giovanni Solimando
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy.,IRCCS Istituto Tumori "Giovanni Paolo II" Bari, Bari, Italy
| | - Marcello Tavio
- Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Rossella Acquaviva
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Marco Rocchi
- Statistica Medica, Dipartimento di Scienze Biomolecolari, Università di Urbino, Urbino, Italy
| | - Cinzia Nitti
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Aldo Salvi
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vincenzo Menditto
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | - Alessandro Russo
- Clinica di Malattie Infettive e Tropicali Dipartimento di Scienze Mediche e Chirurgiche Università "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Marco Moretti
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Marianna Pavani
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Andrea Giacometti
- Clinica di Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | - Laura Romani
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vito Racanelli
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Gianluca Moroncini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | | |
Collapse
|
20
|
Carrara E, Razzaboni E, Azzini AM, De Rui ME, Pinho Guedes MN, Gorska A, Giannella M, Bussini L, Bartoletti M, Arbizzani F, Palacios‐Baena ZR, Caponcello G, Maldonado N, Rodríguez‐Baño J, Visco C, Krampera M, Tacconelli E. Predictors of clinical evolution of SARS-CoV-2 infection in hematological patients: A systematic review and meta-analysis. Hematol Oncol 2023; 41:16-25. [PMID: 36238977 PMCID: PMC9874549 DOI: 10.1002/hon.3084] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/14/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023]
Abstract
Main aim of this systematic review is to quantify the risk and identify predictors of clinical evolution of SARS-CoV-2 in hematological patients compared to different control populations. Two independent reviewers screened the literature assessing clinical outcomes of SARS-CoV-2 infection in adult patients with active hematological malignancies published up to June 2021. Primary outcome was COVID-19 related mortality, secondary outcomes were hospital and intensive-care admission, mechanical ventilation (MV), and thromboembolic events. Variables related to study setting, baseline patients' demographic, comorbidities, underlying hematological disease, ongoing chemotherapy, COVID-19 presentation, and treatments were extracted. A total of 67 studies including 10,061 hematological patients and 111,143 controls were included. Most of the studies were retrospective cohorts (51 studies, 76%) and only 19 (13%) provided data for a control group. A significant increased risk of clinical progression in the hematological population compared to the controls was found in terms of COVID-19 related mortality (OR, 2.12; 95% CI, 1.77-2.54), hospitalization (OR, 1.98; 95% CI, 1.15-3.43), intensive-care admission (OR, 1.77; 95% CI, 1.38-2.26), and MV (OR, 2.17; 95% CI, 1.71-2.75). The risk remained significantly higher in the subgroup analysis comparing hematological patients versus solid cancer. Meta-regression analysis of uncontrolled studies showed that older age, male sex, and hypertension were significantly related to worse clinical outcomes of COVID-19 in hematological population. Older age and hypertension were found to be associated also to thromboembolic events. In conclusion, hematological patients have a higher risk of COVID-19 clinical progression compared to both the general population and to patients with solid cancer.
Collapse
Affiliation(s)
- Elena Carrara
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Elisa Razzaboni
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Anna Maria Azzini
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Maria Elena De Rui
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Mariana Nunes Pinho Guedes
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Anna Gorska
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| | - Maddalena Giannella
- Department of Medical and Surgical SciencesInfectious Diseases UnitIRCCS Policlinico Sant’OrsolaUniversity of BolognaBolognaItaly
| | - Linda Bussini
- Department of Medical and Surgical SciencesInfectious Diseases UnitIRCCS Policlinico Sant’OrsolaUniversity of BolognaBolognaItaly
| | - Michele Bartoletti
- Department of Medical and Surgical SciencesInfectious Diseases UnitIRCCS Policlinico Sant’OrsolaUniversity of BolognaBolognaItaly
| | - Federica Arbizzani
- Department of Medical and Surgical SciencesInfectious Diseases UnitIRCCS Policlinico Sant’OrsolaUniversity of BolognaBolognaItaly
| | - Zaira R. Palacios‐Baena
- Department of MedicineInfectious Diseases and Microbiology UnitHospital Universitario Virgen MacarenaUniversity of SevilleBiomedicine Institute of Seville (IBiS) / CSIC; and Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)SevilleSpain
| | - Giulia Caponcello
- Department of MedicineInfectious Diseases and Microbiology UnitHospital Universitario Virgen MacarenaUniversity of SevilleBiomedicine Institute of Seville (IBiS) / CSIC; and Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)SevilleSpain
| | - Natalia Maldonado
- Department of MedicineInfectious Diseases and Microbiology UnitHospital Universitario Virgen MacarenaUniversity of SevilleBiomedicine Institute of Seville (IBiS) / CSIC; and Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)SevilleSpain
| | - Jesús Rodríguez‐Baño
- Department of MedicineInfectious Diseases and Microbiology UnitHospital Universitario Virgen MacarenaUniversity of SevilleBiomedicine Institute of Seville (IBiS) / CSIC; and Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)SevilleSpain
| | - Carlo Visco
- Department of MedicineSection of HematologyUniversity of VeronaVeronaItaly
| | - Mauro Krampera
- Department of MedicineSection of HematologyUniversity of VeronaVeronaItaly
| | - Evelina Tacconelli
- Department of Diagnostics and Public HealthDivision of Infectious DiseasesUniversity of VeronaVeronaItaly
| |
Collapse
|
21
|
Singh M, Pushpakumar S, Bard N, Zheng Y, Homme RP, Mokshagundam SPL, Tyagi SC. Simulation of COVID-19 symptoms in a genetically engineered mouse model: implications for the long haulers. Mol Cell Biochem 2023; 478:103-119. [PMID: 35731343 PMCID: PMC9214689 DOI: 10.1007/s11010-022-04487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 01/24/2023]
Abstract
The ongoing pandemic (also known as coronavirus disease-19; COVID-19) by a constantly emerging viral agent commonly referred as the severe acute respiratory syndrome corona virus 2 or SARS-CoV-2 has revealed unique pathological findings from infected human beings, and the postmortem observations. The list of disease symptoms, and postmortem observations is too long to mention; however, SARS-CoV-2 has brought with it a whole new clinical syndrome in "long haulers" including dyspnea, chest pain, tachycardia, brain fog, exercise intolerance, and extreme fatigue. We opine that further improvement in delivering effective treatment, and preventive strategies would be benefited from validated animal disease models. In this context, we designed a study, and show that a genetically engineered mouse expressing the human angiotensin converting enzyme 2; ACE-2 (the receptor used by SARS-CoV-2 agent to enter host cells) represents an excellent investigative resource in simulating important clinical features of the COVID-19. The ACE-2 mouse model (which is susceptible to SARS-CoV-2) when administered with a recombinant SARS-CoV-2 spike protein (SP) intranasally exhibited a profound cytokine storm capable of altering the physiological parameters including significant changes in cardiac function along with multi-organ damage that was further confirmed via histological findings. More importantly, visceral organs from SP treated mice revealed thrombotic blood clots as seen during postmortem examination. Thus, the ACE-2 engineered mouse appears to be a suitable model for studying intimate viral pathogenesis thus paving the way for identification, and characterization of appropriate prophylactics as well as therapeutics for COVID-19 management.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Nia Bard
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yuting Zheng
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Rubens P Homme
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|
22
|
Plocque A, Mitri C, Lefèvre C, Tabary O, Touqui L, Philippart F. Should We Interfere with the Interleukin-6 Receptor During COVID-19: What Do We Know So Far? Drugs 2023; 83:1-36. [PMID: 36508116 PMCID: PMC9743129 DOI: 10.1007/s40265-022-01803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 12/14/2022]
Abstract
Severe manifestations of COVID-19 consist of acute respiratory distress syndrome due to an initially local reaction leading to a systemic inflammatory response that results in hypoxia. Many therapeutic approaches have been attempted to reduce the clinical consequences of an excessive immune response to viral infection. To date, systemic corticosteroid therapy is still the most effective intervention. More recently, new hope has emerged with the use of interleukin (IL)-6 receptor inhibitors (tocilizumab and sarilumab). However, the great heterogeneity of the methodology and results of published studies obfuscate the true value of this treatment, leading to a confusing synthesis in recent meta-analyses, and the persistence of doubts in terms of patient groups and the appropriate time to treat. Moreover, their effects on the anti-infectious or pro-healing response are still poorly studied. This review aims to clarify the potential role of IL-6 receptor inhibitors in the treatment of severe forms of COVID-19.
Collapse
Affiliation(s)
- Alexia Plocque
- Medical and Surgical Intensive Care Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Christie Mitri
- Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, 75012, Paris, France
| | - Charlène Lefèvre
- Medical and Surgical Intensive Care Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Olivier Tabary
- Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Inserm, 75012, Paris, France
| | - Lhousseine Touqui
- INSERM U938 Unit, St. Antoine Research Centre, Sorbona University, Paris, France
- Mucoviscidosis and Pulmonary Disease Units, Institute Pasteur, Paris, France
- Cystic fibrosis and Bronchial diseases team-INSERM U938, Institut Pasteur, Paris, France
| | - Francois Philippart
- Medical and Surgical Intensive Care Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France.
- Endotoxins, Structures and Host Response, Department of Microbiology, Institute for Integrative Biology of the Cell, UMR 9891 CNRS-CEA-Paris Saclay University, 98190, Gif-sur-Yvette, France.
| |
Collapse
|
23
|
O'Hayer PJ, Vasbinder A, Anderson E, Catalan T, Bitterman B, Khaleel I, Erne G, Tekumulla A, Tilley C, Presswalla F, Nelapudi N, Chen J, Tripathi M, Rochlen M, Rambo L, Sulaiman N, Blakely P, Huang Y, Zhao L, Pop-Busui R, Hayek SS. Evolution of Care and Outcomes Across Surges in Hospitalized Patients with Coronavirus Disease 2019. Am J Med 2023; 136:63-71.e1. [PMID: 36150511 PMCID: PMC9489963 DOI: 10.1016/j.amjmed.2022.08.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has unfolded in distinct surges. Understanding how surges differ may reveal important insights into the evolution of the pandemic and improve patient care. METHODS We leveraged the Michigan Medicine COVID-19 Cohort, a prospective observational study at an academic tertiary medical center that systematically enrolled 2309 consecutive patients hospitalized for COVID-19, comprising 5 distinct surges. RESULTS As the pandemic evolved, patients hospitalized for COVID-19 tended to have a lower burden of comorbidities and a lower inflammatory burden as measured by admission levels of C-reactive protein, ferritin, lactate dehydrogenase, and D-dimer. Use of hydroxychloroquine and azithromycin decreased substantially after Surge 1, while use of corticosteroids and remdesivir markedly increased (P < .001 for all). In-hospital mortality significantly decreased from 18.3% in Surge 1 to 5.3% in Surge 5 (P < .001). The need for mechanical ventilation significantly decreased from 42.5% in Surge 1 to 7.0% in Surge 5 (P < .001), while the need for renal replacement therapy decreased from 14.4% in Surge 1 to 2.3% in Surge 5 (P < .001). Differences in patient characteristics, treatments, and inflammatory markers accounted only partially for the differences in outcomes between surges. CONCLUSIONS The COVID-19 pandemic has evolved significantly with respect to hospitalized patient populations and therapeutic approaches, and clinical outcomes have substantially improved. Hospitalization after the first surge was independently associated with improved outcomes, even after controlling for relevant clinical covariates.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Grace Erne
- Department of Medicine, Division of Cardiology
| | | | | | | | | | - Jiazi Chen
- Department of Medicine, Division of Cardiology
| | | | | | - Loni Rambo
- Department of Medicine, Division of Cardiology
| | | | | | - Yiyuan Huang
- Department of Biostatistics, School of Public Health
| | - Lili Zhao
- Department of Biostatistics, School of Public Health
| | - Rodica Pop-Busui
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor
| | | |
Collapse
|
24
|
Wang KY, Shah P, Skavla B, Fayaaz F, Chi J, Rhodes JM. Vaccination efficacy in patients with chronic lymphocytic leukemia. Leuk Lymphoma 2023; 64:42-56. [PMID: 36270021 DOI: 10.1080/10428194.2022.2133538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a disorder of mature malignant B cells with multiple elements of immune dysfunction. Infections are common in CLL patients due to complex immunodeficiency. Vaccines are used as preventative measures for common diseases including influenza, pneumococcus, tetanus/diphtheria and shingles in the general population. Vaccines are utilized to mitigate this risk, although there have been some concerns regarding the efficacy of vaccines in the CLL population due to the inherent complex immune dysfunction associated with the disease. In this review, we describe the clinical and laboratory indicators for efficacy of the vaccines in the CLL population (including COVID-19, influenza, pneumonia, herpes zoster, and tetanus) and discuss immunization recommendations for patients with CLL.
Collapse
Affiliation(s)
- Kevin Y Wang
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Pratik Shah
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Brandon Skavla
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Fatima Fayaaz
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA
| | - Jeffrey Chi
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA
| | - Joanna M Rhodes
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
25
|
Elgohary MA, Ali A, El-Masry TA, Faidah H, Bantun F, Elkholy AM, Fahim JS, Elgamal NN, Mohamed ME, Seadawy MG, Helal AM, De Waard M, Shishtawy HM, El-Bouseary MM. Development and validation of a predictive scoring system for in-hospital mortality in COVID-19 Egyptian patients: a retrospective study. Sci Rep 2022; 12:22352. [PMID: 36572690 PMCID: PMC9791155 DOI: 10.1038/s41598-022-26471-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
SARS-CoV-2 virus has rapidly spread worldwide since December 2019, causing COVID-19 disease. In-hospital mortality is a common indicator for evaluating treatment outcomes. Therefore, the developing and validating a simple score system from observational data could assist in modulating the management procedures. A retrospective cohort study included all data records of patients with positive PCR for SARS-CoV-2. The factors that associated with mortality were analyzed, then allocation of potential predictors of mortality was executed using different logistic regression modeling, subsequently scoring system was developed from the most weighted predictors. The mortality rate of patients with COVID-19 pneumonia was 28.5% and 28.74%, respectively. The most significant factors that affected in-hospital mortality were old age (> 60 years), delay in hospital admission (> 4 days), high neutrophil/lymphocyte ratio "NLR" (> 3); higher computed tomography severity score; and CT-SS (> 20), in addition to using remdesivir and tocilizumab in the treatment protocol (P < 0.001 for all). The validity of the newly performed score was significant; the AUC was 85%, P < 0.001, and its prognostic utility was good; the AUC was 75%, P < 0.001. The prognostic utility of newly developed score system (EGY.Score) was excellent and could be used to adjust the treatment strategy of highly at-risk patients with COVID-19 pneumonia.
Collapse
Affiliation(s)
| | - Asmaa Ali
- Department of Pulmonary Medicine, Abbassia Chest Hospital, MOH, Cairo, Egypt ,grid.440785.a0000 0001 0743 511XDepartment of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013 P. R. China
| | - Thanaa A. El-Masry
- grid.412258.80000 0000 9477 7793Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Hani Faidah
- grid.412832.e0000 0000 9137 6644Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Farkad Bantun
- grid.412832.e0000 0000 9137 6644Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmad M. Elkholy
- Department of Tropical Medicine, Almaza Military Fever Hospital, Cairo, Egypt
| | - Jaklin S. Fahim
- Department of Microbiology, Almaza Military Fever Hospital, Cairo, Egypt
| | - Nabila N. Elgamal
- Department of Tropical Medicine, Almaza Military Fever Hospital, Cairo, Egypt
| | | | | | - Amro M. Helal
- Department of Public Health, Almaza Military Fever Hospital, Cairo, Egypt
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120 Saint-Egrève, France ,grid.4817.a0000 0001 2189 0784L’institut du Thorax, INSERM, CNRS, Univ Nantes, F-44007 Nantes, France ,grid.460782.f0000 0004 4910 6551Université de Nice Sophia-Antipolis, LabEx “Ion Channels, Science & Therapeutics”, F-06560 Valbonne, France
| | | | - Maisra M. El-Bouseary
- grid.412258.80000 0000 9477 7793Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
26
|
Gao Q, Yin X, Tan B, Wang J, Chen J, Zhao B, Yang Q, Li Z. Drug-induced liver injury following the use of tocilizumab or sarilumab in patients with coronavirus disease 2019. BMC Infect Dis 2022; 22:929. [PMID: 36503381 PMCID: PMC9742033 DOI: 10.1186/s12879-022-07896-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUNDS Interleukin-6 (IL-6) blockers including tocilizumab and sarilumab were approved by the U.S. Food and Drug Administration (FDA) in June 2021 for the treatment of patients with moderate to severe COVID-19. The use of sarilumab or tocilizumab in COVID-19 patients has been related to a reduction in mortality compared to standard care. Recent evidence has emerged concerning drug-induced liver injury (DILI) after sarilumab or tocilizumab applications in COVID-19 patients. AIMS The study aimed to estimate DILI associated with sarilumab or tocilizumab in treating moderate to severe patients infected with SARS-Cov-2. METHODS We conducted a retrospective pharmacovigilance study by data mining of the FDA's adverse event reporting systems (FAERS) database from the first quarter of 2004 to the fourth quarter of 2021 in confirmed COVID-19 patients. We analyzed DILI cases associated with tocilizumab or sarilumab in treating COVID-19 patients from the FAERS during this period. Disproportionality analysis and Bayesian analysis of COVID-19 patients were utilized for case analysis, and we also next compared the onset time and fatality rates of DILI following tocilizumab or sarilumab. RESULTS A total of 275 cases of TCZ or SAR-related DILI reports were extracted. A total of 192 AEs cases were related to tocilizumab (TCZ), and 83 were related to sarilumab (SAR). In patients treated with TCZ, most were < 75 years old (51.57%), with more male than female (46.35% vs. 13.02%). The correlation between IL-6 receptor antagonists and DILI was stronger in SAR (ROR = 12.94; 95%CI 9.6-17.44) than in TCZ (ROR = 1.33; 95%CI 1.14-1.55). The onset time of DILI was different between TCZ and SAR, and a significant difference was observed in TCZ than SAR (P < 0.0001). A significant difference was observed in the mortality rate of TCZ and SAR (P = 0.0009). DILI associated with COVID-19 patients treated with TCZ appeared to have earlier onset-time (1(0-46) day) VS. SAR (3.5(0-27) day). CONCLUSION This study shows strict monitor ought to be paid for TCZ or SAR when used for COVID-19 patients with poor liver function.
Collapse
Affiliation(s)
- Qian Gao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125 China
| | - Xuedong Yin
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125 China
| | - Boyu Tan
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Junshi Wang
- grid.259384.10000 0000 8945 4455Macau University of Science and Technology, Macau, China
| | - Jiayan Chen
- School of Nursing and Health, Shanghai Zhongqiao Vocational and Technical University, Shanghai, 201514 China
| | - Bin Zhao
- grid.413106.10000 0000 9889 6335Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730 China
| | - Qiaoling Yang
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.412540.60000 0001 2372 7462Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Zhiling Li
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| |
Collapse
|
27
|
Ziegler L, Lundström A, Havervall S, Thålin C, Gigante B. IL-6 signalling biomarkers in hospitalised patients with moderate to severe SARS-CoV-2 infection in a single centre study in Sweden. Cytokine 2022; 159:156020. [PMID: 36057230 PMCID: PMC9420722 DOI: 10.1016/j.cyto.2022.156020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/15/2022] [Accepted: 08/20/2022] [Indexed: 11/03/2022]
Abstract
Background Method Results Conclusion
Collapse
|
28
|
Efficacy and safety of nitazoxanide in treating SARS-CoV-2 infection: a systematic review and meta-analysis of blinded, placebo-controlled, randomized clinical trials. Eur J Clin Pharmacol 2022; 78:1813-1821. [PMID: 36066651 PMCID: PMC9446612 DOI: 10.1007/s00228-022-03380-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/30/2022] [Indexed: 12/15/2022]
Abstract
Purpose Nitazoxanide is a broad-spectrum antiparasitic that has been tested for COVID-19 due to its anti-inflammatory effects and in vitro antiviral activity. This study synthesized the best evidence on the efficacy and safety of nitazoxanide in COVID-19. Methods Searches for studies were performed in peer-reviewed and grey-literature from January 1, 2020 to May 23, 2022. The following elements were used to define eligibility criteria: (1) Population: individuals with COVID-19; (2) Intervention: nitazoxanide; (3) Comparison: placebo; (4) Outcomes: primary outcome was death, and secondary outcomes were viral load, positive RT-PCR status, serum biomarkers of inflammation, composite measure of disease progression (ICU admission or invasive mechanical ventilation), and any adverse events; (5) Study type: blinded, placebo-controlled, randomized clinical trials (RCTs). Treatment effects were reported as relative risk (RR) for dichotomous variables and standardized mean difference (SMD) for continuous variables with 95% confidence intervals (CI). Results Five blinded, placebo-controlled RCTs were included and enrolled individuals with mild or moderate SARS-CoV-2 infection. We found no difference between nitazoxanide and placebo in reducing viral load (SMD = − 0.16; 95% CI − 0.38 to 0.05) and the frequency of positive RTP-PCR results (RR = 0.92; 95% CI 0.81 to 1.06). In addition, there was no decreased risk for disease progression (RR = 0.63; 95% CI 0.38 to 1.04) and death (RR = 0.81; 95% CI 0.36 to 1.78) among patients receiving nitazoxanide. Patients with COVID-19 treated with nitazoxanide had decreased levels of white blood cells (SMD = − 0.15; 95% − 0.29 to − 0.02), lactate dehydrogenase (LDH) (SMD − 0.32; 95% − 0.52 to − 0.13), and D-dimer (SMD − 0.49; 95% CI − 0.68 to − 0.31) compared to placebo, but the magnitude of effect was considered small to moderate. Conclusion This systematic review showed no evidence of clinical benefits of the use of nitazoxanide to treat patients with mild or moderate COVID-19. In addition, we found a reduction in WBC, LDH, and D-dimer levels among nitazoxanide-treated patients, but the effect size was considered small to moderate. Supplementary Information The online version contains supplementary material available at 10.1007/s00228-022-03380-5.
Collapse
|
29
|
Coldewey SM, Neu C, Bloos F, Baumbach P, Schumacher U, Bauer M, Reuken P, Stallmach A. Infliximab in the treatment of patients with severe COVID-19 (INFLIXCOVID): protocol for a randomised, controlled, multicentre, open-label phase II clinical study. Trials 2022; 23:737. [PMID: 36056419 PMCID: PMC9438250 DOI: 10.1186/s13063-022-06566-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
Background Despite the intense global research endeavour to improve the treatment of patients with COVID-19, the current therapy remains insufficient, resulting in persisting high mortality. Severe cases are characterised by a systemic inflammatory reaction driven by the release of pro-inflammatory cytokines such as IL-6 and tumour-necrosis-factor alpha (TNF-α). TNF-α-blocking therapies have proved beneficial in patients with chronic inflammatory diseases and could therefore pose a new treatment option in COVID-19. Hitherto, no results from randomised controlled trials assessing the effectiveness and safety of infliximab—a monoclonal antibody targeting TNF-α—in the treatment of COVID-19 have been published. Methods In this phase-2 clinical trial, patients with COVID-19 and clinical and laboratory signs of hyperinflammation will be randomised to receive either one dose of infliximab (5 mg/kg body weight) in addition to the standard of care or the standard of care alone. The primary endpoint is the difference in 28-day mortality. Further assessments concern the safety of infliximab therapy in COVID-19 and the influence of infliximab on morbidity and the course of the disease. For the supplementary scientific programme, blood and urine samples are collected to assess concomitant molecular changes. The Ethics Committee of the Friedrich Schiller University Jena (2021-2236-AMG-ff) and the Paul-Ehrlich-Institute (4513/01) approved the study. Discussion The results of this study could influence the therapy of patients with COVID-19 and affect the course of the disease worldwide, as infliximab is globally available and approved by several international drug agencies. Trial registration The trial was registered at clinicaltrials.gov (NCT04922827, 11 June 2021) and at EudraCT (2021-002098-25, 19 May 2021).
Collapse
Affiliation(s)
- Sina M Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany. .,Septomics Research Centre, Jena University Hospital, Jena, Germany. .,Centre for Sepsis Control & Care (CSCC), Jena University Hospital, Jena, Germany.
| | - Charles Neu
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Frank Bloos
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Centre for Sepsis Control & Care (CSCC), Jena University Hospital, Jena, Germany
| | - Philipp Baumbach
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Ulrike Schumacher
- Centre for Clinical Studies, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Centre for Sepsis Control & Care (CSCC), Jena University Hospital, Jena, Germany
| | - Philipp Reuken
- Clinic for Internal Medicine IV, Jena University Hospital, Jena, Germany
| | - Andreas Stallmach
- Clinic for Internal Medicine IV, Jena University Hospital, Jena, Germany
| |
Collapse
|
30
|
Miller KD, Ashcraft AM, Pilkerton CS, Shrader CD. Seronegative immunity to SARS-CoV-2: a case study. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2022; 18:80. [PMID: 36042466 PMCID: PMC9425822 DOI: 10.1186/s13223-022-00715-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/31/2022] [Indexed: 12/15/2022]
Abstract
Background COVID-19 presents with a variable clinical course from asymptomatic to severe respiratory distress with nearly 25% mortality in mechanically-ventilated patients. As such, there is uncertainty regarding how host factors modulate the disease course. Case Presentation This report examines these factors in two geriatric patients with multiple comorbid conditions who were residents of the long-term care facility in West Virginia that was the epicenter of COVID-19 in the state. Each patient had substantial, unprotected exposure to SARS-CoV-2 with subsequent negative PCR and antibody testing. Conclusions These cases could represent an important step in understanding host factors that modulate the disease course and susceptibility of patients exposed to SARS-CoV-2, and illustrate the need for further research into host resistance relating to this pandemic.
Collapse
Affiliation(s)
- Kyle D Miller
- Department of Family Medicine, Medical Center Drive, West Virginia University, Box 9152, Morgantown, WV, 26506, USA
| | - Amie M Ashcraft
- Department of Family Medicine, Medical Center Drive, West Virginia University, Box 9152, Morgantown, WV, 26506, USA.
| | - Courtney S Pilkerton
- Department of Family Medicine, Medical Center Drive, West Virginia University, Box 9152, Morgantown, WV, 26506, USA
| | - Carl D Shrader
- Department of Family Medicine, Medical Center Drive, West Virginia University, Box 9152, Morgantown, WV, 26506, USA
| |
Collapse
|
31
|
Hoffmann J, Etati R, Brendel C, Neubauer A, Mack E. The Low Expression of Fc-Gamma Receptor III (CD16) and High Expression of Fc-Gamma Receptor I (CD64) on Neutrophil Granulocytes Mark Severe COVID-19 Pneumonia. Diagnostics (Basel) 2022; 12:2010. [PMID: 36010361 PMCID: PMC9407138 DOI: 10.3390/diagnostics12082010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022] Open
Abstract
Hyperinflammation through neutrophil granulocytes contributes to disease severity in COVID-19 pneumonia and promotes acute lung failure. Understanding the mechanisms of the dysregulations within the myeloid cell compartment may help to improve therapies for severe COVID-19 infection. Here, we investigated the immunopathological characteristics of circulating neutrophil granulocytes and monocytes in 16 patients with COVID-19 pneumonia by multiparameter flow cytometry in comparison to 9 patients with pulmonary infiltrates but without COVID-19. We correlated the immunophenotypes with the scores of the severity-of-disease classification system, APACHE-II. We found that the mean fluorescence intensity (MFI) of CD15, which is important for the transendothelial migration, was significantly reduced in the patients with COVID-19 (difference ± SD; 295.70 ± 117.50 MFI; p = 0.02). In addition, the granularity was significantly lower in the neutrophil granulocytes of patients with COVID-19 (difference ± SD; 1.11 ± 0.43 side-scatter ratio; p = 0.02). Moreover, the Fc-gamma receptor III (CD16) and Fc-gamma receptor I (CD64) on the neutrophil granulocytes were expressed discordantly with COVID-19 severity. CD16 correlated as inversely proportional (ρ = (-)0.72; 95% CI (-)0.92-(-)0.23; p = 0.01) and CD64 as proportional (ρ = 0.76; 95% CI 0.31-0.93; p = 0.01) with the APACHE-II scores of the patients. We conclude that the deviant expression of the Fc-gamma receptors might play role in a dysregulated antibody-mediated phagocytosis in severe cases of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Joerg Hoffmann
- Department of Hematology, Oncology and Immunology, University Hospital Giessen and Marburg, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany
| | | | | | | | | |
Collapse
|
32
|
Chen Z, Luo J, Li S, Xu P, Zeng L, Yu Q, Zhang L. Characteristics of Living Systematic Review for COVID-19. Clin Epidemiol 2022; 14:925-935. [PMID: 35958161 PMCID: PMC9359410 DOI: 10.2147/clep.s367339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose The systematic review aims to analyze and summarize the characteristics of living systematic review (LSR) for coronavirus disease 2019 (COVID-19). Methods Six databases including Medline, Excerpta Medica (Embase), Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang Database and China Science, and Technology Journal Database (VIP), were searched as the source of basic information and methodology of LSR. Descriptive analytical methods were used to analyze the included COVID-19 LSRs, and the study characteristics of COVID-19 LSRs were further assessed. Results Sixty-four COVID-19 LSRs were included. Eighty-nine point one percent of LSRs were published on Science Citation Index (SCI) journals, and 64.1% publication with an impact factor (IF) >5 and 17.2% with an IF >15 among SCI journals. The first unit of the published LSRs for COVID-19 came from 19 countries, with the largest contribution from the UK (17.2%, 11/64). Forty point six percent of LSRs for COVID-19 were related to therapeutics topic which was considered the most concerned perspective for LSRs for COVID-19. Seventy-six point six percent of LSRs focused on the general population, with less attention to children, pregnant women and the elderly. However, the LSR for COVID-19 was reported incomplete on “living” process, including 40.6% of studies without search frequency, 79.7% of studies without screening frequency, 20.3% of studies without update frequency, and 65.6% of studies without the timing or criteria of transitioning LSR out of living mode. Conclusion Although researchers in many countries have applied LSRs to COVID-19, most of the LSRs for COVID-19 were incomplete in reporting on the “living” process and less focused on special populations. This could reduce the confidence of health-care providers and policy makers in the results of COVID-19 LSR, thereby hindering the translation of evidence on COVID-19 LSR into clinical practice. It was necessary to explicitly enact preferred reporting items for systematic reviews and meta-analyses (PRISMA) to improve the reporting quality of LSR and support ongoing efforts of therapeutics research for special patients with COVID-19.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- West China School of Pharmacy, Sichuan University, Chengdu, People’s Republic of China
| | - Jiefeng Luo
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- West China School of Pharmacy, Sichuan University, Chengdu, People’s Republic of China
| | - Siyu Li
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- West China School of Medicine, Sichuan University, Chengdu, People’s Republic of China
| | - Peipei Xu
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- West China School of Medicine, Sichuan University, Chengdu, People’s Republic of China
| | - Linan Zeng
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
| | - Qin Yu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- National Drug Clinical Trial Institute, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Qin Yu, Email
| | - Lingli Zhang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- NMPA Key Laboratory for Technical Research on Drug Products in Vitro and in Vivo Correlation, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- Correspondence: Lingli Zhang, Email
| |
Collapse
|
33
|
Lebedeva A, Molodtsov I, Anisimova A, Berestovskaya A, Dukhin O, Elizarova A, Fitzgerald W, Fomina D, Glebova K, Ivanova O, Kalinskaya A, Lebedeva A, Lysenko M, Maryukhnich E, Misyurina E, Protsenko D, Rosin A, Sapozhnikova O, Sokorev D, Shpektor A, Vorobyeva D, Vasilieva E, Margolis L. Comprehensive Cytokine Profiling of Patients with COVID-19 Receiving Tocilizumab Therapy. Int J Mol Sci 2022; 23:7937. [PMID: 35887283 PMCID: PMC9316906 DOI: 10.3390/ijms23147937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by immune activation in response to viral spread, in severe cases leading to the development of cytokine storm syndrome (CSS) and increased mortality. Despite its importance in prognosis, the pathophysiological mechanisms of CSS in COVID-19 remain to be defined. Towards this goal, we analyzed cytokine profiles and their interrelation in regard to anti-cytokine treatment with tocilizumab in 98 hospitalized patients with COVID-19. We performed a multiplex measurement of 41 circulating cytokines in the plasma of patients on admission and 3-5 days after, during the follow-up. Then we analyzed the patient groups separated in two ways: according to the clusterization of their blood cytokines and based on the administration of tocilizumab therapy. Patients with and without CSS formed distinct clusters according to their cytokine concentration changes. However, the tocilizumab therapy, administered based on the standard clinical and laboratory criteria, did not fully correspond to those clusters of CSS. Furthermore, among all cytokines, IL-6, IL-1RA, IL-10, and G-CSF demonstrated the most prominent differences between patients with and without clinical endpoints, while only IL-1RA was prognostically significant in both groups of patients with and without tocilizumab therapy, decreasing in the former and increasing in the latter during the follow-up period. Thus, CSS in COVID-19, characterized by a correlated release of multiple cytokines, does not fully correspond to the standard parameters of disease severity. Analysis of the cytokine signature, including the IL-1RA level in addition to standard clinical and laboratory parameters may be useful to define the onset of a cytokine storm in COVID-19 as well as the indications for anti-cytokine therapy.
Collapse
Affiliation(s)
- Anna Lebedeva
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
| | - Ivan Molodtsov
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Alexandra Anisimova
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Anastasia Berestovskaya
- Clinical City Hospital №40, Moscow Department of Healthcare, 7 Kasatkina Str., 129301 Moscow, Russia; (A.B.); (D.P.)
| | - Oleg Dukhin
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Antonina Elizarova
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 29B Lincoln Dr., Bethesda, MD 20892, USA; (W.F.); (L.M.)
| | - Darya Fomina
- Clinical City Hospital №52, Moscow Department of Healthcare, 3 Pekhotnaya Str., 123182 Moscow, Russia; (D.F.); (M.L.); (E.M.)
| | - Kseniya Glebova
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Oxana Ivanova
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Anna Kalinskaya
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
- Department of Cardiology, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia
| | - Anastasia Lebedeva
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
- Clinical City Hospital №40, Moscow Department of Healthcare, 7 Kasatkina Str., 129301 Moscow, Russia; (A.B.); (D.P.)
| | - Maryana Lysenko
- Clinical City Hospital №52, Moscow Department of Healthcare, 3 Pekhotnaya Str., 123182 Moscow, Russia; (D.F.); (M.L.); (E.M.)
| | - Elena Maryukhnich
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Elena Misyurina
- Clinical City Hospital №52, Moscow Department of Healthcare, 3 Pekhotnaya Str., 123182 Moscow, Russia; (D.F.); (M.L.); (E.M.)
| | - Denis Protsenko
- Clinical City Hospital №40, Moscow Department of Healthcare, 7 Kasatkina Str., 129301 Moscow, Russia; (A.B.); (D.P.)
| | - Alexander Rosin
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Olga Sapozhnikova
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Denis Sokorev
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Alexander Shpektor
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
- Department of Cardiology, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia
| | - Daria Vorobyeva
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (O.I.); (A.K.); (E.M.); (D.V.)
- Clinical City Hospital Named after I.V. Davidovsky, Moscow Department of Healthcare, 11/6 Yauzskaya Str., 109240 Moscow, Russia; (I.M.); (A.A.); (O.D.); (A.E.); (K.G.); (A.L.); (A.R.); (O.S.); (D.S.); (A.S.)
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 29B Lincoln Dr., Bethesda, MD 20892, USA; (W.F.); (L.M.)
| |
Collapse
|
34
|
Ware CF, Croft M, Neil GA. Realigning the LIGHT signaling network to control dysregulated inflammation. J Exp Med 2022; 219:213236. [PMID: 35604387 PMCID: PMC9130030 DOI: 10.1084/jem.20220236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Advances in understanding the physiologic functions of the tumor necrosis factor superfamily (TNFSF) of ligands, receptors, and signaling networks are providing deeper insight into pathogenesis of infectious and autoimmune diseases and cancer. LIGHT (TNFSF14) has emerged as an important modulator of critical innate and adaptive immune responses. LIGHT and its signaling receptors, herpesvirus entry mediator (TNFRSF14), and lymphotoxin β receptor, form an immune regulatory network with two co-receptors of herpesvirus entry mediator, checkpoint inhibitor B and T lymphocyte attenuator, and CD160. Deciphering the fundamental features of this network reveals new understanding to guide therapeutic development. Accumulating evidence from infectious diseases points to the dysregulation of the LIGHT network as a disease-driving mechanism in autoimmune and inflammatory reactions in barrier organs, including coronavirus disease 2019 pneumonia and inflammatory bowel diseases. Recent clinical results warrant further investigation of the LIGHT regulatory network and application of target-modifying therapeutics for disease intervention.
Collapse
Affiliation(s)
- Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA
| | | |
Collapse
|
35
|
Popp M, Reis S, Schießer S, Hausinger RI, Stegemann M, Metzendorf MI, Kranke P, Meybohm P, Skoetz N, Weibel S. Ivermectin for preventing and treating COVID-19. Cochrane Database Syst Rev 2022; 6:CD015017. [PMID: 35726131 PMCID: PMC9215332 DOI: 10.1002/14651858.cd015017.pub3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Ivermectin, an antiparasitic agent, inhibits the replication of viruses in vitro. The molecular hypothesis of ivermectin's antiviral mode of action suggests an inhibitory effect on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in early stages of infection. Currently, evidence on ivermectin for prevention of SARS-CoV-2 infection and COVID-19 treatment is conflicting. OBJECTIVES To assess the efficacy and safety of ivermectin plus standard of care compared to standard of care plus/minus placebo, or any other proven intervention for people with COVID-19 receiving treatment as inpatients or outpatients, and for prevention of an infection with SARS-CoV-2 (postexposure prophylaxis). SEARCH METHODS We searched the Cochrane COVID-19 Study Register, Web of Science (Emerging Citation Index and Science Citation Index), WHO COVID-19 Global literature on coronavirus disease, and HTA database weekly to identify completed and ongoing trials without language restrictions to 16 December 2021. Additionally, we included trials with > 1000 participants up to April 2022. SELECTION CRITERIA We included randomized controlled trials (RCTs) comparing ivermectin to standard of care, placebo, or another proven intervention for treatment of people with confirmed COVID-19 diagnosis, irrespective of disease severity or treatment setting, and for prevention of SARS-CoV-2 infection. Co-interventions had to be the same in both study arms. For this review update, we reappraised eligible trials for research integrity: only RCTs prospectively registered in a trial registry according to WHO guidelines for clinical trial registration were eligible for inclusion. DATA COLLECTION AND ANALYSIS We assessed RCTs for bias, using the Cochrane RoB 2 tool. We used GRADE to rate the certainty of evidence for outcomes in the following settings and populations: 1) to treat inpatients with moderate-to-severe COVID-19, 2) to treat outpatients with mild COVID-19 (outcomes: mortality, clinical worsening or improvement, (serious) adverse events, quality of life, and viral clearance), and 3) to prevent SARS-CoV-2 infection (outcomes: SARS-CoV-2 infection, development of COVID-19 symptoms, admission to hospital, mortality, adverse events and quality of life). MAIN RESULTS We excluded seven of the 14 trials included in the previous review version; six were not prospectively registered and one was non-randomized. This updated review includes 11 trials with 3409 participants investigating ivermectin plus standard of care compared to standard of care plus/minus placebo. No trial investigated ivermectin for prevention of infection or compared ivermectin to an intervention with proven efficacy. Five trials treated participants with moderate COVID-19 (inpatient settings); six treated mild COVID-19 (outpatient settings). Eight trials were double-blind and placebo-controlled, and three were open-label. We assessed around 50% of the trial results as low risk of bias. We identified 31 ongoing trials. In addition, there are 28 potentially eligible trials without publication of results, or with disparities in the reporting of the methods and results, held in 'awaiting classification' until the trial authors clarify questions upon request. Ivermectin for treating COVID-19 in inpatient settings with moderate-to-severe disease We are uncertain whether ivermectin plus standard of care compared to standard of care plus/minus placebo reduces or increases all-cause mortality at 28 days (risk ratio (RR) 0.60, 95% confidence interval (CI) 0.14 to 2.51; 3 trials, 230 participants; very low-certainty evidence); or clinical worsening, assessed by participants with new need for invasive mechanical ventilation or death at day 28 (RR 0.82, 95% CI 0.33 to 2.04; 2 trials, 118 participants; very low-certainty evidence); or serious adverse events during the trial period (RR 1.55, 95% CI 0.07 to 35.89; 2 trials, 197 participants; very low-certainty evidence). Ivermectin plus standard of care compared to standard of care plus placebo may have little or no effect on clinical improvement, assessed by the number of participants discharged alive at day 28 (RR 1.03, 95% CI 0.78 to 1.35; 1 trial, 73 participants; low-certainty evidence); on any adverse events during the trial period (RR 1.04, 95% CI 0.61 to 1.79; 3 trials, 228 participants; low-certainty evidence); and on viral clearance at 7 days (RR 1.12, 95% CI 0.80 to 1.58; 3 trials, 231 participants; low-certainty evidence). No trial investigated quality of life at any time point. Ivermectin for treating COVID-19 in outpatient settings with asymptomatic or mild disease Ivermectin plus standard of care compared to standard of care plus/minus placebo probably has little or no effect on all-cause mortality at day 28 (RR 0.77, 95% CI 0.47 to 1.25; 6 trials, 2860 participants; moderate-certainty evidence) and little or no effect on quality of life, measured with the PROMIS Global-10 scale (physical component mean difference (MD) 0.00, 95% CI -0.98 to 0.98; and mental component MD 0.00, 95% CI -1.08 to 1.08; 1358 participants; high-certainty evidence). Ivermectin may have little or no effect on clinical worsening, assessed by admission to hospital or death within 28 days (RR 1.09, 95% CI 0.20 to 6.02; 2 trials, 590 participants; low-certainty evidence); on clinical improvement, assessed by the number of participants with all initial symptoms resolved up to 14 days (RR 0.90, 95% CI 0.60 to 1.36; 2 trials, 478 participants; low-certainty evidence); on serious adverse events (RR 2.27, 95% CI 0.62 to 8.31; 5 trials, 1502 participants; low-certainty evidence); on any adverse events during the trial period (RR 1.24, 95% CI 0.87 to 1.76; 5 trials, 1502 participants; low-certainty evidence); and on viral clearance at day 7 compared to placebo (RR 1.01, 95% CI 0.69 to 1.48; 2 trials, 331 participants; low-certainty evidence). None of the trials reporting duration of symptoms were eligible for meta-analysis. AUTHORS' CONCLUSIONS For outpatients, there is currently low- to high-certainty evidence that ivermectin has no beneficial effect for people with COVID-19. Based on the very low-certainty evidence for inpatients, we are still uncertain whether ivermectin prevents death or clinical worsening or increases serious adverse events, while there is low-certainty evidence that it has no beneficial effect regarding clinical improvement, viral clearance and adverse events. No evidence is available on ivermectin to prevent SARS-CoV-2 infection. In this update, certainty of evidence increased through higher quality trials including more participants. According to this review's living approach, we will continually update our search.
Collapse
Affiliation(s)
- Maria Popp
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Stefanie Reis
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Selina Schießer
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Renate Ilona Hausinger
- Department of Nephrology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maria-Inti Metzendorf
- Cochrane Metabolic and Endocrine Disorders Group, Institute of General Practice, Medical Faculty of the Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Peter Kranke
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stephanie Weibel
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
36
|
Pagkratis K, Chrysikos S, Antonakis E, Pandi A, Kosti CN, Markatis E, Hillas G, Digalaki A, Koukidou S, Chaini E, Afthinos A, Dimakou K, Papanikolaou IC. Predictors of Mortality in Tocilizumab-Treated Severe COVID-19. Vaccines (Basel) 2022; 10:vaccines10060978. [PMID: 35746585 PMCID: PMC9230711 DOI: 10.3390/vaccines10060978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: Tocilizumab is associated with positive outcomes in severe COVID-19. We wanted to describe the characteristics of nonresponders to treatment. Methods: This was a retrospective multicenter study in two respiratory departments investigating adverse outcomes at 90 days from diagnosis in subjects treated with tocilizumab (8 mg/kg intravenously single dose) for severe progressive COVID-19. Results: Of 121 subjects, 62% were males, and 9% were fully vaccinated. Ninety-six (79.4%) survived, and 25 died (20.6%). Compared to survivors (S), nonsurvivors (NS) were older (median 57 versus 75 years of age), had more comorbidities (Charlson comorbidity index 2 versus 5) and had higher rates of intubation/mechanical ventilation (p < 0.05). On admission, NS had a lower PO2/FiO2 ratio, higher blood ferritin, and higher troponin, and on clinical progression (day of tocilizumab treatment), NS had a lower PO2/FiO2 ratio, decreased lymphocytes, increased neutrophil to lymphocyte ratio, increased ferritin and lactate dehydrogenase (LDH), disease located centrally on computed tomography scan, and increased late c-reactive protein. Cox proportional hazards regression analysis identified age and LDH on deterioration as predictors of death; admission PO2/FiO2 ratio and LDH as predictors of intubation; PO2/FiO2 ratios, LDH, and central lung disease on radiology as predictors of noninvasive ventilation (NIV) (a < 0.05). The log-rank test of mortality yielded the same results (p < 0.001). ROC analysis of the above predictors in a separate validation cohort yielded significant results. Conclusions: Older age and high serum LDH levels are predictors of mortality in tocilizumab-treated severe COVID-19 patients. Hypoxia levels, LDH, and central pulmonary involvement radiologically are associated with intubation and NIV.
Collapse
Affiliation(s)
- Konstantinos Pagkratis
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Serafeim Chrysikos
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Emmanouil Antonakis
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Aggeliki Pandi
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Chrysavgi Nikolaou Kosti
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Eleftherios Markatis
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Georgios Hillas
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Antonia Digalaki
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Sofia Koukidou
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Eleftheria Chaini
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Andreas Afthinos
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
| | - Katerina Dimakou
- 5th Respiratory Medicine Department, SOTIRIA Chest Hospital, 11527 Athens, Greece; (S.C.); (C.N.K.); (G.H.); (A.D.); (S.K.); (K.D.)
| | - Ilias C. Papanikolaou
- Pulmonary Department, Corfu General Hospital, 49100 Corfu, Greece; (K.P.); (E.A.); (A.P.); (E.M.); (E.C.); (A.A.)
- Correspondence: ; Tel.: +30-266-136-0694; Fax.: +30-266-136-0488
| |
Collapse
|
37
|
Kirillova A, Lado A, Blatt N. Application of Monoclonal Antibody Drugs in Treatment of COVID-19: a Review. BIONANOSCIENCE 2022; 12:1436-1454. [PMID: 35729973 PMCID: PMC9198616 DOI: 10.1007/s12668-022-00997-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 01/08/2023]
Abstract
Coronavirus infection can have various degrees of severity and outcomes. In some cases, it causes excessive production of pro-inflammatory cytokines, a so-called cytokine storm, leading to acute respiratory distress syndrome. Unfortunately, the exact pathophysiology and treatment, especially for severe cases of COVID-19, are still uncertain. Results of preliminary studies showed that immunosuppressive therapy, such as interleukin (IL)-6, IL-1, and TNF-α antagonists commonly used in rheumatology, can be considered as treatment options for COVID-19, especially in severe cases. The review focused on the most common and currently studied monoclonal antibody drugs, as well as up-to-date data on the pathogenesis of COVID-19, host immune response against SARS-CoV-2 and its association with cytokine storm. It also covered effects of interleukin (IL)-6, IL-1, and TNF-α blockers on the course of coronavirus infection and outcome in patients treated for the main autoimmune disease and subsequently infected with COVID-19.
Collapse
Affiliation(s)
- Aleksandra Kirillova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Anna Lado
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Nataliya Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
38
|
Kramer A, Prinz C, Fichtner F, Fischer AL, Thieme V, Grundeis F, Spagl M, Seeber C, Piechotta V, Metzendorf MI, Golinski M, Moerer O, Stephani C, Mikolajewska A, Kluge S, Stegemann M, Laudi S, Skoetz N. Janus kinase inhibitors for the treatment of COVID-19. Cochrane Database Syst Rev 2022; 6:CD015209. [PMID: 35695334 PMCID: PMC9190191 DOI: 10.1002/14651858.cd015209] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND With potential antiviral and anti-inflammatory properties, Janus kinase (JAK) inhibitors represent a potential treatment for symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. They may modulate the exuberant immune response to SARS-CoV-2 infection. Furthermore, a direct antiviral effect has been described. An understanding of the current evidence regarding the efficacy and safety of JAK inhibitors as a treatment for coronavirus disease 2019 (COVID-19) is required. OBJECTIVES To assess the effects of systemic JAK inhibitors plus standard of care compared to standard of care alone (plus/minus placebo) on clinical outcomes in individuals (outpatient or in-hospital) with any severity of COVID-19, and to maintain the currency of the evidence using a living systematic review approach. SEARCH METHODS We searched the Cochrane COVID-19 Study Register (comprising MEDLINE, Embase, ClinicalTrials.gov, World Health Organization (WHO) International Clinical Trials Registry Platform, medRxiv, and Cochrane Central Register of Controlled Trials), Web of Science, WHO COVID-19 Global literature on coronavirus disease, and the US Department of Veterans Affairs Evidence Synthesis Program (VA ESP) Covid-19 Evidence Reviews to identify studies up to February 2022. We monitor newly published randomised controlled trials (RCTs) weekly using the Cochrane COVID-19 Study Register, and have incorporated all new trials from this source until the first week of April 2022. SELECTION CRITERIA We included RCTs that compared systemic JAK inhibitors plus standard of care to standard of care alone (plus/minus placebo) for the treatment of individuals with COVID-19. We used the WHO definitions of illness severity for COVID-19. DATA COLLECTION AND ANALYSIS We assessed risk of bias of primary outcomes using Cochrane's Risk of Bias 2 (RoB 2) tool. We used GRADE to rate the certainty of evidence for the following primary outcomes: all-cause mortality (up to day 28), all-cause mortality (up to day 60), improvement in clinical status: alive and without need for in-hospital medical care (up to day 28), worsening of clinical status: new need for invasive mechanical ventilation or death (up to day 28), adverse events (any grade), serious adverse events, secondary infections. MAIN RESULTS We included six RCTs with 11,145 participants investigating systemic JAK inhibitors plus standard of care compared to standard of care alone (plus/minus placebo). Standard of care followed local protocols and included the application of glucocorticoids (five studies reported their use in a range of 70% to 95% of their participants; one study restricted glucocorticoid use to non-COVID-19 specific indications), antibiotic agents, anticoagulants, and antiviral agents, as well as non-pharmaceutical procedures. At study entry, about 65% of participants required low-flow oxygen, about 23% required high-flow oxygen or non-invasive ventilation, about 8% did not need any respiratory support, and only about 4% were intubated. We also identified 13 ongoing studies, and 9 studies that are completed or terminated and where classification is pending. Individuals with moderate to severe disease Four studies investigated the single agent baricitinib (10,815 participants), one tofacitinib (289 participants), and one ruxolitinib (41 participants). Systemic JAK inhibitors probably decrease all-cause mortality at up to day 28 (95 of 1000 participants in the intervention group versus 131 of 1000 participants in the control group; risk ratio (RR) 0.72, 95% confidence interval (CI) 0.57 to 0.91; 6 studies, 11,145 participants; moderate-certainty evidence), and decrease all-cause mortality at up to day 60 (125 of 1000 participants in the intervention group versus 181 of 1000 participants in the control group; RR 0.69, 95% CI 0.56 to 0.86; 2 studies, 1626 participants; high-certainty evidence). Systemic JAK inhibitors probably make little or no difference in improvement in clinical status (discharged alive or hospitalised, but no longer requiring ongoing medical care) (801 of 1000 participants in the intervention group versus 778 of 1000 participants in the control group; RR 1.03, 95% CI 1.00 to 1.06; 4 studies, 10,802 participants; moderate-certainty evidence). They probably decrease the risk of worsening of clinical status (new need for invasive mechanical ventilation or death at day 28) (154 of 1000 participants in the intervention group versus 172 of 1000 participants in the control group; RR 0.90, 95% CI 0.82 to 0.98; 2 studies, 9417 participants; moderate-certainty evidence). Systemic JAK inhibitors probably make little or no difference in the rate of adverse events (any grade) (427 of 1000 participants in the intervention group versus 441 of 1000 participants in the control group; RR 0.97, 95% CI 0.88 to 1.08; 3 studies, 1885 participants; moderate-certainty evidence), and probably decrease the occurrence of serious adverse events (160 of 1000 participants in the intervention group versus 202 of 1000 participants in the control group; RR 0.79, 95% CI 0.68 to 0.92; 4 studies, 2901 participants; moderate-certainty evidence). JAK inhibitors may make little or no difference to the rate of secondary infection (111 of 1000 participants in the intervention group versus 113 of 1000 participants in the control group; RR 0.98, 95% CI 0.89 to 1.09; 4 studies, 10,041 participants; low-certainty evidence). Subgroup analysis by severity of COVID-19 disease or type of JAK inhibitor did not identify specific subgroups which benefit more or less from systemic JAK inhibitors. Individuals with asymptomatic or mild disease We did not identify any trial for this population. AUTHORS' CONCLUSIONS In hospitalised individuals with moderate to severe COVID-19, moderate-certainty evidence shows that systemic JAK inhibitors probably decrease all-cause mortality. Baricitinib was the most often evaluated JAK inhibitor. Moderate-certainty evidence suggests that they probably make little or no difference in improvement in clinical status. Moderate-certainty evidence indicates that systemic JAK inhibitors probably decrease the risk of worsening of clinical status and make little or no difference in the rate of adverse events of any grade, whilst they probably decrease the occurrence of serious adverse events. Based on low-certainty evidence, JAK inhibitors may make little or no difference in the rate of secondary infection. Subgroup analysis by severity of COVID-19 or type of agent failed to identify specific subgroups which benefit more or less from systemic JAK inhibitors. Currently, there is no evidence on the efficacy and safety of systemic JAK inhibitors for individuals with asymptomatic or mild disease (non-hospitalised individuals).
Collapse
Affiliation(s)
- Andre Kramer
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Carolin Prinz
- Department of Anesthesiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Falk Fichtner
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Anna-Lena Fischer
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Volker Thieme
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Felicitas Grundeis
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Manuel Spagl
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Christian Seeber
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maria-Inti Metzendorf
- Institute of General Practice, Medical Faculty of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Golinski
- Department of Anesthesiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Onnen Moerer
- Department of Anesthesiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Caspar Stephani
- Department of Anesthesiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Agata Mikolajewska
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, Medical Center Hamburg Eppendorf (UKE), Hamburg, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sven Laudi
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
39
|
Lu S, Huang X, Liu R, Lan Y, Lei Y, Zeng F, Tang X, He H. Comparison of COVID-19 Induced Respiratory Failure and Typical ARDS: Similarities and Differences. Front Med (Lausanne) 2022; 9:829771. [PMID: 35712114 PMCID: PMC9196311 DOI: 10.3389/fmed.2022.829771] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/09/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a predominantly respiratory infectious disease caused by novel coronavirus infection (SARS-CoV-2), respiratory failure is the main clinical manifestation and the leading cause of death. Even though it can meet the acute respiratory distress syndrome (ARDS) Berlin definition, only some clinical features of COVID-19 are consistent with typical ARDS, and which has its own peculiar phenotypes. When compared with typical ARDS, in addition to the typical diffuse alveolar injury, COVID-19 has unique pathological and pathophysiological features, such as endothelial injury, extensive microthrombus, and pulmonary capillary hyperplasia. The clinical features of patients with respiratory failure caused by COVID-19 are heterogeneous and can be generally divided into two phenotypes: progressive respiratory distress and unique "silent hypoxemia". The "H-type" characteristics of reduced lung volume, decreased lung compliance, and unmatched ventilator-perfusion ratio. While some patients may have close to normal lung compliance, that is "L-type". Identifying the exact phenotype in whom are suffered with COVID-19 is crucial to guide clinicians to adopt appropriate treatment strategies. This review discussed the similarities and differences in the pathogenesis, pathophysiology, clinical features and treatment strategies of COVID-19 induced acute respiratory failure and typical ARDS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hongli He
- Department of Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Chengdu, China
| |
Collapse
|
40
|
Rosas IO, Bräu N, Waters M, Go RC, Malhotra A, Hunter BD, Bhagani S, Skiest D, Savic S, Douglas IS, Garcia-Diaz J, Aziz MS, Cooper N, Youngstein T, Sorbo LD, Zerda DJDL, Ustianowski A, Gracian AC, Blyth KG, Carratalà J, François B, Benfield T, Haslem D, Bonfanti P, van der Leest CH, Rohatgi N, Wiese L, Luyt CE, Bauer RN, Cai F, Lee IT, Matharu B, Metcalf L, Wildum S, Graham E, Tsai L, Bao M. Tocilizumab in patients hospitalised with COVID-19 pneumonia: Efficacy, safety, viral clearance, and antibody response from a randomised controlled trial (COVACTA). EClinicalMedicine 2022; 47:101409. [PMID: 35475258 PMCID: PMC9022847 DOI: 10.1016/j.eclinm.2022.101409] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In COVACTA, a randomised, placebo-controlled trial in patients hospitalised with coronavirus disease-19 (COVID-19), tocilizumab did not improve 28-day mortality, but shortened hospital and intensive care unit stay. Longer-term effects of tocilizumab in patients with COVID-19 are unknown. Therefore, the efficacy and safety of tocilizumab in COVID-19 beyond day 28 and its impact on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) clearance and antibody response in COVACTA were investigated. METHODS Adults in Europe and North America hospitalised with COVID-19 (N = 452) between April 3, 2020 and May 28, 2020 were randomly assigned (2:1) to double-blind intravenous tocilizumab or placebo and assessed for efficacy and safety through day 60. Assessments included mortality, time to hospital discharge, SARS-CoV-2 viral load in nasopharyngeal swab and serum samples, and neutralising anti-SARS-CoV-2 antibodies in serum. ClinicalTrials.gov registration: NCT04320615. FINDINGS By day 60, 24·5% (72/294) of patients in the tocilizumab arm and 25·0% (36/144) in the placebo arm died (weighted difference -0·5% [95% CI -9·1 to 8·0]), and 67·0% (197/294) in the tocilizumab arm and 63·9% (92/144) in the placebo arm were discharged from the hospital. Serious infections occurred in 24·1% (71/295) of patients in the tocilizumab arm and 29·4% (42/143) in the placebo arm. Median time to negative reverse transcriptase-quantitative polymerase chain reaction result in nasopharyngeal/oropharyngeal samples was 15·0 days (95% CI 14·0 to 21·0) in the tocilizumab arm and 21·0 days (95% CI 14·0 to 28·0) in the placebo arm. All tested patients had positive test results for neutralising anti-SARS-CoV-2 antibodies at day 60. INTERPRETATION There was no mortality benefit with tocilizumab through day 60. Tocilizumab did not impair viral clearance or host immune response, and no new safety signals were observed. Future investigations may explore potential biomarkers to optimize patient selection for tocilizumab treatment and combination therapy with other treatments. FUNDING F. Hoffmann-La Roche Ltd and the US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, under OT number HHSO100201800036C.
Collapse
Affiliation(s)
- Ivan O. Rosas
- Pulmonary, Critical Care, and Sleep Medicine, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA
- Corresponding author.
| | - Norbert Bräu
- James J. Peters Veterans Affairs Medical Center, Bronx, and Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ronaldo C. Go
- Hackensack Meridian School of Medicine and Hackensack University Medical Center, Hackensack, NJ, USA
| | - Atul Malhotra
- University of California San Diego, La Jolla, CA, USA
| | | | | | - Daniel Skiest
- University of Massachusetts Medical School–Baystate, Springfield, MA, USA
| | - Sinisa Savic
- Leeds Teaching Hospitals NHS Trust and National Institute for Health Research–Leeds, Biomedical Research Centre, Leeds, UK
| | - Ivor S. Douglas
- Denver Health Medical Center, Denver, and University of Colorado, Anschutz School of Medicine, Aurora, CO, USA
| | | | | | | | | | | | | | | | - Antonio Cubillo Gracian
- Hospital Universitario HM Sanchinarro, Centro Integral, Oncológico Clara Campal, and Departamento de Ciencias Médicas Clínicas, Facultad de Medicina, Universidad CEU San Pablo, Madrid, Spain
| | - Kevin G. Blyth
- Institute of Cancer Sciences, University of Glasgow/Department of Respiratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Jordi Carratalà
- Department of Infectious Diseases, Bellvitge University Hospital, Bellvitge Biomedical Research Institute, University of Barcelona, and CIBERINFEC, Barcelona, Spain
| | - Bruno François
- Intensive Care Unit and Inserm CIC1435 and UMR1092, CHU Limoges, Limoges, France
| | - Thomas Benfield
- Center of Research and Disruption of Infectious Diseases, Department of Infectious Diseases, Copenhagen University Hospital–Amager and Hvidovre, Hvidovre, Denmark
| | | | - Paolo Bonfanti
- University of Milano–Bicocca and Azienda Ospedaliera San Gerardo di Monza, Monza, Italy
| | | | - Nidhi Rohatgi
- Division of Hospital Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lothar Wiese
- Department of Infectious Diseases, Zealand University Hospital, Roskilde, Denmark
| | | | | | - Fang Cai
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | - Min Bao
- Genentech, South San Francisco, CA, USA
| |
Collapse
|
41
|
Durstenfeld MS, Peluso MJ, Kelly JD, Win S, Swaminathan S, Li D, Arechiga VM, Zepeda VA, Sun K, Shao SJ, Hill C, Arreguin MI, Lu S, Hoh R, Tai VW, Chenna A, Yee BC, Winslow JW, Petropoulos CJ, Kornak J, Henrich TJ, Martin JN, Deeks SG, Hsue PY. Role of antibodies, inflammatory markers, and echocardiographic findings in post-acute cardiopulmonary symptoms after SARS-CoV-2 infection. JCI Insight 2022; 7:157053. [PMID: 35389890 PMCID: PMC9220849 DOI: 10.1172/jci.insight.157053] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
Shortness of breath, chest pain, and palpitations occur as postacute sequelae of COVID-19, but whether symptoms are associated with echocardiographic abnormalities, cardiac biomarkers, or markers of systemic inflammation remains unknown. In a cross-sectional analysis, we assessed symptoms, performed echocardiograms, and measured biomarkers among adults more than 8 weeks after confirmed SARS-CoV-2 infection. We modeled associations between symptoms and baseline characteristics, echocardiographic findings, and biomarkers using logistic regression. We enrolled 102 participants at a median of 7.2 months following COVID-19 onset; 47 individuals reported dyspnea, chest pain, or palpitations. Median age was 52 years, and 41% of participants were women. Female sex, hospitalization, IgG antibody against SARS-CoV-2 receptor binding domain, and C-reactive protein were associated with symptoms. Regarding echocardiographic findings, 4 of 47 participants (9%) with symptoms had pericardial effusions compared with 0 of 55 participants without symptoms; those with effusions had a median of 4 symptoms compared with a median of 1 symptom in those without effusions. There was no strong evidence for a relationship between symptoms and echocardiographic functional parameters or other biomarkers. Among adults more than 8 weeks after SARS-CoV-2 infection, SARS-CoV-2 RBD antibodies, markers of inflammation, and, possibly, pericardial effusions are associated with cardiopulmonary symptoms. Investigation into inflammation as a mechanism underlying postacute sequelae of COVID-19 is warranted.
Collapse
Affiliation(s)
- Matthew S Durstenfeld
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Michael J Peluso
- University of California, San Francisco, San Francisco, United States of America
| | - J Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, United States of America
| | - Sithu Win
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Shreya Swaminathan
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Danny Li
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Victor M Arechiga
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Victor Antonio Zepeda
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| | - Kaiwen Sun
- Department of Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Shirley J Shao
- School of Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Christopher Hill
- School of Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Mireya I Arreguin
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Scott Lu
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Viva W Tai
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Ahmed Chenna
- Oncology Group, Monogram Biosciences, South San Francisco, United States of America
| | - Brandon C Yee
- Monogram Biosciences, South San Francisco, United States of America
| | - John W Winslow
- Oncology Group, Monogram Biosciences, South San Francisco, United States of America
| | | | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, United States of America
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, United States of America
| | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Priscilla Y Hsue
- Division of Cardiology, University of California, San Francisco, San Francisco, United States of America
| |
Collapse
|
42
|
Gonzalez FA, Ângelo-Dias M, Martins C, Gomes R, Bacariza J, Fernandes A, Borrego LM, Group E. Characteristic Immune Dynamics in COVID-19 Patients with Cardiac Dysfunction. J Clin Med 2022; 11:jcm11071880. [PMID: 35407485 PMCID: PMC8999785 DOI: 10.3390/jcm11071880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/13/2022] [Accepted: 03/26/2022] [Indexed: 12/19/2022] Open
Abstract
Background: We aimed to explore immune parameters in COVID-19 patients admitted to the intensive care unit (ICU) to identify distinctive features in patients with cardiac injury. Methods: A total of 30 COVID-19 patients >18 years admitted to the ICU were studied on days D1, D3 and D7 after admission. Cardiac function was assessed using speckle-tracking echocardiography (STE). Peripheral blood immunophenotyping, cardiac (pro-BNP; troponin) and inflammatory biomarkers were simultaneously evaluated. Results: Cardiac dysfunction (DYS) was detected by STE in 73% of patients: 40% left ventricle (LV) systolic dysfunction, 60% LV diastolic dysfunction, 37% right ventricle systolic dysfunction. High-sensitivity cardiac troponin (hs-cTn) was detectable in 43.3% of the patients with a median value of 13.00 ng/L. There were no significant differences between DYS and nDYS patients regarding mortality, organ dysfunction, cardiac (including hs-cTn) or inflammatory biomarkers. Patients with DYS showed persistently lower lymphocyte counts (median 896 [661−1837] cells/µL vs. 2141 [924−3306] cells/µL, p = 0.058), activated CD3 (median 85 [66−170] cells/µL vs. 186 [142−259] cells/µL, p = 0.047) and CD4 T cells (median 33 [28−40] cells/µL vs. 63 [48−79] cells/µL, p = 0.005), and higher effector memory T cells (TEM) at baseline (CD4%: 10.9 [6.4−19.2] vs. 5.9 [4.2−12.8], p = 0.025; CD8%: 15.7 [7.9−22.8] vs. 8.1 [7.7−13.7], p = 0.035; CD8 counts: 40 cells/µL [17−61] vs. 10 cells/µL [7−17], p = 0.011) than patients without cardiac dysfunction. Conclusion: Our study suggests an association between the immunological trait and cardiac dysfunction in severe COVID-19 patients.
Collapse
Affiliation(s)
- Filipe André Gonzalez
- Intensive Care Department, Hospital Garcia de Orta, 2805-267 Almada, Portugal; (R.G.); (J.B.); (A.F.)
- Correspondence: ; Tel.: +351-91-793-2502
| | - Miguel Ângelo-Dias
- CEDOC—Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, 1099-085 Lisbon, Portugal; (M.Â.-D.); (C.M.); (L.M.B.)
| | - Catarina Martins
- CEDOC—Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, 1099-085 Lisbon, Portugal; (M.Â.-D.); (C.M.); (L.M.B.)
| | - Rui Gomes
- Intensive Care Department, Hospital Garcia de Orta, 2805-267 Almada, Portugal; (R.G.); (J.B.); (A.F.)
| | - Jacobo Bacariza
- Intensive Care Department, Hospital Garcia de Orta, 2805-267 Almada, Portugal; (R.G.); (J.B.); (A.F.)
| | - Antero Fernandes
- Intensive Care Department, Hospital Garcia de Orta, 2805-267 Almada, Portugal; (R.G.); (J.B.); (A.F.)
| | - Luís Miguel Borrego
- CEDOC—Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, 1099-085 Lisbon, Portugal; (M.Â.-D.); (C.M.); (L.M.B.)
- Immunoallergy Department, Hospital da Luz Lisboa, 1500-650 Lisbon, Portugal
| | - EchoCrit Group
- Intensive Care Department, Hospital Garcia de Orta, 2805-267 Almada, Portugal; (R.G.); (J.B.); (A.F.)
| |
Collapse
|
43
|
Al-Hajeri H, Baroun F, Abutiban F, Al-Mutairi M, Ali Y, Alawadhi A, Albasri A, Aldei A, AlEnizi A, Alhadhood N, Al-Herz A, Alkadi A, Alkanderi W, Almathkoori A, Almutairi N, Alsayegh S, Alturki A, Bahbahani H, Dehrab A, Ghanem A, Haji Hasan E, Hayat S, Saleh K, Tarakmeh H. Therapeutic role of immunomodulators during the COVID-19 pandemic- a narrative review. Postgrad Med 2022; 134:160-179. [PMID: 35086413 PMCID: PMC8862162 DOI: 10.1080/00325481.2022.2033563] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/18/2022] [Indexed: 02/07/2023]
Abstract
The emergency state caused by COVID-19 saw the use of immunomodulators despite the absence of robust research. To date, the results of relatively few randomized controlled trials have been published, and methodological approaches are riddled with bias and heterogeneity. Anti-SARS-CoV-2 antibodies, convalescent plasma and the JAK inhibitor baricitinib have gained Emergency Use Authorizations and tentative recommendations for their use in clinical practice alone or in combination with other therapies. Anti-SARS-CoV-2 antibodies are predominating the management of non-hospitalized patients, while the inpatient setting is seeing the use of convalescent plasma, baricitinib, tofacitinib, tocilizumab, sarilumab, and corticosteroids, as applicable. Available clinical data also suggest the potential clinical benefit of the early administration of blood-derived products (e.g. convalescent plasma, non-SARS-CoV-2-specific immunoglobins) and the blockade of factors implicated in the hyperinflammatory state of severe COVID-19 (Interleukin 1 and 6; Janus Kinase). Immune therapies seem to have a protective effect and using immunomodulators alone or in combination with viral replication inhibitors and other treatment modalities might prevent progression into severe COVID-19 disease, cytokine storm and death. Future trials should address existing gaps and reshape the landscape of COVID-19 management.
Collapse
Affiliation(s)
- Hebah Al-Hajeri
- Department of Rheumatology and Internal Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Fatemah Baroun
- Department of Rheumatology and Internal Medicine, AlJahra Hospital, Al-Jahra, Kuwait
| | - Fatemah Abutiban
- Department of Rheumatology and Internal Medicine, Jaber Al-Ahmad Hospital, South Surra, Kuwait
| | | | - Yasser Ali
- Rheumatology Unit, Department of Internal Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Adel Alawadhi
- Rheumatology Unit, Department of Internal Medicine, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Anwar Albasri
- Rheumatology Unit, Department of Internal Medicine, Jaber Al-Ahmad Hospital, South Surra, Kuwait
| | - Ali Aldei
- Rheumatology Unit, Department of Internal Medicine, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Ahmad AlEnizi
- Rheumatology Unit, Department of Internal Medicine, AlJahra Hospital, AlJahra, Kuwait
| | - Naser Alhadhood
- Rheumatology Unit, Department of Internal Medicine, Farwaneyah Hospital, AlFarwaniya, Kuwait
| | - Adeeba Al-Herz
- Rheumatology Unit, Department of Internal Medicine, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Amjad Alkadi
- Rheumatology Unit, Department of Internal Medicine, Al-Sabah Hospital, Alsabah, Kuwait
| | - Waleed Alkanderi
- Rheumatology Unit, Department of Internal Medicine, Farwaneyah Hospital, AlFarwaniya, Kuwait
| | - Ammar Almathkoori
- Rheumatology Unit, Department of Internal Medicine, Al-Adan Hospital, Hadiya, Kuwait
| | - Nora Almutairi
- Rheumatology Unit, Department of Internal Medicine, Al-Sabah Hospital, Alsabah, Kuwait
| | - Saud Alsayegh
- Rheumatology Unit, Department of Internal Medicine, Jaber Al-Ahmad Armed Forces, Kuwait City, Kuwait
| | - Ali Alturki
- Rheumatology Unit, Department of Internal Medicine, Al-Adan Hospital, Hadiya, Kuwait
| | - Husain Bahbahani
- Rheumatology Unit, Department of Internal Medicine, Farwaneyah Hospital, AlFarwaniya, Kuwait
| | - Ahmad Dehrab
- Rheumatology Unit, Department of Internal Medicine, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Aqeel Ghanem
- Rheumatology Unit, Department of Internal Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Eman Haji Hasan
- Rheumatology Unit, Department of Internal Medicine, Al-Amiri Hospital, Kuwait City, Kuwait
| | - Sawsan Hayat
- Rheumatology Unit, Department of Internal Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Khuloud Saleh
- Rheumatology Unit, Department of Internal Medicine, Farwaneyah Hospital, AlFarwaniya, Kuwait
| | - Hoda Tarakmeh
- Rheumatology Unit, Department of Internal Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| |
Collapse
|
44
|
Zizzo G, Tamburello A, Castelnovo L, Laria A, Mumoli N, Faggioli PM, Stefani I, Mazzone A. Immunotherapy of COVID-19: Inside and Beyond IL-6 Signalling. Front Immunol 2022; 13:795315. [PMID: 35340805 PMCID: PMC8948465 DOI: 10.3389/fimmu.2022.795315] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/25/2022] [Indexed: 01/08/2023] Open
Abstract
Acting on the cytokine cascade is key to preventing disease progression and death in hospitalised patients with COVID-19. Among anti-cytokine therapies, interleukin (IL)-6 inhibitors have been the most used and studied since the beginning of the pandemic. Going through previous observational studies, subsequent randomised controlled trials, and meta-analyses, we focused on the baseline characteristics of the patients recruited, identifying the most favourable features in the light of positive or negative study outcomes; taking into account the biological significance and predictivity of IL-6 and other biomarkers according to specific thresholds, we ultimately attempted to delineate precise windows for therapeutic intervention. By stimulating scavenger macrophages and T-cell responsivity, IL-6 seems protective against viral replication during asymptomatic infection; still protective on early tissue damage by modulating the release of granzymes and lymphokines in mild-moderate disease; importantly pathogenic in severe disease by inducing the proinflammatory activation of immune and endothelial cells (through trans-signalling and trans-presentation); and again protective in critical disease by exerting homeostatic roles for tissue repair (through cis-signalling), while IL-1 still drives hyperinflammation. IL-6 inhibitors, particularly anti-IL-6R monoclonal antibodies (e.g., tocilizumab, sarilumab), are effective in severe disease, characterised by baseline IL-6 concentrations ranging from 35 to 90 ng/mL (reached in the circulation within 6 days of hospital admission), a ratio of partial pressure arterial oxygen (PaO2) and fraction of inspired oxygen (FiO2) between 100 and 200 mmHg, requirement of high-flow oxygen or non-invasive ventilation, C-reactive protein levels between 120 and 160 mg/L, ferritin levels between 800 and 1600 ng/mL, D-dimer levels between 750 and 3000 ng/mL, and lactate dehydrogenase levels between 350 and 500 U/L. Granulocyte-macrophage colony-stimulating factor inhibitors might have similar windows of opportunity but different age preferences compared to IL-6 inhibitors (over or under 70 years old, respectively). Janus kinase inhibitors (e.g., baricitinib) may also be effective in moderate disease, whereas IL-1 inhibitors (e.g., anakinra) may also be effective in critical disease. Correct use of biologics based on therapeutic windows is essential for successful outcomes and could inform future new trials with more appropriate recruiting criteria.
Collapse
Affiliation(s)
- Gaetano Zizzo
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale (ASST) Ovest Milanese, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abidi E, El Nekidy WS, Alefishat E, Rahman N, Petroianu GA, El-Lababidi R, Mallat J. Tocilizumab and COVID-19: Timing of Administration and Efficacy. Front Pharmacol 2022; 13:825749. [PMID: 35250575 PMCID: PMC8894855 DOI: 10.3389/fphar.2022.825749] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Elevated concentrations of interleukin-6 have been demonstrated to be an important key factor in COVID-19 host immune impairment. It represents an important prognostic factor of harm associated with COVID-19 infection by stimulating a vigorous proinflammatory response, leading to the so-called "cytokine storm". Therefore, immunomodulatory interventions targeting interleukin-6 receptor antagonism have been investigated as potential treatments to counterbalance the host immune dysregulation and to support the advantageous effects of corticosteroids. Tocilizumab is a recombinant humanized monoclonal antibody that has gained much interest during the COVID-19 pandemic as an interleukin-6 receptor antagonist. Various early observational studies have reported beneficial effects of tocilizumab. Moreover, consequent randomized controlled trials have subsequently shown significant positive results about tocilizumab efficacy and safety, focusing on outcomes like mortality, risk of intensive care unit admission, and the need for mechanical ventilation, while others presented conflicting findings. In this review, we first described the pathophysiology of COVID-19 infection while highlighting the role of interleukin-6. Furthermore, we also discussed the non-conclusive evidence about tocilizumab to be used as the standard of care therapy for all patients with COVID-19 pneumonia, as well as its beneficial effects in selected patients.
Collapse
Affiliation(s)
- Emna Abidi
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Wasim S. El Nekidy
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nadeem Rahman
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
- Critical Care Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Georg A. Petroianu
- Department of Pharmacology, College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Rania El-Lababidi
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Jihad Mallat
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
- Critical Care Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
- Normandy University, UNICAEN, Caen, France
| |
Collapse
|
46
|
Imeneo A, Alessio G, Di Lorenzo A, Campogiani L, Lodi A, Barreca F, Zordan M, Barchi V, Massa B, Tedde S, Crea A, Vitale P, Spalliera I, Compagno M, Coppola L, Dori L, Malagnino V, Teti E, Andreoni M, Sarmati L, Iannetta M. In Patients with Severe COVID-19, the Profound Decrease in the Peripheral Blood T-Cell Subsets Is Correlated with an Increase of QuantiFERON-TB Gold Plus Indeterminate Rates and Reflecting a Reduced Interferon-Gamma Production. Life (Basel) 2022; 12:life12020244. [PMID: 35207531 PMCID: PMC8880410 DOI: 10.3390/life12020244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/15/2022] Open
Abstract
Increased rates of indeterminate QuantiFERON-TB Gold Plus Assay (QFT-Plus) were demonstrated in patients hospitalized with Coronavirus Disease (COVID)-19. We aimed to define the prevalence and characteristics of hospitalized COVID-19 patients with indeterminate QFT-Plus. A retrospective study was performed including hospitalized COVID-19 patients, stratified in survivors and non-survivors, non-severe and severe according to the maximal oxygen supply required. Statistical analysis was performed using JASP ver0.14.1 and GraphPad Prism ver8.2.1. A total of 420 patients were included, median age: 65 years, males: 66.4%. The QFT-Plus was indeterminate in 22.1% of patients. Increased rate of indeterminate QFT-Plus was found in non-survivors (p = 0.013) and in severe COVID-19 patients (p < 0.001). Considering the Mitogen-Nil condition of the QFT-Plus, an impaired production of interferon-gamma (IFN-γ) was found in non-survivors (p < 0.001) and in severe COVID-19 patients (p < 0.001). A positive correlation between IFN-γ levels in the Mitogen-Nil condition and the absolute counts of CD3+ (p < 0.001), CD4+ (p < 0.001), and CD8+ (p < 0.001) T-lymphocytes was found. At the multivariable analysis, CD3+ T-cell absolute counts and CD4/CD8 ratio were confirmed as independent predictors of indeterminate results at the QFT-Plus. Our study confirmed the increased rate of indeterminate QFT-Plus in COVID-19 patients, mainly depending on the peripheral blood T-lymphocyte depletion found in the most severe cases.
Collapse
Affiliation(s)
- Alessandra Imeneo
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Grazia Alessio
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Andrea Di Lorenzo
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Laura Campogiani
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Alessandra Lodi
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Filippo Barreca
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Marta Zordan
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Virginia Barchi
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Barbara Massa
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Simona Tedde
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Angela Crea
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
| | - Pietro Vitale
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Ilaria Spalliera
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Mirko Compagno
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Luigi Coppola
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Luca Dori
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Vincenzo Malagnino
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Elisabetta Teti
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Massimo Andreoni
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Loredana Sarmati
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
| | - Marco Iannetta
- Department of System Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (A.I.); (G.A.); (A.D.L.); (A.L.); (F.B.); (M.Z.); (V.B.); (B.M.); (S.T.); (A.C.); (V.M.); (M.A.); (L.S.)
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy; (L.C.); (P.V.); (I.S.); (M.C.); (L.C.); (L.D.); (E.T.)
- Correspondence:
| |
Collapse
|
47
|
Kuwahara M, Kamigaito M, Nitta S, Hasegawa K, Murakami H, Kobayashi T, Shirai K, Kohama K, Hirata JI. Effect of Tocilizumab Treatment on Patients with Coronavirus Disease 2019 and Bacteremia: A Retrospective Cohort Study. Infect Dis Ther 2022; 11:533-541. [PMID: 35060111 PMCID: PMC8775152 DOI: 10.1007/s40121-022-00592-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION This study aimed to determine if tocilizumab treatment for coronavirus disease 2019 (COVID-19) increases bacteremia and suppresses fever and inflammatory reactants. METHODS In this single-center, retrospective, observational study, all patients with COVID-19 admitted to our emergency intensive care unit from March 2020 to August 2021 were categorized into tocilizumab-treated and tocilizumab-naïve groups, and the incidence of bacteremia and other factors between the two groups were compared. Patients with bacteremia were further classified into tocilizumab-treated and tocilizumab-naïve groups to determine if fever and inflammatory reactants were suppressed. RESULTS Overall, 144 patients were included in the study, 51 of whom received tocilizumab, which was administered on the day of admission. Further, of the 24 (16.7%) patients with bacteremia, 13 were in the tocilizumab-treated group. Results revealed a significant difference in the C-reactive protein level (p < 0.001) at the onset of bacteremia between the tocilizumab-treated group [median 0.42 mg/dL (0.27-0.44 mg/dL)] and the tocilizumab-naïve group [7.48 mg/dL (4.56-13.9 mg/dL)]. The median number of days from admission to onset of bacteremia was not significantly different between the tocilizumab-treated group [10 days (9-12 days)] and the tocilizumab-naïve group [9 days (7.5-11 days)] (p = 0.48). There was no significant difference in fever between the groups. Multivariate logistic analysis showed that tocilizumab treatment did not affect the probability of bacteremia. CONCLUSION Treatment of patients with COVID-19 with tocilizumab does not increase the risk of bacteremia. Tocilizumab suppresses C-reactive protein levels but not fever. Therefore, careful monitoring of fever can reduce the risk of missed bacteremia.
Collapse
Affiliation(s)
- Masaatsu Kuwahara
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Misa Kamigaito
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Shou Nitta
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kana Hasegawa
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hiromoto Murakami
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Tomoyuki Kobayashi
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kunihiro Shirai
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keisuke Kohama
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Jun-Ichi Hirata
- Department of Emergency and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
48
|
Zhang X, Shang L, Fan G, Gu X, Xu J, Wang Y, Huang L, Cao B. The Efficacy and Safety of Janus Kinase Inhibitors for Patients With COVID-19: A Living Systematic Review and Meta-Analysis. Front Med (Lausanne) 2022; 8:800492. [PMID: 35155477 PMCID: PMC8828941 DOI: 10.3389/fmed.2021.800492] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cytokine storm observed in patients with severe Coronavirus Disease 2019 (COVID-19) contributes to poor clinical outcomes and increased mortality. Janus kinases (JAKs) are important mediators in the cytokine storm. Therefore, we conduct a living systematic review and meta-analysis of the literature investigating efficacy and safety of JAK inhibitors for patients with COVID-19. METHODS Databases were searched up to December 1, 2021 for interventional and observational studies comparing JAK inhibitor treatment with concurrent control in patients with COVID-19. Efficacy and safety outcomes were evaluated by pooled risk ratio (RR). RESULTS Of 3,170 records retrieved, 15 studies were eligible and 13 were evaluated in the meta-analysis (n = 3,977). Based on data from three randomized controlled trials (RCTs), baricitinib treatment significantly decreased mortality by day 28 in hospitalized patients with COVID-19 (RR = 0.64, 95% CI 0.51-0.80) without increasing the incidence of adverse outcomes. In subgroup analysis, patients who required supplemental oxygen (RR = 0.62, 95% CI 0.41-0.95) or high-flow oxygen/non-invasive ventilation (RR = 0.59, 95% CI 0.42-0.85) at baseline benefited most. Pooled analysis of all eligible studies for JAK inhibitors (baricitinib, ruxolitinib, tofacitinib, and nezulcitinib) demonstrated a significant decrease in mortality (RR = 0.62, 95% CI 0.49-0.78) with no increase in the risk of adverse events. CONCLUSION Baricitinib probably decreases mortality in hospitalized adult patients with COVID-19, especially for patients who required supplemental oxygen or high-flow oxygen/non-invasive ventilation at baseline. The efficacy and safety of other JAK inhibitors, such as ruxolitinib, tofacitinib, and nezulcitinib, await more evidence. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021261414, identifier: CRD42021261414.
Collapse
Affiliation(s)
- Xueyang Zhang
- School of Medicine, Tsinghua University, Beijing, China
| | - Lianhan Shang
- Beijing University of Chinese Medicine, Beijing, China,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Guohui Fan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoying Gu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jiuyang Xu
- School of Medicine, Tsinghua University, Beijing, China,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yeming Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lixue Huang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China,Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing, China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China,Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing, China,Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China,*Correspondence: Bin Cao
| |
Collapse
|
49
|
Garbo R, Valent F, Gigli GL, Valente M. Pre-Existing Lymphopenia Increases the Risk of Hospitalization and Death after SARS-CoV-2 Infection. Infect Dis Rep 2022; 14:20-25. [PMID: 35076526 PMCID: PMC8788278 DOI: 10.3390/idr14010003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
There is limited information regarding the severity of COVID-19 in immunocompromized patients. We conducted a retrospective cohort study considering the period from 1 March 2020 to 31 December 2020 to determine whether previously existing lymphopenia increases the risk of hospitalization and death after SARS-CoV-2 infection in the general population. The laboratory and hospital discharge databases of the Azienda Sanitaria Universitaria Friuli Centrale were used, and 5415 subjects infected with SARS-CoV-2 and with at least one recent absolute lymphocyte count determination before SARS-CoV-2 positivity were included. In total, 817 (15.1%) patients had severe COVID-19. Patients developing severe COVID-19 were more frequently males (44.9% of the severe COVID-19 group vs. 41.5% in the non-severe COVID-19 group; p < 0.0001) and were older (73.2 ± 13.8 vs. 58.4 ± 20.3 years; p < 0.0001). Furthermore, 29.9% of the lymphopenic patients developed severe COVID-19 vs. 14.5% of the non-lymphopenic patients (p < 0.0001). In a logistic regression model, female sex remained a protective factor (OR = 0.514, 95%CI 0.438–0.602, p < 0.0001), while age and lymphopenia remained risk factors for severe COVID-19 (OR = 1.047, 95%CI 1.042–1.053, p < 0.0001 for each additional year of age; OR = 1.715, 95%CI 1.239–2.347, p = 0.0011 for lymphopenia). This provides further information to stratify the risk of COVID-19 severity, which may be an important element in the management of immunosuppressive therapies.
Collapse
Affiliation(s)
- Riccardo Garbo
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (G.L.G.); (M.V.)
- Correspondence:
| | - Francesca Valent
- Clinical and Evaluational Epidemiology Service, Department of Governance, Local Health Authority, 38123 Trento, Italy;
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (G.L.G.); (M.V.)
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (G.L.G.); (M.V.)
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| |
Collapse
|
50
|
Campochiaro C, Tomelleri A, Matucci-Cerinic M, Dagna L. One year later: The case of tocilizumab in COVID-19. Eur J Intern Med 2022; 95:5-6. [PMID: 34711474 PMCID: PMC8536517 DOI: 10.1016/j.ejim.2021.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Hospital, Via Olgettina 60, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Hospital, Via Olgettina 60, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Hospital, Via Olgettina 60, Milan 20132, Italy; Department of Experimental and Clinical Medicine, and Division of Rheumatology AOUC, University of Florence, Florence, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Hospital, Via Olgettina 60, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|