401
|
Abstract
The published results of the post-marketing ORAL Surveillance study, which compared the Janus kinase (JAK) inhibitor tofacitinib with anti-TNF therapy in older patients with rheumatoid arthritis who have cardiovascular risk factors, have led to changes in the recommendations for the use of JAK inhibitors. Although new safety signals have emerged for tofacitinib, namely malignancy and cardiovascular disease, it should be noted that these signals are relative to those seen with TNF blockers. The new data further raise our intrigue that venous thromboembolism might be a true risk related to JAK inhibition. Reassuringly, the totality of the findings from this newly published study and the other data collected to date suggest that JAK inhibitors can be used safely at approved doses by many patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Kevin L Winthrop
- Division of Infectious Diseases, Schools of Medicine and Public Health, Oregon Health and Sciences University, Portland, OR, USA.
| | - Stanley B Cohen
- Division of Rheumatology, Presbyterian Hospital, Dallas, TX, USA
| |
Collapse
|
402
|
Ho WS, Zhang R, Tan YL, Chai CLL. COVID-19 and the promise of small molecule therapeutics: Are there lessons to be learnt? Pharmacol Res 2022; 179:106201. [PMID: 35367622 PMCID: PMC8970615 DOI: 10.1016/j.phrs.2022.106201] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic had grounded the world to a standstill. As the disease continues to rage two years on, it is apparent that effective therapeutics are critical for a successful endemic living with COVID-19. A dearth in suitable antivirals has prompted researchers and healthcare professionals to investigate existing and developmental drugs against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although some of these drugs initially appeared to be promising for the treatment of COVID-19, they were ultimately found to be ineffective. In this review, we provide a retrospective analysis on the merits and limitations of some of these drugs that were tested against SARS-CoV-2 as well as those used for adjuvant therapy. While many of these drugs are no longer part of our arsenal for the treatment of COVID-19, important lessons can be learnt. The recent inclusion of molnupiravir and Paxlovid™ as treatment options for COVID-19 represent our best hope to date for endemic living with COVID-19. Our viewpoints on these two drugs and their prospects as current and future antiviral agents will also be provided.
Collapse
Affiliation(s)
- Wei Shen Ho
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Ruirui Zhang
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Yeong Lan Tan
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Christina Li Lin Chai
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
403
|
Olschewski H, Eber E, Bucher B, Hackner K, Handzhiev S, Hoetzenecker K, Idzko M, Klepetko W, Kovacs G, Lamprecht B, Löffler-Ragg J, Meilinger M, Müller A, Prior C, Schindler O, Täubl H, Zacharasiewicz A, Zwick RH, Arns BM, Bolitschek J, Cima K, Gingrich E, Hochmair M, Horak F, Jaksch P, Kropfmüller R, Pfleger A, Puchner B, Puelacher C, Rodriguez P, Salzer HJF, Schenk P, Stelzmüller I, Strenger V, Urban M, Wagner M, Wimberger F, Flick H. Management of patients with SARS-CoV-2 infections with focus on patients with chronic lung diseases (as of 10 January 2022) : Updated statement of the Austrian Society of Pneumology (ASP). Wien Klin Wochenschr 2022; 134:399-419. [PMID: 35449467 PMCID: PMC9022736 DOI: 10.1007/s00508-022-02018-x] [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: 01/19/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022]
Abstract
The Austrian Society of Pneumology (ASP) launched a first statement on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in May 2020, at a time when in Austria 285 people had died from this disease and vaccinations were not available. Lockdown and social distancing were the only available measures to prevent more infections and the breakdown of the health system. Meanwhile, in Austria over 13,000 patients have died in association with a SARS-CoV‑2 infection and coronavirus disease 2019 (COVID-19) was among the most common causes of death; however, SARS-CoV‑2 has been mutating all the time and currently, most patients have been affected by the delta variant where the vaccination is very effective but the omicron variant is rapidly rising and becoming predominant. Particularly in children and young adults, where the vaccination rate is low, the omicron variant is expected to spread very fast. This poses a particular threat to unvaccinated people who are at elevated risk of severe COVID-19 disease but also to people with an active vaccination. There are few publications that comprehensively addressed the special issues with SARS-CoV‑2 infection in patients with chronic lung diseases. These were the reasons for this updated statement. Pulmonologists care for many patients with an elevated risk of death in case of COVID-19 but also for patients that might be at an elevated risk of vaccination reactions or vaccination failure. In addition, lung function tests, bronchoscopy, respiratory physiotherapy and training therapy may put both patients and health professionals at an increased risk of infection. The working circles of the ASP have provided statements concerning these risks and how to avoid risks for the patients.
Collapse
Affiliation(s)
- Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.
| | - Ernst Eber
- Division of Paediatric Pulmonology and Allergology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Brigitte Bucher
- Department of Pulmonology, Tirol Kliniken, Hospital Hochzirl-Natters, Natters, Austria
| | - Klaus Hackner
- Department of Pneumology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Sabin Handzhiev
- Department of Pneumology, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Gabor Kovacs
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Bernd Lamprecht
- Department of Pulmonology, Faculty of Medicine, Johannes-Kepler-University, Linz, Austria
| | - Judith Löffler-Ragg
- Pulmonology, Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Meilinger
- Department of Internal Medicine and Pulmonology, Klinik Floridsdorf, Vienna, Austria
| | - Alexander Müller
- Department of Physical Medicine and Rehabilitation, Klinik Floridsdorf, Vienna, Austria
| | | | - Otmar Schindler
- Department of Internal and Respiratory Medicine, Hospital Graz II, Hospital Enzenbach, Gratwein, Austria
| | - Helmut Täubl
- Department of Pulmonology, Tirol Kliniken, Hospital Hochzirl-Natters, Natters, Austria
| | | | - Ralf Harun Zwick
- Outpatient Pulmonary Rehabilitation, Therme Wien Med, Vienna, Austria
| | | | - Josef Bolitschek
- Department of Pneumology, Ordensklinikum Linz Elisabethinen Hospital, Linz, Austria
| | - Katharina Cima
- Department of Pulmonology, Tirol Kliniken, Hospital Hochzirl-Natters, Natters, Austria
| | | | - Maximilian Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
| | | | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Roland Kropfmüller
- Department of Pulmonology, Faculty of Medicine, Johannes-Kepler-University, Linz, Austria
| | - Andreas Pfleger
- Division of Paediatric Pulmonology and Allergology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Bernhard Puchner
- Department of Pulmonology, Reha Zentrum Münster, Münster, Austria
| | | | - Patricia Rodriguez
- Division of Paediatric Pulmonology and Allergology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Helmut J F Salzer
- Department of Pulmonology, Faculty of Medicine, Johannes-Kepler-University, Linz, Austria
| | - Peter Schenk
- Department of Pulmonology, Landesklinikum Hochegg, Grimmenstein, Austria
| | | | - Volker Strenger
- Division of Paediatric Pulmonology and Allergology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Matthias Urban
- Department of Internal Medicine and Pulmonology, Klinik Floridsdorf, Vienna, Austria
| | - Marlies Wagner
- Division of Paediatric Pulmonology and Allergology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Franz Wimberger
- Department of Pneumology, Ordensklinikum Linz Elisabethinen Hospital, Linz, Austria
| | - Holger Flick
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
404
|
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a devastating pandemic. Although most people infected with SARS-CoV-2 develop a mild to moderate disease with virus replication restricted mainly to the upper airways, some progress to having a life-threatening pneumonia. In this Review, we explore recent clinical and experimental advances regarding SARS-CoV-2 pathophysiology and discuss potential mechanisms behind SARS-CoV-2-associated acute respiratory distress syndrome (ARDS), specifically focusing on new insights obtained using novel technologies such as single-cell omics, organoid infection models and CRISPR screens. We describe how SARS-CoV-2 may infect the lower respiratory tract and cause alveolar damage as a result of dysfunctional immune responses. We discuss how this may lead to the induction of a 'leaky state' of both the epithelium and the endothelium, promoting inflammation and coagulation, while an influx of immune cells leads to overexuberant inflammatory responses and immunopathology. Finally, we highlight how these findings may aid the development of new therapeutic interventions against COVID-19.
Collapse
|
405
|
Biddle K, White JPE, Sofat N. What is the full potential of baricitinib in treating patients with COVID-19? Expert Rev Clin Immunol 2022; 18:545-549. [PMID: 35486502 DOI: 10.1080/1744666x.2022.2072298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This has been removed as per the suggestions from the editorsEditorial.
Collapse
Affiliation(s)
- Kathryn Biddle
- St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, UK.,nstitute for Infection & Immunity, St George's, University of London and Department of Rheumatology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jonathan P E White
- St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, UK
| | - Nidhi Sofat
- St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, UK.,nstitute for Infection & Immunity, St George's, University of London and Department of Rheumatology, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
406
|
Levy G, Guglielmelli P, Langmuir P, Constantinescu S. JAK inhibitors and COVID-19. J Immunother Cancer 2022; 10:jitc-2021-002838. [PMID: 35459733 PMCID: PMC9035837 DOI: 10.1136/jitc-2021-002838] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
During SARS-CoV-2 infection, the innate immune response can be inhibited or delayed, and the subsequent persistent viral replication can induce emergency signals that may culminate in a cytokine storm contributing to the severe evolution of COVID-19. Cytokines are key regulators of the immune response and virus clearance, and, as such, are linked to the—possibly altered—response to the SARS-CoV-2. They act via a family of more than 40 transmembrane receptors that are coupled to one or several of the 4 Janus kinases (JAKs) coded by the human genome, namely JAK1, JAK2, JAK3, and TYK2. Once activated, JAKs act on pathways for either survival, proliferation, differentiation, immune regulation or, in the case of type I interferons, antiviral and antiproliferative effects. Studies of graft-versus-host and systemic rheumatic diseases indicated that JAK inhibitors (JAKi) exert immunosuppressive effects that are non-redundant with those of corticotherapy. Therefore, they hold the potential to cut-off pathological reactions in COVID-19. Significant clinical experience already exists with several JAKi in COVID-19, such as baricitinib, ruxolitinib, tofacitinib, and nezulcitinib, which were suggested by a meta-analysis (Patoulias et al.) to exert a benefit in terms of risk reduction concerning major outcomes when added to standard of care in patients with COVID-19. Yet, only baricitinib is recommended in first line for severe COVID-19 treatment by the WHO, as it is the only JAKi that has proven efficient to reduce mortality in individual randomized clinical trials (RCT), especially the Adaptive COVID-19 Treatment Trial (ACTT-2) and COV-BARRIER phase 3 trials. As for secondary effects of JAKi treatment, the main caution with baricitinib consists in the induced immunosuppression as long-term side effects should not be an issue in patients treated for COVID-19. We discuss whether a class effect of JAKi may be emerging in COVID-19 treatment, although at the moment the convincing data are for baricitinib only. Given the key role of JAK1 in both type I IFN action and signaling by cytokines involved in pathogenic effects, establishing the precise timing of treatment will be very important in future trials, along with the control of viral replication by associating antiviral molecules.
Collapse
Affiliation(s)
- Gabriel Levy
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium.,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| | - Paola Guglielmelli
- Department of Clinical and Experimental Medicine, University of Florence, Firenze, Italy.,Center of Research and Innovation for Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Peter Langmuir
- Oncology Targeted Therapeutics, Incyte Corp, Wilmington, Delaware, USA
| | - Stefan Constantinescu
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium .,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium.,Nuffield Department of Medicine, Oxford University, Ludwig Institute for Cancer Research, Oxford, UK
| |
Collapse
|
407
|
SARS-CoV-2 Infection Associated with Aplastic Anemia and Pure Red Cell Aplasia. Blood Adv 2022; 6:3840-3843. [PMID: 35452511 PMCID: PMC9040401 DOI: 10.1182/bloodadvances.2022007174] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
|
408
|
Niedźwiedzka-Rystwej P, Majchrzak A, Kurkowska S, Małkowska P, Sierawska O, Hrynkiewicz R, Parczewski M. Immune Signature of COVID-19: In-Depth Reasons and Consequences of the Cytokine Storm. Int J Mol Sci 2022; 23:4545. [PMID: 35562935 PMCID: PMC9105989 DOI: 10.3390/ijms23094545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
In the beginning of the third year of the fight against COVID-19, the virus remains at least still one step ahead in the pandemic "war". The key reasons are evolving lineages and mutations, resulting in an increase of transmissibility and ability to evade immune system. However, from the immunologic point of view, the cytokine storm (CS) remains a poorly understood and difficult to combat culprit of the extended number of in-hospital admissions and deaths. It is not fully clear whether the cytokine release is a harmful result of suppression of the immune system or a positive reaction necessary to clear the virus. To develop methods of appropriate treatment and therefore decrease the mortality of the so-called COVID-19-CS, we need to look deeply inside its pathogenesis, which is the purpose of this review.
Collapse
Affiliation(s)
| | - Adam Majchrzak
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| | - Sara Kurkowska
- Department of Nuclear Medicine, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Paulina Małkowska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Olga Sierawska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| |
Collapse
|
409
|
Russo A, Olivadese V, Trecarichi EM, Torti C. Bacterial Ventilator-Associated Pneumonia in COVID-19 Patients: Data from the Second and Third Waves of the Pandemic. J Clin Med 2022; 11:jcm11092279. [PMID: 35566405 PMCID: PMC9100863 DOI: 10.3390/jcm11092279] [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] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/30/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, many patients requiring invasive mechanical ventilation were admitted to intensive care units (ICU) for COVID-19-related severe respiratory failure. As a matter of fact, ICU admission and invasive ventilation increased the risk of ventilator-associated pneumonia (VAP), which is associated with high mortality rate and a considerable burden on length of ICU stay and healthcare costs. The objective of this review was to evaluate data about VAP in COVID-19 patients admitted to ICU that developed VAP, including their etiology (limiting to bacteria), clinical characteristics, and outcomes. The analysis was limited to the most recent waves of the epidemic. The main conclusions of this review are the following: (i) P. aeruginosa, Enterobacterales, and S. aureus are more frequently involved as etiology of VAP; (ii) obesity is an important risk factor for the development of VAP; and (iii) data are still scarce and increasing efforts should be put in place to optimize the clinical management and preventative strategies for this complex and life-threatening disease.
Collapse
|
410
|
Abstract
Purpose of Review Due to the rapidly changing landscape of COVID-19, the purpose of this review is to provide a concise and updated summary of pediatric COVID-19 diagnosis and management. Recent Findings The relative proportion of pediatric cases have significantly increased following the emergence of the Omicron variant (from < 2% in the early pandemic to 25% from 1/27 to 2/3/22). While children present with milder symptoms than adults, severe disease can still occur, particularly in children with comorbidities. There is a relative paucity of pediatric data in the management of COVID-19 and the majority of recommendations remain based on adult data. Summary Fever and cough remain the most common clinical presentations, although atypical presentations such as "COVID toes," anosmia, and croup may be present. Children are at risk for post-infectious complications such as MIS-C and long COVID. Nucleic acid amplification tests through respiratory PCR remain the mainstay of diagnosis. The mainstay of management remains supportive care and prevention through vaccination is highly recommended. In patients at increased risk of progression, interventions such as monoclonal antibody therapy, PO Paxlovid, or IV remdesivir × 3 days should be considered. In patients with severe disease, the use of remdesivir, dexamethasone, and immunomodulatory agents (tocilizumab, baricitinib) is recommended. Children can be at risk for thrombosis from COVID-19 and anticoagulation is recommended in children with markedly elevated D-dimer levels or superimposed clinical risk factors for hospital associated venous thromboembolism.
Collapse
Affiliation(s)
- Frank Zhu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Medical College of Wisconsin, Suite 450C, 999 North 92nd Street, Wauwatosa, Milwaukee, WI 53226 USA
| | - Jocelyn Y. Ang
- Division of Pediatric Infectious Diseases, Children’s Hospital of Michigan, Detroit, MI USA
- Department of Pediatrics, Central Michigan University College of Medicine, Mount Pleasant, MI USA
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI USA
| |
Collapse
|
411
|
Comparative Efficacy of Tocilizumab and Baricitinib Administration in COVID-19 Treatment: A Retrospective Cohort Study. Medicina (B Aires) 2022; 58:medicina58040513. [PMID: 35454352 PMCID: PMC9030469 DOI: 10.3390/medicina58040513] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
Background and Objectives: Tocilizumab and baricitinib have been observed to improve the outcomes of patients with coronavirus disease 2019 (COVID-19). However, a comparative evaluation of these drugs has not been performed. Materials and Methods: A retrospective, single-center study was conducted using the data of COVID-19 patients admitted to Hokkaido University hospital between April 2020 and September 2021, who were treated with tocilizumab or baricitinib. The clinical characteristics of the patients who received tocilizumab were compared to those of patients who received baricitinib. Univariate and multivariate logistic regression analyses of the outcomes of all-cause mortality and improvement in respiratory status were performed. The development of secondary infection events was analyzed using the Kaplan–Meier method and the log-rank test. Results: Of the 459 patients hospitalized with COVID-19 during the study, 64 received tocilizumab treatment and 34 baricitinib treatment, and those 98 patients were included in the study. Most patients were treated with concomitant steroids and exhibited the same severity level at the initiation of drug treatment. When compared to each other, neither tocilizumab nor baricitinib use were associated with all-cause mortality or improvement in respiratory status within 28 days from drug administration. Conclusions: Age, chronic renal disease and early administration of TCZ or BRT from the onset of COVID-19 were independent prognostic factors for all-cause mortality, whereas anti-viral drug use and the severity of COVID-19 at baseline were associated with an improvement in respiratory status. Secondary infection-free survival rates of patients treated with tocilizumab and those treated with baricitinib did not significantly differ. The results suggest that both tocilizumab and baricitinib could be clinically equivalent agents of choice in treatment of COVID-19.
Collapse
|
412
|
Panahi Y, Dadkhah M, Talei S, Gharari Z, Asghariazar V, Abdolmaleki A, Matin S, Molaei S. Can anti-parasitic drugs help control COVID-19? Future Virol 2022. [PMID: 35359702 PMCID: PMC8940209 DOI: 10.2217/fvl-2021-0160] [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: 07/06/2021] [Accepted: 02/28/2022] [Indexed: 01/18/2023]
Abstract
Novel COVID-19 is a public health emergency that poses a serious threat to people worldwide. Given the virus spreading so quickly, novel antiviral medications are desperately needed. Repurposing existing drugs is the first strategy. Anti-parasitic drugs were among the first to be considered as a potential treatment option for this disease. Even though many papers have discussed the efficacy of various anti-parasitic drugs in treating COVID-19 separately, so far, no single study comprehensively discussed these drugs. This study reviews some anti-parasitic recommended drugs to treat COVID-19, in terms of function and in vitro as well as clinical results. Finally, we briefly review the advanced techniques, such as artificial intelligence, that have been used to find effective drugs for the treatment of COVID-19.
Collapse
Affiliation(s)
- Yasin Panahi
- Department of Pharmacology & Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sahand Talei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Gharari
- Department of Biotechnology, Faculty of Biological Sciences, Al-Zahra University, Tehran, Iran
| | - Vahid Asghariazar
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Arash Abdolmaleki
- Department of Engineering Sciences, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Iran.,Bio Science & Biotechnology Research center (BBRC), Sabalan University of Advanced Technologies (SUAT), Namin, Iran
| | - Somayeh Matin
- Department of Internal Medicine, Imam Khomeini Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheila Molaei
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.,Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
413
|
Hoepler W, Traugott M, Zoufaly A, Schatzl M, Hind J, Wenisch C, Neuhold S. [Diagnosis and treatment of COVID-19 in intensive care units]. Med Klin Intensivmed Notfmed 2022; 117:177-186. [PMID: 35347341 PMCID: PMC8959071 DOI: 10.1007/s00063-022-00909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/22/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022]
Abstract
Treatment of coronavirus disease 2019 (COVID-19) is particularly challenging due to the rapid scientific advances and the often significant hypoxemia. Use of high-flow oxygen, noninvasive mask ventilation, and the technique of awake proning can sometimes avoid the need for intubation. Mechanical ventilation follows the principles of ventilation for acute respiratory distress syndrome (ARDS; lung protective ventilation) and is generally supplemented by consequent positioning therapy (with at least 16 h in prone position in multiple cycles). Antiviral therapy options such as remdesivir usually come too late for patients with COVID-19 in the ICU, the only exception being the administration of monoclonal antibodies for patients without seroconversion. The value of immunomodulatory therapy such as dexamethasone is undisputed. Interleukin‑6 antagonists, on the other hand, are rather problematic for ICU patients, and for Janus kinase inhibitors, data and experience are still insufficient in this context.
Collapse
Affiliation(s)
- Wolfgang Hoepler
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
| | - Marianna Traugott
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
| | - Alexander Zoufaly
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
- Tropeninstitut Wien, Wien, Österreich
| | - Martina Schatzl
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
| | - Julian Hind
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
| | - Christoph Wenisch
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich.
| | - Stephanie Neuhold
- 4. Medizinische Abteilung mit Infektions- und Tropenmedizin, Intensivstation, Klinik Favoriten, Wiener Gesundheitsverbund, Kundratstr. 3, 1100, Wien, Österreich
| |
Collapse
|
414
|
Shimada A, Ohnaka S, Kubo K, Nakashima M, Nagai A. COVID-19-Associated Pulmonary Aspergillosis in a Patient Treated With Remdesivir, Dexamethasone, and Baricitinib: A Case Report. Cureus 2022; 14:e23755. [PMID: 35518522 PMCID: PMC9064704 DOI: 10.7759/cureus.23755] [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] [Accepted: 04/02/2022] [Indexed: 12/15/2022] Open
Abstract
Remdesivir, dexamethasone, and baricitinib have recently been used to treat patients with coronavirus disease 2019 (COVID-19) and respiratory failure. However, the adverse effects of combination therapy have not been fully explored. A 64-year-old man was diagnosed with COVID-19 and was treated with remdesivir, dexamethasone, and baricitinib. His respiratory condition worsened on day 17, and in the following days, he was diagnosed with pneumomediastinum and COVID-19-associated pulmonary aspergillosis (CAPA). His condition improved with a reduction in the corticosteroid regime and antifungal treatment. This is the first case of pulmonary aspergillosis in a patient with COVID-19 that was treated with remdesivir, dexamethasone, and baricitinib.
Collapse
Affiliation(s)
- Ayako Shimada
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Shinnosuke Ohnaka
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Kosumi Kubo
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Masanao Nakashima
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| | - Atsushi Nagai
- Department of Respiratory Medicine, Shin-Yurigaoka General Hospital, Kanagawa, JPN
| |
Collapse
|
415
|
Henderson LA, Canna SW, Friedman KG, Gorelik M, Lapidus SK, Bassiri H, Behrens EM, Kernan KF, Schulert GS, Seo P, Son MBF, Tremoulet AH, VanderPluym C, Yeung RSM, Mudano AS, Turner AS, Karp DR, Mehta JJ. American College of Rheumatology Clinical Guidance for Multisystem Inflammatory Syndrome in Children Associated With SARS-CoV-2 and Hyperinflammation in Pediatric COVID-19: Version 3. Arthritis Rheumatol 2022; 74:e1-e20. [PMID: 35118829 PMCID: PMC9011620 DOI: 10.1002/art.42062] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To provide guidance on the management of Multisystem Inflammatory Syndrome in Children (MIS-C), a condition characterized by fever, inflammation, and multiorgan dysfunction that manifests late in the course of SARS-CoV-2 infection. Recommendations are also provided for children with hyperinflammation during COVID-19, the acute, infectious phase of SARS-CoV-2 infection. METHODS The Task Force is composed of 9 pediatric rheumatologists and 2 adult rheumatologists, 2 pediatric cardiologists, 2 pediatric infectious disease specialists, and 1 pediatric critical care physician. Preliminary statements addressing clinical questions related to MIS-C and hyperinflammation in COVID-19 were developed based on evidence reports. Consensus was built through a modified Delphi process that involved anonymous voting and webinar discussion. A 9-point scale was used to determine the appropriateness of each statement (median scores of 1-3 for inappropriate, 4-6 for uncertain, and 7-9 for appropriate). Consensus was rated as low, moderate, or high based on dispersion of the votes. Approved guidance statements were those that were classified as appropriate with moderate or high levels of consensus, which were prespecified before voting. RESULTS The guidance was approved in June 2020 and updated in November 2020 and October 2021, and consists of 41 final guidance statements accompanied by flow diagrams depicting the diagnostic pathway for MIS-C and recommendations for initial immunomodulatory treatment of MIS-C. CONCLUSION Our understanding of SARS-CoV-2-related syndromes in the pediatric population continues to evolve. This guidance document reflects currently available evidence coupled with expert opinion, and will be revised as further evidence becomes available.
Collapse
Affiliation(s)
| | - Scott W. Canna
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| | - Kevin G. Friedman
- Boston Children's Hospital and Harvard Medical SchoolBostonMassachusetts
| | - Mark Gorelik
- Morgan Stanley Children’s Hospital and Columbia UniversityNew YorkNew York
| | - Sivia K. Lapidus
- Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center and Hackensack Meridian School of MedicineHackensackNew Jersey
| | - Hamid Bassiri
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| | - Edward M. Behrens
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| | - Kate F. Kernan
- University of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Grant S. Schulert
- Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of MedicineCincinnatiOhio
| | - Philip Seo
- Johns Hopkins University School of MedicineBaltimoreMaryland
| | - Mary Beth F. Son
- Boston Children's Hospital and Harvard Medical SchoolBostonMassachusetts
| | - Adriana H. Tremoulet
- University of California San Diego and Rady Children’s Hospital San DiegoCalifornia
| | | | - Rae S. M. Yeung
- The Hospital for Sick Children and University of TorontoTorontoOntarioCanada
| | | | | | - David R. Karp
- University of Texas Southwestern Medical CenterDallas
| | - Jay J. Mehta
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| |
Collapse
|
416
|
Ely EW, Ramanan AV, Kartman CE, de Bono S, Liao R, Piruzeli MLB, Goldman JD, Saraiva JFK, Chakladar S, Marconi VC. Efficacy and safety of baricitinib plus standard of care for the treatment of critically ill hospitalised adults with COVID-19 on invasive mechanical ventilation or extracorporeal membrane oxygenation: an exploratory, randomised, placebo-controlled trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10. [PMID: 35123660 PMCID: PMC8813065 DOI: 10.1016/s2213-2600(22)00006-6 10.1016/s2213-2600(22)00057-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
BACKGROUND The oral, selective Janus kinase 1/2 inhibitor baricitinib has shown efficacy in studies of hospitalised adults with COVID-19. COV-BARRIER (NCT04421027) was a multinational, phase 3, randomised, double-blind, placebo-controlled trial of baricitinib in patients with confirmed SARS-CoV-2 infection. We aimed to evaluate the efficacy and safety of baricitinib plus standard of care in critically ill hospitalised adults with COVID-19 requiring invasive mechanical ventilation or extracorporeal membrane oxygenation. METHODS This exploratory trial followed the study design of COV-BARRIER in a critically ill cohort not included in the main phase 3 trial. The study was conducted across 18 hospitals in Argentina, Brazil, Mexico, and the USA. Participants (aged ≥18 years) hospitalised with laboratory-confirmed SARS-CoV-2 infection on baseline invasive mechanical ventilation or extracorporeal membrane oxygenation were randomly assigned (1:1) to baricitinib (4 mg) or placebo once daily for up to 14 days in combination with standard of care. Participants, study staff, and investigators were masked to study group assignment. Prespecified endpoints included all-cause mortality through days 28 and 60, number of ventilator-free days, duration of hospitalisation, and time to recovery through day 28. The efficacy analysis was done in the intention-to-treat population and the safety analysis was done in the safety population. This trial is registered with ClinicalTrials.gov, NCT04421027. FINDINGS Between Dec 23, 2020, and April 10, 2021, 101 participants were enrolled into the exploratory trial and assigned to baricitinib (n=51) or placebo (n=50) plus standard of care. Standard of care included baseline systemic corticosteroid use in 87 (86%) participants. Treatment with baricitinib significantly reduced 28-day all-cause mortality compared with placebo (20 [39%] of 51 participants died in the baricitinib group vs 29 [58%] of 50 in the placebo group; hazard ratio [HR] 0·54 [95% CI 0·31-0·96]; p=0·030; 46% relative reduction; absolute risk reduction 19%). A significant reduction in 60-day mortality was also observed in the baricitinib group compared with the placebo group (23 [45%] events vs 31 [62%]; HR 0·56 [95% CI 0·33-0·97]; p=0·027; 44% relative reduction; absolute risk reduction 17%). In every six baricitinib-treated participants, one additional death was prevented compared with placebo at days 28 and 60. The number of ventilator-free days did not differ significantly between treatment groups (mean 8·1 days [SD 10·2] in the baricitinib group vs 5·5 days [8·4] in the placebo group; p=0·21). The mean duration of hospitalisation in baricitinib-treated participants was not significantly shorter than in placebo-treated participants (23·7 days [SD 7·1] vs 26·1 days [3·9]; p=0·050). The rates of infections, blood clots, and adverse cardiovascular events were similar between treatment groups. INTERPRETATION In critically ill hospitalised patients with COVID-19 who were receiving invasive mechanical ventilation or extracorporeal membrane oxygenation, treatment with baricitinib compared with placebo (in combination with standard of care, including corticosteroids) reduced mortality, which is consistent with the mortality reduction observed in less severely ill patients in the hospitalised primary COV-BARRIER study population. However, this was an exploratory trial with a relatively small sample size; therefore, further phase 3 trials are needed to confirm these findings. FUNDING Eli Lilly and Company.
Collapse
Affiliation(s)
- E Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center (GRECC), Nashville, TN, USA.
| | - Athimalaipet V Ramanan
- Translational Health Sciences, University of Bristol, Bristol, UK; Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
| | | | | | - Ran Liao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Providence St Joseph Health, Seattle, WA, USA; Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Vincent C Marconi
- Emory University School of Medicine, Rollins School of Public Health, Emory Vaccine Center, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
417
|
Supady A, Zeiser R. Baricitinib for patients with severe COVID-19—time to change the standard of care? THE LANCET RESPIRATORY MEDICINE 2022; 10:314-315. [PMID: 35123659 PMCID: PMC8813061 DOI: 10.1016/s2213-2600(22)00021-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Alexander Supady
- Department of Medicine III, Interdisciplinary Medical Intensive Care, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau 79106, Germany; Department of Cardiology and Angiology I, Heart Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau 79106, Germany; Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany.
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau 79106, Germany
| |
Collapse
|
418
|
Ely EW, Ramanan AV, Kartman CE, de Bono S, Liao R, Piruzeli MLB, Goldman JD, Saraiva JFK, Chakladar S, Marconi VC. Efficacy and safety of baricitinib plus standard of care for the treatment of critically ill hospitalised adults with COVID-19 on invasive mechanical ventilation or extracorporeal membrane oxygenation: an exploratory, randomised, placebo-controlled trial. THE LANCET RESPIRATORY MEDICINE 2022; 10:327-336. [PMID: 35123660 PMCID: PMC8813065 DOI: 10.1016/s2213-2600(22)00006-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
Background The oral, selective Janus kinase 1/2 inhibitor baricitinib has shown efficacy in studies of hospitalised adults with COVID-19. COV-BARRIER (NCT04421027) was a multinational, phase 3, randomised, double-blind, placebo-controlled trial of baricitinib in patients with confirmed SARS-CoV-2 infection. We aimed to evaluate the efficacy and safety of baricitinib plus standard of care in critically ill hospitalised adults with COVID-19 requiring invasive mechanical ventilation or extracorporeal membrane oxygenation. Methods This exploratory trial followed the study design of COV-BARRIER in a critically ill cohort not included in the main phase 3 trial. The study was conducted across 18 hospitals in Argentina, Brazil, Mexico, and the USA. Participants (aged ≥18 years) hospitalised with laboratory-confirmed SARS-CoV-2 infection on baseline invasive mechanical ventilation or extracorporeal membrane oxygenation were randomly assigned (1:1) to baricitinib (4 mg) or placebo once daily for up to 14 days in combination with standard of care. Participants, study staff, and investigators were masked to study group assignment. Prespecified endpoints included all-cause mortality through days 28 and 60, number of ventilator-free days, duration of hospitalisation, and time to recovery through day 28. The efficacy analysis was done in the intention-to-treat population and the safety analysis was done in the safety population. This trial is registered with ClinicalTrials.gov, NCT04421027. Findings Between Dec 23, 2020, and April 10, 2021, 101 participants were enrolled into the exploratory trial and assigned to baricitinib (n=51) or placebo (n=50) plus standard of care. Standard of care included baseline systemic corticosteroid use in 87 (86%) participants. Treatment with baricitinib significantly reduced 28-day all-cause mortality compared with placebo (20 [39%] of 51 participants died in the baricitinib group vs 29 [58%] of 50 in the placebo group; hazard ratio [HR] 0·54 [95% CI 0·31–0·96]; p=0·030; 46% relative reduction; absolute risk reduction 19%). A significant reduction in 60-day mortality was also observed in the baricitinib group compared with the placebo group (23 [45%] events vs 31 [62%]; HR 0·56 [95% CI 0·33–0·97]; p=0·027; 44% relative reduction; absolute risk reduction 17%). In every six baricitinib-treated participants, one additional death was prevented compared with placebo at days 28 and 60. The number of ventilator-free days did not differ significantly between treatment groups (mean 8·1 days [SD 10·2] in the baricitinib group vs 5·5 days [8·4] in the placebo group; p=0·21). The mean duration of hospitalisation in baricitinib-treated participants was not significantly shorter than in placebo-treated participants (23·7 days [SD 7·1] vs 26·1 days [3·9]; p=0·050). The rates of infections, blood clots, and adverse cardiovascular events were similar between treatment groups. Interpretation In critically ill hospitalised patients with COVID-19 who were receiving invasive mechanical ventilation or extracorporeal membrane oxygenation, treatment with baricitinib compared with placebo (in combination with standard of care, including corticosteroids) reduced mortality, which is consistent with the mortality reduction observed in less severely ill patients in the hospitalised primary COV-BARRIER study population. However, this was an exploratory trial with a relatively small sample size; therefore, further phase 3 trials are needed to confirm these findings. Funding Eli Lilly and Company.
Collapse
Affiliation(s)
- E Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center (GRECC), Nashville, TN, USA.
| | - Athimalaipet V Ramanan
- Translational Health Sciences, University of Bristol, Bristol, UK; Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
| | | | | | - Ran Liao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Providence St Joseph Health, Seattle, WA, USA; Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Vincent C Marconi
- Emory University School of Medicine, Rollins School of Public Health, Emory Vaccine Center, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
419
|
García-Lledó A, Gómez-Pavón J, González Del Castillo J, Hernández-Sampelayo T, Martín-Delgado MC, Martín Sánchez FJ, Martínez-Sellés M, Molero García JM, Moreno Guillén S, Rodríguez-Artalejo FJ, Ruiz-Galiana J, Cantón R, De Lucas Ramos P, García-Botella A, Bouza E. Pharmacological treatment of COVID-19: an opinion paper. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2022; 35:115-130. [PMID: 34894208 PMCID: PMC8972693 DOI: 10.37201/req/158.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The precocity and efficacy of the vaccines developed so far against COVID-19 has been the most significant and saving advance against the pandemic. The development of vaccines has not prevented, during the whole period of the pandemic, the constant search for therapeutic medicines, both among existing drugs with different indications and in the development of new drugs. The Scientific Committee of the COVID-19 of the Illustrious College of Physicians of Madrid wanted to offer an early, simplified and critical approach to these new drugs, to new developments in immunotherapy and to what has been learned from the immune response modulators already known and which have proven effective against the virus, in order to help understand the current situation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - E Bouza
- Servicio de Microbiología Clínica y Enfermedades Infecciosas del Hospital General Universitario Gregorio Marañón, Universidad Complutense. CIBERES. Ciber de Enfermedades Respiratorias. Madrid, Spain.
| |
Collapse
|
420
|
Alfano G, Morisi N, Frisina M, Ferrari A, Fontana F, Tonelli R, Franceschini E, Meschiari M, Donati G, Guaraldi G. Awaiting a cure for COVID-19: therapeutic approach in patients with different severity levels of COVID-19. LE INFEZIONI IN MEDICINA 2022; 30:11-21. [PMID: 35350263 DOI: 10.53854/liim-3001-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
Abstract
COVID-19 is an unpredictable infectious disease caused by SARS-CoV-2. The development of effective anti-COVID-19 vaccines has enormously minimized the risk of severe illness in most immunocompetent patients. However, unvaccinated patients and non-responders to the COVID-19 vaccine are at risk of shortand long-term consequences. In these patients, the outcome of COVID-19 relies on an interplay of multiple factors including age, immunocompetence, comorbidities, inflammatory response triggered by the virus as well as the virulence of SARS-CoV-2 variants. Generally, COVID-19 is asymptomatic or mildly symptomatic in young people, but it may manifest with respiratory insufficiency requiring mechanical ventilation in certain susceptible groups of patients. Furthermore, severe SARS-CoV-2 infection induces multiorgan failure syndrome by affecting liver, kidney heart and nervous system. Since December 2019, multiple drugs have been tested to treat COVID-19, but only a few have been proven effective to mitigate the course of the disease that continues to cause death and comorbidity worldwide. Current treatment of COVID-19 patients is essentially based on the administration of supportive oxygen therapy and the use of specific drugs such as steroids, anticoagulants, antivirals, anti-SARS-CoV-2 antibodies and immunomodulators. However, the rapid spread of new variants and the release of new data coming from the numerous ongoing clinical trials have created the conditions for maintaining a continuous updating of the therapeutic management of COVID-19 patients. Furthermore, we believe that a well-established therapeutic strategy along with the continuum of medical care for all patients with COVID-19 is pivotal to improving disease outcomes and restoring healthcare care fragmentation caused by the pandemic. This narrative review, focusing on the therapeutic management of COVID-19 patients, aimed to provide an overview of current therapies for (i) asymptomatic or mildly/moderate symptomatic patients, (ii) hospitalized patients requiring low-flow oxygen, (iii) high-flow oxygen and (iv) mechanical ventilation.
Collapse
Affiliation(s)
- Gaetano Alfano
- Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy.,Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Italy.,Clinical and Experimental Medicine, PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Morisi
- Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Monica Frisina
- Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy
| | - Annachiara Ferrari
- Internal and Emergency Medicine, Baggiovara Hospital, Baggiovara, Modena, Italy.,Department of Specialistic Medicine, Azienda USL-IRCCS di Reggio Emilia, Italy
| | - Francesco Fontana
- Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Italy
| | - Roberto Tonelli
- Respiratory Diseases Unit and Center for Rare Lung Disease, Department of Surgical and Medical Sciences, University Hospital of Modena, Italy.,Clinical and Experimental Medicine, PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Gabriele Donati
- Surgical, Medical and Dental Department of Morphological Sciences, Section of Nephrology, University of Modena and Reggio Emilia, Modena, Italy.,Nephrology Dialysis and Transplant Unit, University Hospital of Modena, Italy
| | | |
Collapse
|
421
|
Fluvoxamine in Nonhospitalized Patients With Acute COVID-19 Infection and the Lack of Efficacy in Reducing Rates of Hospitalization, Mechanical Ventilation, and Mortality in Placebo-Controlled Trials: A Systematic Review and Meta-Analysis. Am J Ther 2022; 29:e298-e304. [PMID: 35383578 DOI: 10.1097/mjt.0000000000001496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Recent clinical trials have investigated the use of fluvoxamine in preventing clinical deterioration in nonhospitalized patients with acute COVID-19 infection via stimulation of sigma-1 receptors, which regulates cytokine production and functional inhibition of acid sphingomyelinase activity, which may prevent infection of epithelial cells with SARS-CoV-2. However, the role of fluvoxamine is currently unclear because of a paucity of studies, particularly because the drug is being repurposed as an immunomodulatory and antiviral agent. STUDY QUESTION Aim of our meta-analysis was to investigate the efficacy of fluvoxamine in nonhospitalized patients with acute COVID-19 infection. DATA SOURCE Comprehensive literature search of PubMed, Embase, Cochrane Library databases, and Web of Science was performed from inception to February 10, 2022, for studies comparing fluvoxamine versus placebo for outpatient management of COVID-19. STUDY DESIGN The primary outcome of interest was rate of hospitalization. The secondary outcomes were rates of patients requiring mechanical ventilation and mortality. The random-effects model was used to calculate the risk ratios (RR) and confidence intervals (CI). A P value <0.05 was considered statistically significant. Heterogeneity was assessed using the Higgins I2 index. RESULTS Three studies (2 randomized controlled trials and one prospective cohort trial) involving 1762 patients were included in the meta-analysis. In patients who received fluvoxamine compared with placebo, there was no significant difference in rates of hospitalization (RR 0.26, 95% CI, 0.04-1.73, P = 0.16, I2 = 62%), mechanical ventilation (RR 0.73, 95% CI, 0.45-1.19, P = 0.21, I2 = 0%), and mortality (RR 0.67, 95% CI, 0.37-1.22, P = 0.19, I2 = 0%). CONCLUSION Current evidence does not indicate a significant effect of fluvoxamine on the rates of hospitalization, mechanical ventilation, and mortality of patients with COVID-19 infection.
Collapse
|
422
|
Coutre SE, Barnett C, Osiyemi O, Hoda D, Ramgopal M, Fort AC, Qaqish R, Hu Y, Ninomoto J, Alami NN, Styles L, Treon SP. Ibrutinib for Hospitalized Adults with Severe COVID-19 Infection: Results of the Randomized, Double-Blind, Placebo-Controlled iNSPIRE Study. Open Forum Infect Dis 2022; 9:ofac104. [PMID: 35493119 PMCID: PMC8992313 DOI: 10.1093/ofid/ofac104] [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: 12/08/2021] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
Abstract
Background Few therapies are approved for hospitalized patients with severe coronavirus disease 2019 (COVID-19). Ibrutinib, a once-daily Bruton tyrosine kinase inhibitor, may mitigate COVID-19–induced lung damage by reducing inflammatory cytokines. The multicenter, randomized, double-blind phase 2 iNSPIRE study evaluated ibrutinib for prevention of respiratory failure in hospitalized patients with severe COVID-19. Methods Adult patients with severe COVID-19 requiring hospitalization and supplemental oxygen but without respiratory failure were randomized 1:1 (stratified by remdesivir prescription) to ibrutinib 420 mg or placebo once daily for up to 28 days plus standard of care (SOC), including remdesivir and/or dexamethasone. Results Forty-six patients were randomized to ibrutinib plus SOC (n = 22) or placebo plus SOC (n = 24). The primary endpoint (proportion of patients alive and without respiratory failure through day 28) was not met, with no statistically significant difference adjusting for remdesivir prescription (86% with ibrutinib plus SOC vs 79% with placebo plus SOC; adjusted difference, 5.8% [80% confidence interval, –9.2% to 20.4%]; P = .599). Secondary endpoints also showed no statistically significant improvement with ibrutinib plus SOC. Median treatment duration was 14 days for ibrutinib and placebo. Adverse events were similar with ibrutinib plus SOC vs placebo plus SOC (overall: 55% vs 50%; serious: 18% vs 13%) and were consistent with the known safety profile of ibrutinib. Conclusions Addition of ibrutinib to SOC did not improve the proportion of patients alive and without respiratory failure through day 28 in hospitalized patients with severe COVID-19. Ibrutinib had a manageable safety profile, with similar safety to placebo. Clinical Trials Registration NCT04375397.
Collapse
Affiliation(s)
- Steven E Coutre
- Stanford Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher Barnett
- MedStar Washington Hospital Center, Washington, DC, USA, and University of California San Francisco, San Francisco, CA, USA
| | | | - Daanish Hoda
- Intermountain Healthcare, Salt Lake City, UT, USA
| | - Moti Ramgopal
- Midway Immunology and Research Center, Fort Pierce, FL, USA
| | | | | | | | - Joi Ninomoto
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA, USA
| | | | - Lori Styles
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA, USA
| | | |
Collapse
|
423
|
Londres HD, Armada JJ, Martínez AH, Abdo Cuza AA, Sánchez YH, Rodríguez AG, Figueroa SS, Llanez Gregorich EM, Torres Lahera ML, Peire FG, González TM, González YZ, Añé Kouri AL, Palomo AG, Concepción MT, Pérez LM, Luaces-Alvarez PL, Iglesias DE, Hernández DS, Suzarte MR, Ramos TC. Blocking EGFR with nimotuzumab: a novel strategy for COVID-19 treatment. Immunotherapy 2022; 14:521-530. [PMID: 35306855 PMCID: PMC8936166 DOI: 10.2217/imt-2022-0027] [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] [Indexed: 12/15/2022] Open
Abstract
Background: Lung injury and STAT1 deficit induce EGFR overexpression in SARS-CoV-2 infection. Patients & methods: A phase I/II trial was done to evaluate the safety and preliminary effect of nimotuzumab, an anti-EGFR antibody, in COVID-19 patients. Patients received from one to three infusions together with other drugs included in the national guideline. Results: 41 patients (31 severe and 10 moderate) received nimotuzumab. The median age was 62 years and the main comorbidities were hypertension, diabetes and cardiovascular disease. The antibody was very safe and the 14-day recovery rate was 82.9%. Inflammatory markers decreased over time. Patients did not show signs of fibrosis. Conclusion: Nimotuzumab is a safe antibody that might reduce inflammation and prevent fibrosis in severe and moderate COVID-19 patients. Clinical Trial Registration: RPCEC00000369 (rpcec.sld.cu). Background: After SARS-CoV-2 infection, many cells in the lung express a new receptor called EGFR. Overexpression of EGFR can worsen the pulmonary disease and provoke fibrosis. Patients & methods: The initial impact of using a drug that blocks EGFR, nimotuzumab, was evaluated in COVID-19 patients. Results: 41 patients received nimotuzumab by the intravenous route together with other medications. The median age was 62 years, and patients had many chronic conditions including hypertension, diabetes and cardiac problems. Treatment was well tolerated and 82.9% of the patients were discharged by day 14. Serial laboratory tests, x-rays and CT scan evaluations showed the improvement of the patients. Conclusion: Nimotuzumab is a safe drug that can be useful to treat COVID-19 patients.
Collapse
|
424
|
Barletta JF, Erstad BL. Drug dosing in hospitalized obese patients with COVID-19. Crit Care 2022; 26:60. [PMID: 35287690 PMCID: PMC8919144 DOI: 10.1186/s13054-022-03941-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
Obesity is highly prevalent in hospitalized patients admitted with COVID-19. Evidence based guidelines are available for COVID-19-related therapies but dosing information specific to patients with obesity is lacking. Failure to account for the pharmacokinetic alterations that exist in this population can lead to underdosing, and treatment failure, or overdosing, resulting in an adverse effect. The objective of this manuscript is to provide clinicians with guidance for making dosing decisions for medications used in the treatment of patients with COVID-19. A detailed literature search was conducted for medications listed in evidence-based guidelines from the National Institutes of Health with an emphasis on pharmacokinetics, dosing and obesity. Retrieved manuscripts were evaluated and the following prioritization strategy was used to form the decision framework for recommendations: clinical outcome data > pharmacokinetic studies > adverse effects > physicochemical properties. Most randomized controlled studies included a substantial number of patients who were obese but few had large numbers of patients more extreme forms of obesity. Pharmacokinetic data have described alterations with volume of distribution and clearance but this variability does not appear to warrant dosing modifications. Future studies should provide more information on size descriptors and stratification of data according to obesity and body habitus.
Collapse
Affiliation(s)
- Jeffrey F Barletta
- Department of Pharmacy Practice, College of Pharmacy, Midwestern University, 19555 N 59th Avenue, Glendale, AZ, 85038, USA.
| | - Brian L Erstad
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, 1295 N Martin Ave, PO Box 210202, Tucson, AZ, 85721, USA
| |
Collapse
|
425
|
Racial/ethnic disparities on inflammation and response to methylprednisolone in severe COVID-19 pneumonia. BMC Infect Dis 2022; 22:254. [PMID: 35287602 PMCID: PMC8919360 DOI: 10.1186/s12879-022-07237-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Background Racial/ethnic minorities are at higher risk for severe COVID-19. This may be related to social determinants that lead to chronic inflammatory states. The aims of the study were to determine if there are racial/ethnic disparities with inflammatory markers and association of methylprednisolone to in hospital survival. Methods This was a secondary analysis of a retrospective cohort study of patients ≥ 18 years of age and admitted for severe COVID-19 pneumonia between March and June 2020 in 13 Hospitals in New Jersey, United States. Patients who received other formulation of corticosteroids were not included. Area under the receiver operating characteristics curves were performed to test for discriminatory ability of each inflammatory makers. Univariate and multivariate Cox regression assessed the association of variables to in hospital survival. Results Propensity matched sample (n = 759) between no methylprednisolone (n = 380) and methylprednisolone (n = 379) had 338 Whites, 102 Blacks, 61 Asian/Indians, and 251 non-Black non-White Hispanics. Compared to CRP, area under receiving operating characteristic curve for d-dimer in Hispanics (0.742) was statistically different (DeLong Test P = 0.0041). Multivariate cox regression showed that different variables in Blacks [age ≥ 60 years (HR = 3.71, P = 0.0281), mechanical ventilation (HR = 5.07, P = 0.0281) and creatinine ≥ 1.5 mg/dL (HR = 3.61, P = 0.0007)], Whites [cancer (HR = 1.68, P = 0.0213), qSOFA score of 1 (HR = 1.81, P = 0.0213), qSOFA score of 2 (HR = 5.16, P < 0.0001), qSOFA score of 3 (HR = 11.81, P < 0.0001) and creatinine ≥ 1.5 mg/dL (HR = 2.16, P = 0.0006)], Hispanics [hypertension (HR = 2.52, P = 0.0007), cancer (HR = 2.99, P = 0.0244 and D-dimer ≥ 2 mcg/mL (HR = 2.22, P = 0.0077)], and Asian/Indians [
chronic kidney disease (HR = 6.36, P = 0.0031) and CRP > 20 mg/L (HR = 5.02, P = 0.0032)] were statistically significant for mortality. Low dose and high dose methylprednisolone were significantly associated with prolonged survival in Whites [low dose (HR = 0.37, P < 0.0001) and high dose (HR = 0.48, P < 0.0183)] and Asian/Indians [low dose (HR = 0.13, P = 0.0101) and high dose (HR = 0.15, P = 0.01)]. However, high dose was not associated with improved survival compared to low dose. Methylprednisolone was not associated with prolonged survival in Blacks and Hispanics. Conclusion Racial/Ethnic disparities with inflammatory markers preclude the use of one marker as a predictor of survival. Methylprednisolone is associated with prolonged survival in Asian/Indians and Whites. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07237-1.
Collapse
|
426
|
Association of high SARS-CoV-2 RNAemia with diabetes and mortality in COVID-19 critically ill patient. iScience 2022; 25:104075. [PMID: 35309726 PMCID: PMC8920087 DOI: 10.1016/j.isci.2022.104075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
It has been suggested that during the period of respiratory worsening of severe COVID-19 patients, viral replication plays a less important role than inflammation. Using the droplet-based digital PCR (ddPCR) for precise quantification of plasma SARS-CoV-2 viral load (SARS-CoV-2 RNAemia), we investigated the relationship between plasma viral load, comorbidities, and mortality of 122 critically ill COVID-19 patients. SARS-CoV-2 RNAemia was detected by ddPCR in 90 (74%) patients, ranging from 70 to 213,152 copies per mL. A high (>1 000 copies/ml) or very high (>10,000 copies/ml) SARS-Cov-2 RNAemia was observed in 46 patients (38%), of which 26 were diabetic. Diabetes was independently associated with a higher SARS-CoV-2 RNAemia. In multivariable logistic regression models, SARS-CoV-2 RNAemia was strongly and independently associated with day-60 mortality. Early initiation of antiviral therapies might be considered in COVID-19 critically ill patients with high RNAemia. Plasma SARS-CoV-2 RNA was detected in 90 of 122 COVID-19 critically ill patients Plasma SARS-CoV-2 RNA was high (>1000 copies per mL) in 46 patients Diabetic patients had significantly higher levels of plasma SARS-CoV-2 RNA Plasma SARS-CoV-2 RNA levels were independently associated with day-60 mortality
Collapse
|
427
|
Jansen‐van Vuuren RD, Vohra R. Synthesis of [
2
H
5
]baricitinib via [
2
H
5
]ethanesulfonyl chloride. J Labelled Comp Radiopharm 2022; 65:156-161. [PMID: 35277889 PMCID: PMC9313827 DOI: 10.1002/jlcr.3969] [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: 01/25/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/23/2022]
Abstract
Baricitinib, typically applied as a treatment for rheumatoid arthritis, has recently attracted the attention of clinicians and researchers as a potential treatment for COVID‐19. Naturally, there has been a need for the preparation of the isotope‐labelled analogue of baricitinib to probe the pharmacokinetics of baricitinib in this new role. As such, we have developed a simple synthetic route to deuterated [2H5]baricitinib, facilitating its formation over four steps and in a 29% overall yield based on starting [2H5]ethanethiol (19% if we start with [2H5]bromoethane instead). A critical component of the overall process involves the synthesis of [2H5]ethanesulfonyl chloride, and we describe in detail the two routes that were explored to optimize this step.
Collapse
Affiliation(s)
- Ross D. Jansen‐van Vuuren
- Department of Chemistry Queen’s University Kingston ON CANADA
- Faculty of Chemistry and Chemical Technology University of Ljubljana Ljubljana CANADA
| | | |
Collapse
|
428
|
Arcani R, Cauchois R, Suchon P, Jean R, Jarrot P, Pinho QGD, Dalmas J, Jean E, Andre B, Veit V, Koubi M, Kaplanski G. Factors associated with dexamethasone efficacy in COVID‐19. A retrospective investigative cohort study. J Med Virol 2022; 94:3169-3175. [PMID: 35277862 PMCID: PMC9088322 DOI: 10.1002/jmv.27712] [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: 12/28/2021] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 11/09/2022]
Abstract
Dexamethasone has demonstrated efficacy in reducing mortality in COVID‐19. However, its practical use is badly defined. We aimed to investigate factors associated with dexamethasone efficacy in real life. Our retrospective study was conducted in two university hospitals between September and November 2020 and included all the consecutive hospitalized patients with a laboratory‐confirmed SARS‐CoV‐2 infection assessed by RT‐PCR, treated with intravenous dexamethasone (6 mg/day). Among 111 patients, 10.6% necessitated a transfer into the intensive care unit (ICU) and the 28‐day mortality rate was 17.1%. The 28‐day mortality rate was significantly lower in patients who demonstrated improvement at 48 h (hazard ratio [HR]: 0.17, 95% confidence interval [CI]: 0.04–0.78, p = 0.02) and 96 h (HR: 0.07, 95% CI: 0.02–0.31, p = 0.0005) after dexamethasone initiation. Apart from well‐known risk factors (age, hypertension, active cancer, severe lesions on chest computed tomography [CT] scan), we found that a high viral load in nasopharyngeal swab (Cycle threshold <30) at dexamethasone initiation was associated with higher 28‐day mortality (66.6% vs. 36.7%, p = 0.03). Patients who did not receive antibiotics at dexamethasone initiation had a higher rate of transfer into the ICU (55.6% vs. 23.5%, p = 0.045) with a trend towards higher mortality in case of severe or critical lesions on CT scan (75.0% vs. 25.0%, p = 0.053). Patients who did not improve within 2–4 days after steroid initiation have a bad prognosis and should receive additional anti‐inflammatory drugs. Our data suggest better efficacy of dexamethasone in patients with a low or negative viral load, receiving broad‐spectrum antibiotics. At dexamethasone initiation, a high viral load was associated with higher mortality. Patients without antibiotics were more transferred into the ICU. Mortality was lower when improvement 48 h and 96 h after dexamethasone initiation.
Collapse
Affiliation(s)
- Robin Arcani
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
- Center for Cardiovascular and Nutrition research (C2VN), INRA 1260, INSERM UMR_S 1263Aix‐Marseille University27 Boulevard Jean Moulin13385MarseilleFrance
| | - Raphael Cauchois
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
- Center for Cardiovascular and Nutrition research (C2VN), INRA 1260, INSERM UMR_S 1263Aix‐Marseille University27 Boulevard Jean Moulin13385MarseilleFrance
| | - Pierre Suchon
- Hematology Laboratory, CHU La Timone, Assistance Publique‐Hôpitaux de Marseille (AP‐HM)264 Rue Saint‐Pierre13005MarseilleFrance
| | - Rodolphe Jean
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
| | - Pierre‐André Jarrot
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
- Center for Cardiovascular and Nutrition research (C2VN), INRA 1260, INSERM UMR_S 1263Aix‐Marseille University27 Boulevard Jean Moulin13385MarseilleFrance
| | - Quentin Gomes De Pinho
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
| | - Jean‐Baptiste Dalmas
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
| | - Estelle Jean
- Department of Internal Medicine, CHU La TimoneAssistance Publique‐Hôpitaux de Marseille (AP‐HM)264 Rue Saint‐Pierre13005MarseilleFrance
| | - Baptiste Andre
- Department of Internal Medicine, CHU La TimoneAssistance Publique‐Hôpitaux de Marseille (AP‐HM)264 Rue Saint‐Pierre13005MarseilleFrance
| | - Véronique Veit
- Department of Internal Medicine, CHU La TimoneAssistance Publique‐Hôpitaux de Marseille (AP‐HM)264 Rue Saint‐Pierre13005MarseilleFrance
| | - Marie Koubi
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
| | - Gilles Kaplanski
- Department of Internal Medicine and Clinical Immunology, CHU La ConceptionAssistance Publique‐Hôpitaux de Marseille (AP‐HM)147 Boulevard Baille13005MarseilleFrance
- Center for Cardiovascular and Nutrition research (C2VN), INRA 1260, INSERM UMR_S 1263Aix‐Marseille University27 Boulevard Jean Moulin13385MarseilleFrance
| |
Collapse
|
429
|
Oo A, Zandi K, Shepard C, Bassit LC, Musall K, Goh SL, Cho YJ, Kim DH, Schinazi RF, Kim B. Elimination of Aicardi-Goutières syndrome protein SAMHD1 activates cellular innate immunity and suppresses SARS-CoV-2 replication. J Biol Chem 2022; 298:101635. [PMID: 35085552 PMCID: PMC8786443 DOI: 10.1016/j.jbc.2022.101635] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/23/2022] Open
Abstract
The lack of antiviral innate immune responses during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections is characterized by limited production of interferons (IFNs). One protein associated with Aicardi-Goutières syndrome, SAMHD1, has been shown to negatively regulate the IFN-1 signaling pathway. However, it is unclear whether elevated IFN signaling associated with genetic loss of SAMHD1 would affect SARS-CoV-2 replication. In this study, we established in vitro tissue culture model systems for SARS-CoV-2 and human coronavirus OC43 infections in which SAMHD1 protein expression was absent as a result of CRISPR-Cas9 gene KO or lentiviral viral protein X-mediated proteosomal degradation. We show that both SARS-CoV-2 and human coronavirus OC43 replications were suppressed in SAMHD1 KO 293T and differentiated THP-1 macrophage cell lines. Similarly, when SAMHD1 was degraded by virus-like particles in primary monocyte-derived macrophages, we observed lower levels of SARS-CoV-2 RNA. The loss of SAMHD1 in 293T and differentiated THP-1 cells resulted in upregulated gene expression of IFNs and innate immunity signaling proteins from several pathways, with STAT1 mRNA being the most prominently elevated ones. Furthermore, SARS-CoV-2 replication was significantly increased in both SAMHD1 WT and KO cells when expression and phosphorylation of STAT1 were downregulated by JAK inhibitor baricitinib, which over-rode the activated antiviral innate immunity in the KO cells. This further validates baricitinib as a treatment of SARS-CoV-2-infected patients primarily at the postviral clearance stage. Overall, our tissue culture model systems demonstrated that the elevated innate immune response and IFN activation upon genetic loss of SAMHD1 effectively suppresses SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Adrian Oo
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Keivan Zandi
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Caitlin Shepard
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Leda C Bassit
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Katie Musall
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Shu Ling Goh
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Young-Jae Cho
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Dong-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Kyung-Hee University, Seoul, South Korea
| | - Raymond F Schinazi
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Baek Kim
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA; Center for Drug Discovery, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| |
Collapse
|
430
|
Falavigna M, Belli KC, Barbosa AN, Zavascki AP, Nastri ACDSS, Santana CM, Stein C, Gräf DD, Cadegiani FA, Guimarães HP, Monteiro JT, Ferreira JC, de Azevedo LCP, Magri MMC, Sobreira ML, Dias MBGDS, de Oliveira MS, Corradi MDFDB, Rosa R, Heinzelmann RS, da Silva RM, Junior RB, Cimerman S, Colpani V, Veiga VC, de Carvalho CRR. Brazilian guidelines for the treatment of outpatients with suspected or confirmed COVID-19. A joint guideline of the Brazilian Association of Emergency Medicine (ABRAMEDE), Brazilian Medical Association (AMB), Brazilian Society of Angiology and Vascular Surgery (SBACV), Brazilian Society of Geriatrics and Gerontology (SBGG), Brazilian Society of Infectious Diseases (SBI), Brazilian Society of Family and Community Medicine (SBFMC), and Brazilian Thoracic Society (SBPT). Braz J Infect Dis 2022; 26:102347. [PMID: 35341739 PMCID: PMC8926872 DOI: 10.1016/j.bjid.2022.102347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 11/26/2022] Open
|
431
|
Tanimoto T, Tada S, Fujita S, Hirakawa T, Matsumura M, Isoyama S, Ueno S, Hamai K, Tsuji N, Hirosawa H, Taniguchi T, Okamoto T, Omoto T, Kusunoki S, Maeda H, Ishikawa N. Effect of baricitinib in patients with coronavirus disease 2019 and respiratory failure: a propensity score–matched retrospective cohort study. Respir Investig 2022; 60:418-424. [PMID: 35307364 PMCID: PMC8898743 DOI: 10.1016/j.resinv.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
Abstract
In this retrospective cohort study, we evaluated the efficacy of baricitinib in the treatment of coronavirus disease 2019 (COVID-19). Among 404 adult patients with COVID-19 who were admitted to our hospital between October 23, 2020, and July 31, 2021, 229 patients with respiratory failure were included. Among these, 41 patients in the baricitinib group and 41 patients in the control group were selected by propensity score matching to adjust for background factors. We compared the survival rates of the two groups at 30 and 60 days after admission. The 30-day survival rate was significantly higher in the baricitinib group than in the control group. However, there was no significant difference in 60-day survival in the two groups. Baricitinib may improve the early prognosis of patients with respiratory failure associated with COVID-19. However, efforts should be made to improve the long-term prognosis.
Collapse
|
432
|
Tanaka Y, Luo Y, O'Shea JJ, Nakayamada S. Janus kinase-targeting therapies in rheumatology: a mechanisms-based approach. Nat Rev Rheumatol 2022; 18:133-145. [PMID: 34987201 PMCID: PMC8730299 DOI: 10.1038/s41584-021-00726-8] [Citation(s) in RCA: 282] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
The four Janus kinase (JAK) proteins and seven signal transducer and activator of transcription (STAT) transcription factors mediate intracellular signal transduction downstream of cytokine receptors, which are implicated in the pathology of autoimmune, allergic and inflammatory diseases. Development of targeted small-molecule therapies such as JAK inhibitors, which have varied selective inhibitory profiles, has enabled a paradigm shift in the treatment of diverse disorders. JAK inhibitors suppress intracellular signalling mediated by multiple cytokines involved in the pathological processes of rheumatoid arthritis and many other immune and inflammatory diseases, and therefore have the capacity to target multiple aspects of those diseases. In addition to rheumatoid arthritis, JAK inhibition has potential for treatment of autoimmune diseases including systemic lupus erythematosus, spondyloarthritis, inflammatory bowel disease and alopecia areata, in which stimulation of innate immunity activates adaptive immunity, leading to generation of autoreactive T cells and activation and differentiation of B cells. JAK inhibitors are also effective in the treatment of allergic disorders, such as atopic dermatitis, and can even be used for the COVID-19-related cytokine storm. Mechanism-based treatments targeting JAK-STAT pathways have the potential to provide positive outcomes by minimizing the use of glucocorticoids and/or non-specific immunosuppressants in the treatment of systemic immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.
| | - Yiming Luo
- Vasculitis Translational Research Program Systemic Autoimmunity Branch, National Institute of Arthritis, Musculoskeletal, and Skin Diseases NIH, Bethesda, MD, USA
| | - John J O'Shea
- Molecular Immunology & Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis & Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
433
|
Adas MA, Alveyn E, Cook E, Dey M, Galloway JB, Bechman K. The infection risks of JAK inhibition. Expert Rev Clin Immunol 2022; 18:253-261. [PMID: 34860621 PMCID: PMC8935945 DOI: 10.1080/1744666x.2022.2014323] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Janus Kinase inhibitors (JAKi) have shown to be highly effective in the treatment of immune-mediated inflammatory diseases. As with all immunomodulatory therapies, careful assessment of any treatment-associated infection risk is essential to inform clinical decision-making. AREAS COVERED We summarize current literature on infection rates among the licensed JAKi using published phase II/III trial results, post-licensing and registry data. EXPERT OPINION licensed JAKi show increased risk of infection across the class compared to placebo, most commonly affecting respiratory and urinary tracts, nasopharynx and skin. This risk is dose-dependent. Risks are similar at licensed JAKi doses to that seen with biologic therapies. The risk is compounded by other risk factors for infection, such as age and steroid co-prescription. Herpes zoster reactivation is more common with JAKi compared to other targeted immune modulation, making screening for varicella exposure and vaccination in appropriate cohorts an advisable strategy. Crucially, these small risk increases must be balanced against the known harms (including infection) of uncontrolled autoimmune disease. JAKi are a safe and potentially transformative treatment when used for appropriately selected patients.
Collapse
Affiliation(s)
- Maryam A. Adas
- Centre of Rheumatic diseases, School of Immunology & Microbial Sciences, King’s College London, London, UK
- Department of Physiology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Edward Alveyn
- Centre of Rheumatic diseases, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Emma Cook
- Centre of Rheumatic diseases, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Mrinalini Dey
- Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Countess of Chester Hospital Nhs Foundation Trust, Chester, UK
| | - James B Galloway
- Centre of Rheumatic diseases, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Katie Bechman
- Centre of Rheumatic diseases, School of Immunology & Microbial Sciences, King’s College London, London, UK
| |
Collapse
|
434
|
Heldman MR, Kates OS, Safa K, Kotton CN, Multani A, Georgia SJ, Steinbrink JM, Alexander BD, Blumberg EA, Haydel B, Hemmige V, Hemmersbach-Miller M, La Hoz RM, Moni L, Condor Y, Flores S, Munoz CG, Guitierrez J, Diaz EI, Diaz D, Vianna R, Guerra G, Loebe M, Yabu JM, Kramer KH, Tanna SD, Ison MG, Rakita RM, Malinis M, Azar MM, McCort ME, Singh PP, Velioglu A, Mehta SA, van Duin D, Goldman JD, Lease ED, Wald A, Limaye AP, Fisher CE. Delayed mortality among solid organ transplant recipients hospitalized for COVID-19. Clin Infect Dis 2022; 78:ciac159. [PMID: 35212363 PMCID: PMC9383518 DOI: 10.1093/cid/ciac159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Most studies of solid organ transplant (SOT) recipients with COVID-19 focus on outcomes within one month of illness onset. Delayed mortality in SOT recipients hospitalized for COVID-19 has not been fully examined. METHODS We used data from a multicenter registry to calculate mortality by 90 days following initial SARS-CoV-2 detection in SOT recipients hospitalized for COVID-19 and developed multivariable Cox proportional-hazards models to compare risk factors for death by days 28 and 90. RESULTS Vital status at day 90 was available for 936 of 1117 (84%) SOT recipients hospitalized for COVID-19: 190 of 936 (20%) died by 28 days and an additional 56 of 246 deaths (23%) occurred between days 29 and 90. Factors associated with mortality by day 90 included: age > 65 years [aHR 1.8 (1.3-2.4), p =<0.001], lung transplant (vs. non-lung transplant) [aHR 1.5 (1.0-2.3), p=0.05], heart failure [aHR 1.9 (1.2-2.9), p=0.006], chronic lung disease [aHR 2.3 (1.5-3.6), p<0.001] and body mass index ≥ 30 kg/m 2 [aHR 1.5 (1.1-2.0), p=0.02]. These associations were similar for mortality by day 28. Compared to diagnosis during early 2020 (March 1-June 19, 2020), diagnosis during late 2020 (June 20-December 31, 2020) was associated with lower mortality by day 28 [aHR 0.7 (0.5-1.0, p=0.04] but not by day 90 [aHR 0.9 (0.7-1.3), p=0.61]. CONCLUSIONS In SOT recipients hospitalized for COVID-19, >20% of deaths occurred between 28 and 90 days following SARS-CoV-2 diagnosis. Future investigations should consider extending follow-up duration to 90 days for more complete mortality assessment.
Collapse
Affiliation(s)
- Madeleine R Heldman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Olivia S Kates
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kassem Safa
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Ashrit Multani
- Department of Medicine, David Geffen School of Medicine at the University of California–Los Angeles, Los Angeles, California, USA
| | | | - Julie M Steinbrink
- Division of Infectious Diseases, Duke University, Durham, North Carolina, USA
| | - Barbara D Alexander
- Division of Infectious Diseases, Duke University, Durham, North Carolina, USA
| | - Emily A Blumberg
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brandy Haydel
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vagish Hemmige
- Division of Infectious Disease, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, New York, USA
| | | | - Ricardo M La Hoz
- Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lisset Moni
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Yesabeli Condor
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Sandra Flores
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Carlos G Munoz
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Juan Guitierrez
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Esther I Diaz
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Daniela Diaz
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Rodrigo Vianna
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Giselle Guerra
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Matthias Loebe
- University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Julie M Yabu
- Department of Medicine, David Geffen School of Medicine at the University of California–Los Angeles, Los Angeles, California, USA
| | - Kailey Hughes Kramer
- Transplant Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sajal D Tanna
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael G Ison
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert M Rakita
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Maricar Malinis
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marwan M Azar
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Margaret E McCort
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pooja P Singh
- Division of Nephrology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Arzu Velioglu
- Marmara University, School of Medicine, Department of Internal Medicine, Division of Nephrology, Istanbul, Turkey
| | - Sapna A Mehta
- New York University Langone Transplant Institute, New York, New York, USA
| | - David van Duin
- Division of Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jason D Goldman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Swedish Medical Center, Seattle, Washington, USA
| | - Erika D Lease
- Division of Pulmonology, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Anna Wald
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ajit P Limaye
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Cynthia E Fisher
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
435
|
Significance of Immune Status of SARS-CoV-2 Infected Patients in Determining the Efficacy of Therapeutic Interventions. J Pers Med 2022; 12:jpm12030349. [PMID: 35330349 PMCID: PMC8955701 DOI: 10.3390/jpm12030349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is now being investigated for its distinctive patterns in the course of disease development which can be indicated with miscellaneous immune responses in infected individuals. Besides this series of investigations on the pathophysiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), significant fundamental immunological and physiological processes are indispensable to address clinical markers of COVID-19 disease and essential to identify or design effective therapeutics. Recent developments in the literature suggest that deficiency of type I interferon (IFN) in serum samples can be used to represent a severe progression of COVID-19 disease and can be used as the basis to develop combined immunotherapeutic strategies. Precise control over inflammatory response is a significant aspect of targeting viral infections. This account presents a brief review of the pathophysiological characteristics of the SARS-CoV-2 virus and the understanding of the immune status of infected patients. We further discuss the immune system’s interaction with the SARS-CoV-2 virus and their subsequent involvement of dysfunctional immune responses during the progression of the disease. Finally, we highlight some of the implications of the different approaches applicable in developing promising therapeutic interventions that redirect immunoregulation and viral infection.
Collapse
|
436
|
Zhao L, Li S, Zhong W. Mechanism of Action of Small-Molecule Agents in Ongoing Clinical Trials for SARS-CoV-2: A Review. Front Pharmacol 2022; 13:840639. [PMID: 35281901 PMCID: PMC8916227 DOI: 10.3389/fphar.2022.840639] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 01/18/2023] Open
Abstract
Since the first reports from December 2019, COVID-19 caused an overwhelming global pandemic that has affected 223 countries, seriously endangering public health and creating an urgent need for effective drugs to treat SARS-CoV-2 infection. Currently, there is a lack of safe, effective, and specific therapeutic drugs for COVID-19, with mainly supportive and symptomatic treatments being administered to patients. The preferred option for responding to an outbreak of acute infectious disease is through drug repurposing, saving valuable time that would otherwise be lost in preclinical and clinical research, hastening clinical introduction, and lowering treatment costs. Alternatively, researchers seek to design and discover novel small-molecule candidate drugs targeting the key proteins in the life cycle of SARS-CoV-2 through an in-depth study of the infection mechanism, thus obtaining a number of candidate compounds with favorable antiviral effects in preclinical and clinical settings. There is an urgent need to further elucidate the efficacy and mechanism of action of potential anti-SARS-CoV-2 small-molecule drugs. Herein, we review the candidate small-molecule anti-SARS-CoV-2 drugs in ongoing clinical trials, with a major focus on their mechanisms of action in an attempt to provide useful insight for further research and development of small-molecule compounds against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lei Zhao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Beijing Sunho Pharmaceutical Co., Ltd., Beijing, China
| | - Song Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
437
|
Kita T, Kitamura K. Adrenomedullin Therapy in Moderate to Severe COVID-19. Biomedicines 2022; 10:biomedicines10030533. [PMID: 35327335 PMCID: PMC8945653 DOI: 10.3390/biomedicines10030533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
The 2019 coronavirus (COVID-19) pandemic is still in progress, and a significant number of patients have presented with severe illness. Recently introduced vaccines, antiviral medicines, and antibody formulations can suppress COVID-19 symptoms and decrease the number of patients exhibiting severe disease. However, complete avoidance of severe COVID-19 has not been achieved, and more importantly, there are insufficient methods to treat it. Adrenomedullin (AM) is an endogenous peptide that maintains vascular tone and endothelial barrier function. The AM plasma level is markedly increased during severe inflammatory disorders, such as sepsis, pneumonia, and COVID-19, and is associated with the severity of inflammation and its prognosis. In this study, exogenous AM administration reduced inflammation and related organ damage in rodent models. The results of this study strongly suggest that AM could be an alternative therapy in severe inflammation disorders, including COVID-19. We have previously developed an AM formulation to treat inflammatory bowel disease and are currently conducting an investigator-initiated phase 2a trial for moderate to severe COVID-19 using the same formulation. This review presents the basal AM information and the most recent translational AM/COVID-19 study.
Collapse
|
438
|
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
|
439
|
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
|
440
|
Teixeira JP, Barone S, Zahedi K, Soleimani M. Kidney Injury in COVID-19: Epidemiology, Molecular Mechanisms and Potential Therapeutic Targets. Int J Mol Sci 2022; 23:2242. [PMID: 35216358 PMCID: PMC8877127 DOI: 10.3390/ijms23042242] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/08/2023] Open
Abstract
As of December 2021, SARS-CoV-2 had caused over 250 million infections and 5 million deaths worldwide. Furthermore, despite the development of highly effective vaccines, novel variants of SARS-CoV-2 continue to sustain the pandemic, and the search for effective therapies for COVID-19 remains as urgent as ever. Though the primary manifestation of COVID-19 is pneumonia, the disease can affect multiple organs, including the kidneys, with acute kidney injury (AKI) being among the most common extrapulmonary manifestations of severe COVID-19. In this article, we start by reflecting on the epidemiology of kidney disease in COVID-19, which overwhelmingly demonstrates that AKI is common in COVID-19 and is strongly associated with poor outcomes. We also present emerging data showing that COVID-19 may result in long-term renal impairment and delve into the ongoing debate about whether AKI in COVID-19 is mediated by direct viral injury. Next, we focus on the molecular pathogenesis of SARS-CoV-2 infection by both reviewing previously published data and presenting some novel data on the mechanisms of cellular viral entry. Finally, we relate these molecular mechanisms to a series of therapies currently under investigation and propose additional novel therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- J. Pedro Teixeira
- Department of Internal Medicine, Division of Nephrology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.B.); (K.Z.)
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Sharon Barone
- Department of Internal Medicine, Division of Nephrology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.B.); (K.Z.)
- Research/Medicine Services, New Mexico Veterans Healthcare Medical Center, Albuquerque, NM 87108, USA
| | - Kamyar Zahedi
- Department of Internal Medicine, Division of Nephrology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.B.); (K.Z.)
- Research/Medicine Services, New Mexico Veterans Healthcare Medical Center, Albuquerque, NM 87108, USA
| | - Manoocher Soleimani
- Department of Internal Medicine, Division of Nephrology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.B.); (K.Z.)
- Research/Medicine Services, New Mexico Veterans Healthcare Medical Center, Albuquerque, NM 87108, USA
| |
Collapse
|
441
|
van den Berg K, Glatt TN, Vermeulen M, Little F, Swanevelder R, Barrett C, Court R, Bremer M, Nyoni C, Swarts A, Mmenu C, Crede T, Kritzinger G, Naude J, Szymanski P, Cowley J, Moyo-Gwete T, Moore PL, Black J, Singh J, Bhiman JN, Baijnath P, Mody P, Malherbe J, Potgieter S, van Vuuren C, Maasdorp S, Wilkinson RJ, Louw VJ, Wasserman S. Convalescent plasma in the treatment of moderate to severe COVID-19 pneumonia: a randomized controlled trial (PROTECT-Patient Trial). Sci Rep 2022; 12:2552. [PMID: 35169169 PMCID: PMC8847351 DOI: 10.1038/s41598-022-06221-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/24/2022] [Indexed: 01/08/2023] Open
Abstract
There is a need for effective therapy for COVID-19 pneumonia. Convalescent plasma has antiviral activity and early observational studies suggested benefit in reducing COVID-19 severity. We investigated the safety and efficacy of convalescent plasma in hospitalized patients with COVID-19 in a population with a high HIV prevalence and where few therapeutic options were available. We performed a double-blinded, multicenter, randomized controlled trial in one private and three public sector hospitals in South Africa. Adult participants with COVID-19 pneumonia requiring non-invasive oxygen were randomized 1:1 to receive a single transfusion of 200 mL of either convalescent plasma or 0.9% saline solution. The primary outcome measure was hospital discharge and/or improvement of ≥ 2 points on the World Health Organisation Blueprint Ordinal Scale for Clinical Improvement by day 28 of enrolment. The trial was stopped early for futility by the Data and Safety Monitoring Board. 103 participants, including 21 HIV positive individuals, were randomized at the time of premature trial termination: 52 in the convalescent plasma and 51 in the placebo group. The primary outcome occurred in 31 participants in the convalescent plasma group and and 32 participants in the placebo group (relative risk 1.03 (95% CI 0.77 to 1.38). Two grade 1 transfusion-related adverse events occurred. Participants who improved clinically received convalescent plasma with a higher median anti-SARS-CoV-2 neutralizing antibody titre compared with those who did not (298 versus 205 AU/mL). Our study contributes additional evidence for recommendations against the use of convalescent plasma for COVID-19 pneumonia. Safety and feasibility in this population supports future investigation for other indications.
Collapse
Affiliation(s)
- Karin van den Berg
- Medical Division, Translational Research Department, South African National Blood Service, 1 Constantia Blvd, Roodepoort, 1715, South Africa.
- Division of Clinical Haematology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa.
- Division of Clinical Haematology, Department of Internal Medicine, University of the Free State, Bloemfontein, South Africa.
| | - Tanya Nadia Glatt
- Medical Division, Translational Research Department, South African National Blood Service, 1 Constantia Blvd, Roodepoort, 1715, South Africa
| | - Marion Vermeulen
- Division of Clinical Haematology, Department of Internal Medicine, University of the Free State, Bloemfontein, South Africa
- Operations Division, Operations Testing Department, South African National Blood Service, Roodepoort, South Africa
| | - Francesca Little
- Department of Statistical Sciences, University of Cape Town, Observatory, South Africa
| | - Ronel Swanevelder
- Medical Division, Translational Research Department, South African National Blood Service, 1 Constantia Blvd, Roodepoort, 1715, South Africa
| | - Claire Barrett
- School of Clinical Medicine, University of the Free State, Bloemfontein, South Africa
| | - Richard Court
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Marise Bremer
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Cynthia Nyoni
- Medical Division, Translational Research Department, South African National Blood Service, 1 Constantia Blvd, Roodepoort, 1715, South Africa
| | - Avril Swarts
- Medical Division, Translational Research Department, South African National Blood Service, 1 Constantia Blvd, Roodepoort, 1715, South Africa
| | - Cordelia Mmenu
- Operations Division, Operations Testing Department, South African National Blood Service, Roodepoort, South Africa
| | - Thomas Crede
- Mitchells Plain Hospital and the University of Cape Town's Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Gerdien Kritzinger
- Division of Clinical Haematology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Jonathan Naude
- Mitchells Plain Hospital and the University of Cape Town's Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Patryk Szymanski
- Mitchells Plain Hospital and the University of Cape Town's Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - James Cowley
- Operations Division, Processing Department, South African National Blood Service, Roodepoort, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L Moore
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - John Black
- Department of Medicine, Walter Sisulu University, Livingstone Hospital, Gqeberha, South Africa
| | - Jaimendra Singh
- Capital Haematology Hospital and Bone Marrow Transplant Unit, Durban, South Africa
| | - Jinal N Bhiman
- Centre for Respiratory Diseases and Meningitis (CRDM), National Institute for Communicable Diseases, Johannesburg, South Africa
- Department of Virology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Priyesh Mody
- Life Westville Hospital, Westville, South Africa
| | - Jacques Malherbe
- School of Clinical Medicine, University of the Free State, Bloemfontein, South Africa
| | - Samantha Potgieter
- Division of Infectious Diseases, Department of Internal Medicine, University of the Free State, Bloemfontein, South Africa
| | - Cloete van Vuuren
- 3 Military Hospital and Department of Internal Medicine, University of the Free State, Bloemfontein, South Africa
| | - Shaun Maasdorp
- Pulmonology and Critical Care, Universitas Academic Hospital and Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Francis Crick Institute, London, NW1 1AT, UK
- Department of Infectious Diseases, Imperial College London, London, W12 0NN, UK
| | - Vernon J Louw
- Division of Clinical Haematology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Sean Wasserman
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Observatory, South Africa
| |
Collapse
|
442
|
Mandell LA, Zhanel GG, Rotstein C, Muscedere J, Loeb M, Johnstone J. Community-Acquired Pneumonia in Canada During COVID-19. Open Forum Infect Dis 2022; 9:ofac043. [PMID: 35211634 PMCID: PMC8863085 DOI: 10.1093/ofid/ofac043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/03/2022] [Indexed: 12/15/2022] Open
Abstract
Dealing with coronavirus disease 2019 (COVID-19) has been a monumental test of medical skills and resources worldwide. The management of community-acquired pneumonia (CAP) can at times be difficult, but treating CAP in the setting of COVID-19 can be particularly trying and confusing and raises a number of challenging questions relating to etiology, diagnosis, and treatment. This article is based on the authors’ experiences and presents an overview of how CAP during COVID-19 is handled in Canada. We touch on the issues of microbial etiology in patients with CAP in the setting of COVID-19 as well as diagnostic, site of care, and treatment approaches. Published guidelines are the basis of management of CAP and are discussed in the context of Canadian data. We also outline the usual treatment approaches to COVID-19, particularly in patients who have been hospitalized.
Collapse
Affiliation(s)
- L A Mandell
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - G G Zhanel
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - C Rotstein
- Department of Medicine, University of Toronto, Toronto, Canada
| | - J Muscedere
- Department of Critical Care Medicine, Queens University, Kingston, Canada
| | - M Loeb
- Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - J Johnstone
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
443
|
Elkrief A, Wu JT, Jani C, Enriquez KT, Glover M, Shah MR, Shaikh HG, Beeghly-Fadiel A, French B, Jhawar SR, Johnson DB, McKay RR, Rivera DR, Reuben DY, Shah S, Tinianov SL, Vinh DC, Mishra S, Warner JL. Learning through a Pandemic: The Current State of Knowledge on COVID-19 and Cancer. Cancer Discov 2022; 12:303-330. [PMID: 34893494 PMCID: PMC8831477 DOI: 10.1158/2159-8290.cd-21-1368] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/26/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has left patients with current or past history of cancer facing disparate consequences at every stage of the cancer trajectory. This comprehensive review offers a landscape analysis of the current state of the literature on COVID-19 and cancer, including the immune response to COVID-19, risk factors for severe disease, and impact of anticancer therapies. We also review the latest data on treatment of COVID-19 and vaccination safety and efficacy in patients with cancer, as well as the impact of the pandemic on cancer care, including the urgent need for rapid evidence generation and real-world study designs. SIGNIFICANCE: Patients with cancer have faced severe consequences at every stage of the cancer journey due to the COVID-19 pandemic. This comprehensive review offers a landscape analysis of the current state of the field regarding COVID-19 and cancer. We cover the immune response, risk factors for severe disease, and implications for vaccination in patients with cancer, as well as the impact of the COVID-19 pandemic on cancer care delivery. Overall, this review provides an in-depth summary of the key issues facing patients with cancer during this unprecedented health crisis.
Collapse
Affiliation(s)
- Arielle Elkrief
- Division of Medical Oncology (Department of Medicine), McGill University Health Centre, Montreal, Quebec, Canada
| | - Julie T Wu
- Stanford University, Palo Alto, California
| | - Chinmay Jani
- Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts
| | - Kyle T Enriquez
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Mansi R Shah
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | | | | | | - Sachin R Jhawar
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Rana R McKay
- University of California San Diego, San Diego, California
| | - Donna R Rivera
- Division of Cancer Control and Population Services, National Cancer Institute, Rockville, Maryland
| | - Daniel Y Reuben
- Medical University of South Carolina, Charleston, South Carolina
| | - Surbhi Shah
- Hematology and Oncology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Stacey L Tinianov
- Advocates for Collaborative Education, UCSF Breast Science Advocacy Core, San Francisco, California
| | - Donald Cuong Vinh
- Division of Infectious Diseases (Department of Medicine), Divisions of Medical Microbiology and of Molecular Diagnostics (OptiLab), McGill University Health Centre, Montreal, Quebec, Canada
| | - Sanjay Mishra
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeremy L Warner
- Vanderbilt University Medical Center, Nashville, Tennessee.
- Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
444
|
Lanthier L. Chez les adultes atteints de la COVID-19 d’intensité légère à modérée, non vaccinés et à risque de complications, est-ce que le nirmatrelvir/ritonavir diminue le risque d’hospitalisation reliée à la COVID-19 ou la mortalité toute cause tout en étant sécuritaire? Rev Med Interne 2022; 43:270-271. [PMID: 35279350 PMCID: PMC8882428 DOI: 10.1016/j.revmed.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/15/2022]
|
445
|
McConnell MJ, Kondo R, Kawaguchi N, Iwakiri Y. Covid-19 and Liver Injury: Role of Inflammatory Endotheliopathy, Platelet Dysfunction, and Thrombosis. Hepatol Commun 2022; 6:255-269. [PMID: 34658172 PMCID: PMC8652692 DOI: 10.1002/hep4.1843] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/18/2021] [Accepted: 10/10/2021] [Indexed: 02/06/2023] Open
Abstract
Liver injury, characterized predominantly by elevated aspartate aminotransferase and alanine aminotransferase, is a common feature of coronavirus disease 2019 (COVID-19) symptoms caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). Additionally, SARS-CoV-2 infection is associated with acute-on-chronic liver failure in patients with cirrhosis and has a notably elevated mortality in patients with alcohol-related liver disease compared to other etiologies. Direct viral infection of the liver with SARS-CoV-2 remains controversial, and alternative pathophysiologic explanations for its hepatic effects are an area of active investigation. In this review, we discuss the effects of SARS-CoV-2 and the inflammatory environment it creates on endothelial cells and platelets more generally and then with a hepatic focus. In doing this, we present vascular inflammation and thrombosis as a potential mechanism of liver injury and liver-related complications in COVID-19.
Collapse
Affiliation(s)
- Matthew J. McConnell
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Reiichiro Kondo
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of PathologyKurume University School of MedicineKurumeJapan
| | - Nao Kawaguchi
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Yasuko Iwakiri
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
446
|
Chen JT, Ostermann M. Review of Anti-inflammatory and Anti-viral therapeutics for hospitalized patients infected with SARS-CoV-2. Crit Care Clin 2022; 38:587-600. [PMID: 35667745 PMCID: PMC8841241 DOI: 10.1016/j.ccc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
447
|
Bell TD. COVID-19 in the Critically Ill Patient. Infect Dis Clin North Am 2022; 36:365-377. [PMID: 35636905 PMCID: PMC8847096 DOI: 10.1016/j.idc.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
448
|
Jing H, Zuo N, Novakovic VA, Shi J. The Central Role of Extracellular Vesicles in the Mechanisms of Thrombosis in COVID-19 Patients With Cancer and Therapeutic Strategies. Front Cell Dev Biol 2022; 9:792335. [PMID: 35096822 PMCID: PMC8790316 DOI: 10.3389/fcell.2021.792335] [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: 10/10/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer patients have increased SARS-CoV-2 susceptibility and are prone to developing severe COVID-19 infections. The incidence of venous thrombosis is approximately 20% in COVID-19 patients with cancer. It has been suggested that thrombus formation has been suggested to correlate with severe clinical manifestations, mortality, and sequelae. In this review, we primarily elaborate on the pathophysiological mechanisms of thrombosis in COVID-19 patients with cancer, emphasize the role of microparticles (MPs) and phosphatidylserine (PS) in coagulation, and propose an antithrombotic strategy. The coagulation mechanisms of COVID-19 and cancer synergistically amplify the coagulation cascade, and collectively promotes pulmonary microvascular occlusion. During systemic coagulation, the virus activates immune cells to release abundant proinflammatory cytokines, referred to as cytokine storm, resulting in the apoptosis of tumor and blood cells and subsequent MPs release. Additionally, we highlight that tumor cells contribute to MPs and coagulation by apoptosis owing to insufficient blood supply. A positive feedback loop of cytokines storm and MPs storm promotes microvascular coagulation storm, leading to microthrombi formation and inadequate blood perfusion. Microthrombi-damaged endothelial cells (ECs), tumor, and blood cells further aggravate the apoptosis of the cells and facilitate MPs storm. PS, especially on MPs, plays a pivotal role in the blood coagulation process, contributing to clot initiation, amplification, and propagation. Since coagulation is a common pathway of COVID-19 and cancer, and associated with mortality, patients would benefit from antithrombotic therapy. The above results lead us to assert that early stage antithrombotic therapy is optimal. This strategy is likely to maintain blood flow patency contributing to viral clearance, attenuating the formation of cytokines and MPs storm, maintaining oxygen saturation, and avoiding the progress of the disease.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Nan Zuo
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China.,Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
449
|
Kim S. COVID-19 Drug Development. J Microbiol Biotechnol 2022; 32:1-5. [PMID: 34866128 PMCID: PMC9628783 DOI: 10.4014/jmb.2110.10029] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022]
Abstract
Diagnostics, vaccines, and drugs are indispensable tools and control measures employed to overcome infectious diseases such as coronavirus disease 2019 (COVID-19). Diagnostic tools based on RT-PCR were developed early in the COVID-19 pandemic and were urgently required for quarantine (testing, tracing and isolation). Vaccines such as mRNA vaccines and virus-vectored vaccines were also successfully developed using new platform technologies within one year after identifying severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the causative agent of COVID-19. Drug development has been conducted in various ways including drug repurposing, convalescent plasma therapy, and monoclonal antibody development. Among the above efforts, this review examines COVID-19 drug development along with the related and upcoming challenges.
Collapse
Affiliation(s)
- Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea
| |
Collapse
|
450
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a respiratory virus that gains entry via angiotensin-converting enzyme 2 (ACE2) within airway epithelium. Patients exhibit a spectrum of respiratory symptoms from asymptomatic to respiratory failure. Patient factors including obesity, tobacco use, and black race are all associated with increased ACE2 expression and may contribute to increased complications. Consolidation and ground-glass opacities on chest imaging are typical but not specific for coronavirus disease 2019 (COVID-19). Venous thromboembolism occurs infrequently when prophylactic anticoagulation is provided. However, capillary microthrombosis is nearly ubiquitous, suggesting that it contributes to hypoxemia. Remdesivir and glucocorticoids may benefit some hospitalized patients. Many of those afflicted remain symptomatic two weeks following diagnosis and continue to require health care. Total lung capacity, diffusion capacity, and maximal oxygen consumption may be reduced for months in some survivors. Lung transplant offers chronically critically ill patients new hope, and this option may have increasing potential for outpatients with COVID-19-associated fibrosis.
Collapse
Affiliation(s)
- Kevin C Doerschug
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; ,
| | - Gregory A Schmidt
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; ,
| |
Collapse
|