151
|
Mitsushima S, Horiguchi H, Taniguchi K. Alpha and Delta variants and vaccination effectiveness against severity in COVID-19 inpatients based on medical claims in Japan. GHM OPEN 2023; 3:28-36. [PMID: 40143836 PMCID: PMC11933968 DOI: 10.35772/ghmo.2023.01005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/12/2023] [Accepted: 07/13/2023] [Indexed: 03/28/2025]
Abstract
Some mutated strains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presumably have high infectivity and pathogenicity. Using Japanese medical claims data, we assessed the pathogenicity of Alpha and Delta variants and vaccine effectiveness by severity. Inpatient records from the Medical Information Analysis Databank for the National Hospital Organization were used. Severity was defined as the proportion of inpatients using respiratory ventilators among inpatients with oxygen administration. We regressed severity and fatality on the proportion of patients with Alpha or Delta variant and on vaccination coverage, while allowing for some lag to reflect development from infection to hospitalization. We also examined results obtained when using data for all new inpatients, instead of inpatients with oxygen administration, as the denominator for severity. Estimation results were better when using severity defined by inpatients with oxygen administration as the denominator than when using all new inpatients. Especially for severity measures for inpatients 65 years old or older with oxygen administration, we confirmed an association of vaccination with lower severity and an association of Delta variant infection with high severity. Vaccines were most effective for people 65 years old or older. The age distributions of inpatients and confirmed patients were greater than for people younger than 65 years old. Vaccination reduced severity and fatality and Alpha and Delta variants might increase severity and fatality among inpatients 65 years old or older receiving oxygen therapy.
Collapse
Affiliation(s)
- Shingo Mitsushima
- Center for Field Epidemic Intelligence, Research and Professional Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiromasa Horiguchi
- Department of Clinical Data Management and Research, Clinical Research Center, National Hospital Organization Headquarters, Tokyo, Japan
| | - Kiyosu Taniguchi
- National Hospital Organization Mie National Hospital, Mie, Japan
- The Tokyo Foundation for Policy Research, Tokyo, Japan
| |
Collapse
|
152
|
Guérin E, Belin L, Franchineau G, Le Guennec L, Hajage D, Diallo MH, Frapard T, Le Fèvre L, Luyt CE, Combes A, Germain S, Hayon J, Asfar P, Bréchot N. FX06 to rescue SARS-CoV-2-induced acute respiratory distress syndrome: a randomized clinical trial. Crit Care 2023; 27:331. [PMID: 37641136 PMCID: PMC10463389 DOI: 10.1186/s13054-023-04616-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Vascular leakage is a major feature of acute respiratory distress syndrome (ARDS). We aimed to evaluate the efficacy of FX06, a drug under development that stabilizes interendothelial cell junctions, at reducing vascular leakage during SARS-CoV-2-induced ARDS. METHODS This multicenter, double-blinded, randomized trial included adults with COVID-19-associated ARDS who had received invasive mechanical ventilation for < 5 days and were randomized to receive either intravenous FX06 (400 mg/d, for 5 days) or its vehicle as placebo. The primary endpoint was the lowering-from day 1 to day 7-of the transpulmonary thermodilution-derived extravascular lung-water index (EVLWi). RESULTS Twenty-five patients were randomized to receive FX06 and 24 the placebo. Although EVLWi was elevated at baseline (median [IQR] 15.6 mL/kg [13.5; 18.5]), its declines from day 1 to day 7 were comparable for FX06 recipients and controls (respectively, - 1.9 [- 3.3; - 0.5] vs. - 0.8 [- 5.5; - 1.1] mL/kg; estimated effect - 0.8 [- 3.1; + 2.4], p = 0.51). Cardiac indexes, pulmonary vascular permeability indexes, and fluid balances were also comparable, as were PaO2/FiO2 ratios and durations of mechanical ventilation. Adverse event rates were similar for the 2 groups, although more FX06 recipients developed ventilator-associated pneumonia (16/25 (64%) vs. 6/24 (24%), p = 0.009). CONCLUSIONS In this unique-dosing-regimen study, FX06 did not lower SARS-CoV-2-induced pulmonary vascular leakage. Future investigations will need to evaluate its efficacy at earlier times during the disease or using other regimens. Trial registration NCT04618042. Registered 5 November 2020.
Collapse
Affiliation(s)
- Emmanuelle Guérin
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France
- Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), Université PSL, Paris, France
| | - Lisa Belin
- Sorbonne Université, INSERM, Institut Pierre Louis d' Epidémiologie et de Santé Publique, AP-HP, Hôpital Pitié Salpêtrière, Département de Santé Publique, Unité de Recherche Clinique PSL-CFX, CIC-1901, 75013, Paris, France
| | - Guillaume Franchineau
- Intensive Care Unit, Centre Hospitalier Intercommunal de Poissy-Saint-Germain-en-Laye, Poissy, France
- INSERM U1018, Centre de Recherche en Épidémiologie Et Santé Des Populations (CESP), Equipe "Rein et Cœur", Université Paris Saclay, Villejuif, France
| | - Loïc Le Guennec
- Médecine Intensive-Réanimation Neurologique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
- Sorbonne Universités, Paris, France
| | - David Hajage
- Sorbonne Université, INSERM, Institut Pierre Louis d' Epidémiologie et de Santé Publique, AP-HP, Hôpital Pitié Salpêtrière, Département de Santé Publique, Unité de Recherche Clinique PSL-CFX, CIC-1901, 75013, Paris, France
| | - Mamadou Hassimiou Diallo
- Sorbonne Université, INSERM, Institut Pierre Louis d' Epidémiologie et de Santé Publique, AP-HP, Hôpital Pitié Salpêtrière, Département de Santé Publique, Unité de Recherche Clinique PSL-CFX, CIC-1901, 75013, Paris, France
| | - Thomas Frapard
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France
- Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), Université PSL, Paris, France
| | - Lucie Le Fèvre
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France
- Sorbonne Université INSERM-UMRS 1166, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Alain Combes
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France
- Sorbonne Université INSERM-UMRS 1166, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), Université PSL, Paris, France
| | - Jan Hayon
- Intensive Care Unit, Centre Hospitalier Intercommunal de Poissy-Saint-Germain-en-Laye, Poissy, France
| | - Pierre Asfar
- Service de Médecine Intensive-Réanimation et Médecine Hyperbare, Centre Universitaire Hospitalier d'Angers, Angers, France
| | - Nicolas Bréchot
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Paris, France.
- Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), Université PSL, Paris, France.
- Service de Médecine Intensive-Réanimation, Hôpital Européen Georges-Pompidou, APHP, Paris, France.
- Université Paris Cité, Paris, France.
| |
Collapse
|
153
|
Hawkins BK, Walker SD, Shorman MA. Missed Opportunities for Antifungal Stewardship during the COVID-19 Era. Antibiotics (Basel) 2023; 12:1352. [PMID: 37760649 PMCID: PMC10526074 DOI: 10.3390/antibiotics12091352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 09/29/2023] Open
Abstract
Significant increases in antibacterial use were observed during the COVID-19 pandemic. However, subsequent analyses found this increase in antibiotic use to be excessive in comparison with the relatively low rates of bacterial coinfection. Although patients who are critically ill with COVID-19 may be at an increased risk for pulmonary aspergillosis, antifungal use in these populations remained underreported, particularly in later phases of the pandemic. This single-center, population-level cohort analysis compares the monthly use rates of mold-active antifungal drugs in the medical intensive care unit during April 2019-March 2020 (baseline) with those during April 2020-November 2022. The antifungal drugs included in the analysis were liposomal amphotericin B, anidulafungin, isavuconazonium, posaconazole, and voriconazole. We found that during 2020-2022, the usage of antifungal drugs was not significantly different from baseline for all included agents except isavuconazonium, which was used significantly more (p = 0.009). There were no changes in diagnostic modalities between the two time periods. The reported prevalence of and mortality from COVID-19-associated pulmonary aspergillosis (CAPA) may have resulted in higher rates of prescribing antifungal drugs for critically ill patients with COVID-19. Antimicrobial stewardship programs should develop and apply tools to facilitate more effective and appropriate antifungal use.
Collapse
Affiliation(s)
- Brandon K. Hawkins
- College of Pharmacy, University of Tennessee Health Science Center, Knoxville, TN 37920, USA
| | - Samantha D. Walker
- Department of Pharmacy, University of Tennessee Medical Center, Knoxville, TN 37920, USA
| | - Mahmoud A. Shorman
- Division of Infectious Diseases, University of Tennessee Medical Center, Knoxville, TN 37920, USA
| |
Collapse
|
154
|
Tam D, Lorenzo-Leal AC, Hernández LR, Bach H. Targeting SARS-CoV-2 Non-Structural Proteins. Int J Mol Sci 2023; 24:13002. [PMID: 37629182 PMCID: PMC10455537 DOI: 10.3390/ijms241613002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped respiratory β coronavirus that causes coronavirus disease (COVID-19), leading to a deadly pandemic that has claimed millions of lives worldwide. Like other coronaviruses, the SARS-CoV-2 genome also codes for non-structural proteins (NSPs). These NSPs are found within open reading frame 1a (ORF1a) and open reading frame 1ab (ORF1ab) of the SARS-CoV-2 genome and encode NSP1 to NSP11 and NSP12 to NSP16, respectively. This study aimed to collect the available literature regarding NSP inhibitors. In addition, we searched the natural product database looking for similar structures. The results showed that similar structures could be tested as potential inhibitors of the NSPs.
Collapse
Affiliation(s)
- Donald Tam
- Division of Infectious Disease, Department of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.T.); (A.C.L.-L.)
| | - Ana C. Lorenzo-Leal
- Division of Infectious Disease, Department of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.T.); (A.C.L.-L.)
| | - Luis Ricardo Hernández
- Laboratorio de Investigación Fitoquímica, Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Ex Hacienda Sta. Catarina Mártir S/N, San Andrés Cholula 72810, Mexico;
| | - Horacio Bach
- Division of Infectious Disease, Department of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.T.); (A.C.L.-L.)
| |
Collapse
|
155
|
Pawar VA, Tyagi A, Verma C, Sharma KP, Ansari S, Mani I, Srivastva SK, Shukla PK, Kumar A, Kumar V. Unlocking therapeutic potential: integration of drug repurposing and immunotherapy for various disease targeting. Am J Transl Res 2023; 15:4984-5006. [PMID: 37692967 PMCID: PMC10492070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023]
Abstract
Drug repurposing, also known as drug repositioning, entails the application of pre-approved or formerly assessed drugs having potentially functional therapeutic amalgams for curing various disorders or disease conditions distinctive from their original remedial indication. It has surfaced as a substitute for the development of drugs for treating cancer, cardiovascular diseases, neurodegenerative disorders, and various infectious diseases like Covid-19. Although the earlier lines of findings in this area were serendipitous, recent advancements are based on patient centered approaches following systematic, translational, drug targeting practices that explore pathophysiological ailment mechanisms. The presence of definite information and numerous records with respect to beneficial properties, harmfulness, and pharmacologic characteristics of repurposed drugs increase the chances of approval in the clinical trial stages. The last few years have showcased the successful emergence of repurposed drug immunotherapy in treating various diseases. In this light, the present review emphasises on incorporation of drug repositioning with Immunotherapy targeted for several disorders.
Collapse
Affiliation(s)
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied ScienceDelhi 110054, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State UniversityColumbus, Ohio 43201, USA
| | - Kanti Prakash Sharma
- Department of Nutrition Biology, Central University of HaryanaMahendragarh 123029, India
| | - Sekhu Ansari
- Division of Pathology, Cincinnati Children’s Hospital Medical CenterCincinnati, Ohio 45229, USA
| | - Indra Mani
- Department of Microbiology, Gargi College, University of DelhiNew Delhi 110049, India
| | | | - Pradeep Kumar Shukla
- Department of Biological Sciences, Faculty of Science, Sam Higginbottom University of Agriculture, Technology of SciencePrayagraj 211007, UP, India
| | - Antresh Kumar
- Department of Biochemistry, Central University of HaryanaMahendergarh 123031, Haryana, India
| | - Vinay Kumar
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, Ohio 43210, USA
| |
Collapse
|
156
|
Barbosa AN, Chebabo A, Starling C, Pérez C, Cunha CA, de Luna D, Nunes EP, Zambrano G, Ferreira JC, Croda J, Falavigna M, Gomes-da-Silva MM, Thormann M, Cimerman S, Parahiba SM, Tanni S, Bernardo WM, Rodriguez-Morales AJ. Pan-American Guidelines for the treatment of SARS-CoV-2/COVID-19: a joint evidence-based guideline of the Brazilian Society of Infectious Diseases (SBI) and the Pan-American Association of Infectious Diseases (API). Ann Clin Microbiol Antimicrob 2023; 22:67. [PMID: 37550690 PMCID: PMC10408214 DOI: 10.1186/s12941-023-00623-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Since the beginning of the COVID-19 pandemic, therapeutic options for treating COVID-19 have been investigated at different stages of clinical manifestations. Considering the particular impact of COVID-19 in the Americas, this document aims to present recommendations for the pharmacological treatment of COVID-19 specific to this population. METHODS Fifteen experts, members of the Brazilian Society of Infectious Diseases (SBI) and the Pan-American Association of Infectious Diseases (API) make up the panel responsible for developing this guideline. Questions were formulated regarding prophylaxis and treatment of COVID-19 in outpatient and inpatient settings. The outcomes considered in decision-making were mortality, hospitalisation, need for mechanical ventilation, symptomatic COVID-19 episodes, and adverse events. In addition, a systematic review of randomised controlled trials was conducted. The quality of evidence assessment and guideline development process followed the GRADE system. RESULTS Nine technologies were evaluated, and ten recommendations were made, including the use of tixagevimab + cilgavimab in the prophylaxis of COVID-19, tixagevimab + cilgavimab, molnupiravir, nirmatrelvir + ritonavir, and remdesivir in the treatment of outpatients, and remdesivir, baricitinib, and tocilizumab in the treatment of hospitalised patients with severe COVID-19. The use of hydroxychloroquine or chloroquine and ivermectin was discouraged. CONCLUSION This guideline provides recommendations for treating patients in the Americas following the principles of evidence-based medicine. The recommendations present a set of drugs that have proven effective in the prophylaxis and treatment of COVID-19, emphasising the strong recommendation for the use of nirmatrelvir/ritonavir in outpatients as the lack of benefit from the use of hydroxychloroquine and ivermectin.
Collapse
Affiliation(s)
- Alexandre Naime Barbosa
- Infectious Diseases Department - Botucatu School of Medicine - UNESP, Av. Prof. Mário R. G. Montenegro, s/n, Botucatu, SP, CEP 18.618-687, Brazil.
- Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubiao Jr, s/n, Botucatu, SP, CEP 18618-970, Brazil.
| | - Alberto Chebabo
- Universidade Federal do Rio de Janeiro, Avenida Professor Rodolpho Paulo Rocco, 255, 50. Andar, Rio de Janeiro, RJ, CEP 21941-913, Brazil
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
| | - Carlos Starling
- Sociedade Mineira de Infectologia - SMI, Avenida João Pinheiro, 161, Belo Horizonte, MG, CEP 30130-180, Brazil
| | - Clevy Pérez
- Universidad Autónoma de Santo Domingo (UASD), Avenida Simón Bolívar, 902, Santo Domingo, 10108, República Dominicana
| | - Clóvis Arns Cunha
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
- Universidade Federal do Paraná, Rua XV de Novembro, 1299, Curitiba, PR, CEP 80060-000, Brazil
| | - David de Luna
- Comisión Nacional de Arbitraje Médico, C Mitla, 250, Ciudad de México, 03020, México
| | - Estevão Portela Nunes
- Instituto Nacional de Infectologia (INI), Fiocruz, Avenida Brasil, 4365, Rio de Janeiro, RJ, CEP 21040-360, Brazil
| | - Gabriela Zambrano
- Faculty of Medicine, Department of Infectious Diseases, Universidad Central del Ecuador, Quito, Ecuador
- Pontificia Universidad Católica del Ecuador, Facultad de Medicina, Posgrado de Medicina Interna, Quito, Ecuador
| | - Juliana Carvalho Ferreira
- Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, Avenida Dr. Enéas Carvalho de Aguiar, 44, São Paulo, SP, CEP 05403-900, Brazil
- Intensive Care Unit, AC Camargo Cancer Center, Rua Prof. Antônio Prudente, 211, São Paulo, SP, CEP 01509-001, Brazil
| | - Julio Croda
- Oswaldo Cruz Foundation, Avenida Costa e Silva, s/n, Cidade Universitária, Campo Grande, MS, CEP 79070-900, Brazil
| | - Maicon Falavigna
- HTAnalyze Consulting and Training, Rua João Abbott, 109, Porto Alegre, RS, CEP 90460-150, Brazil
| | - Monica Maria Gomes-da-Silva
- Infectious Disease Control Service, Clinical Hospital, Universidade Federal Do Paraná, Rua General Carneiro, 181, Curitiba, PR, CEP 80060-900, Brazil
| | - Monica Thormann
- Hospital Salvador Bienvenido Gautier, Calle Alexander Fleming, 177, Santo Domingo, 10514, Dominican Republic
| | - Sergio Cimerman
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
- Institute of Infectious Diseases Emilio Ribas, Avenida Dr. Arnaldo, 165, São Paulo, SP, CEP 05402-000, Brazil
| | - Suena Medeiros Parahiba
- HTAnalyze Consulting and Training, Rua João Abbott, 109, Porto Alegre, RS, CEP 90460-150, Brazil
| | - Suzana Tanni
- Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubiao Jr, s/n, Botucatu, SP, CEP 18618-970, Brazil
| | - Wanderley Marques Bernardo
- Medical Education Development Center (CEDEM) of Medical Faculty of São Paulo University (FMUSP), São Paulo, SP, Brazil
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de Las Américas-Institución Universitaria Visión de Las Américas, 660003, Pereira, Risaralda, Colombia.
- Clinical Epidemiology and Biostatistics, Faculty of Health Sciences, Universidad Científica del Sur, Lima, 4861, Peru.
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Beirut, Lebanon.
- Latin American Network of Coronavirus Disease 2019 - COVID-19 Research (LANCOVID-19), Pereira, Risaralda, Colombia.
| |
Collapse
|
157
|
Metry A, Pandor A, Ren S, Shippam A, Clowes M, Dark P, McMullan R, Stevenson M. Cost-effectiveness of therapeutics for COVID-19 patients: a rapid review and economic analysis. Health Technol Assess 2023; 27:1-92. [PMID: 37840452 PMCID: PMC10591210 DOI: 10.3310/nafw3527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 is the virus that causes coronavirus disease 2019. Over six million deaths worldwide have been associated with coronavirus disease 2019. Objective To assess the cost-effectiveness of treatments used for the treatment of coronavirus disease 2019 in hospital or used in the community in patients with coronavirus disease 2019 at high risk of hospitalisation. Setting Treatments provided in United Kingdom hospital and community settings. Methods Clinical effectiveness estimates were taken from the coronavirus disease-network meta-analyses initiative and the metaEvidence initiative. A mathematical model was constructed to explore how the interventions impacted on patient health, measured in quality-adjusted life-years gained. The costs associated with treatment, including those of hospital care, were also estimated and used to form a cost per quality-adjusted life-year gained value which was compared with thresholds published by the National Institute for Health and Care Excellence. Estimates of cost-effectiveness compared against current standard of care were produced in both the hospital and community settings at three different levels of efficacy: mean, low and high. Public list prices were used for interventions with neither confidential patient access schemes nor confidential list prices considered. Results incorporating confidential pricing data were provided to the National Institute for Health and Care Excellence appraisal committee. Results The treatments were estimated to be clinically effective although not all reached statistical significance. All treatments in the hospital setting, or community, were estimated to plausibly have a cost per quality-adjusted life-year gained value below National Institute for Health and Care Excellence's thresholds when compared with standard of care. However, almost all drugs could plausibly have cost per quality-adjusted life-years above National Institute for Health and Care Excellence's thresholds. However, there is considerable uncertainty in the results as the prevalent severe acute respiratory syndrome coronavirus 2 variant, vaccination status, history of being infected with severe acute respiratory syndrome coronavirus 2 and standard of care have all evolved since the pivotal studies were conducted which could have significant impact on the efficacy of each drug. For drugs used in high-risk patients in the community setting, the proportion of people at high risk who need hospital admission was a large driver of the cost per quality-adjusted life-year. Limitations No studies were identified that were conducted in current conditions. This may be a large limitation as the severe acute respiratory syndrome coronavirus 2 variant changes. No head-to-head studies of interventions were identified. Conclusions The results produced could be informative to decision-makers, although conclusions regarding the most clinical - and cost-effectiveness of each intervention should be tentative due to the evolving nature of the decision problem and, in this report, the use of list prices only. Comparisons between interventions should also be treated with caution due to potentially large heterogeneity between studies. Future work Research assessing the relative clinical effectiveness of interventions within head-to-head studies in current conditions would be beneficial. Contemporary information related to the probability of hospital admission and death for patients at high risk in the community would improve the precision of the estimates generated. Funding This project was funded by the National Institute for Health and Care Research (NIHR) Evidence Synthesis programme (NIHR135564) and will be published in full in Health Technology Assessment; Vol. 27, No. 14. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Andrew Metry
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| | - Abdullah Pandor
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| | - Shijie Ren
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| | - Andrea Shippam
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| | - Mark Clowes
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| | - Paul Dark
- The University of Manchester, Manchester, UK
| | - Ronan McMullan
- School of Medicine, Dentistry and Biomedical Sciences, Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Matt Stevenson
- School of Health and Related Research (ScHARR), The University of Sheffield, Sheffield, UK
| |
Collapse
|
158
|
Pandharipande P, Williams Roberson S, Harrison FE, Wilson JE, Bastarache JA, Ely EW. Mitigating neurological, cognitive, and psychiatric sequelae of COVID-19-related critical illness. THE LANCET. RESPIRATORY MEDICINE 2023; 11:726-738. [PMID: 37475124 PMCID: PMC10706757 DOI: 10.1016/s2213-2600(23)00238-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023]
Abstract
Despite advances in the treatment and mitigation of critical illness caused by infection with SARS-CoV-2, millions of survivors have a devastating, post-acute infection syndrome known as long COVID. A large proportion of patients with long COVID have nervous system dysfunction, which is also seen in the distinct but overlapping condition of post-intensive care syndrome (PICS), putting survivors of COVID-19-related critical illness at high risk of long-lasting morbidity affecting multiple organ systems and, as a result, engendering measurable deficits in quality of life and productivity. In this Series paper, we discuss neurological, cognitive, and psychiatric sequelae in patients who have survived critical illness due to COVID-19. We review current knowledge of the epidemiology and pathophysiology of persistent neuropsychological impairments, and outline potential preventive strategies based on safe, evidence-based approaches to the management of pain, agitation, delirium, anticoagulation, and ventilator weaning during critical illness. We highlight priorities for current and future research, including possible therapeutic approaches, and offer considerations for health services to address the escalating health burden of long COVID.
Collapse
Affiliation(s)
- Pratik Pandharipande
- Department of Anesthesiology, Division of Anesthesiology Critical Care Medicine, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Shawniqua Williams Roberson
- Departments of Neurology and Biomedical Engineering, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jo Ellen Wilson
- Department of Psychiatry and Behavioral Sciences, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center, VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Julie A Bastarache
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - E Wesley Ely
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center, VA Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
159
|
Sunny S, Tran A, Lee J, Abdallah M, Chaudhry N, Quale J. Comparison of Tocilizumab vs Baricitinib in Clinical Outcomes Among Hospitalized Patients With COVID-19: Experience From a Public Hospital System in New York City. Open Forum Infect Dis 2023; 10:ofad426. [PMID: 37608917 PMCID: PMC10442059 DOI: 10.1093/ofid/ofad426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023] Open
Abstract
Background Tocilizumab and baricitinib are immunomodulators that have been repurposed for the treatment of coronavirus disease 2019 (COVID-19). Whether one medication should be preferred over the other has not been established. Methods This multicenter retrospective cohort study comprised hospitalized patients with COVID-19 who received either tocilizumab or baricitinib. The primary outcome was improvement in respiratory status (at least 1-point reduction on the respiratory ordinal scale) at day 7 and up to day 28. Secondary outcomes included mortality, disposition, deep vein thrombosis, pulmonary embolism, or positive blood culture. Outcomes were stratified by baseline respiratory status and variant-predominating periods. Results were reported for the overall and propensity-matched cohorts. Results A total of 921 patients received tocilizumab and 638 received baricitinib. The propensity-matched cohort included 597 patients in each group. At day 7 in the overall and propensity-matched cohorts, significantly more patients had improvement in respiratory status in the baricitinib group. These improvements were seen in patients requiring supplemental oxygen and noninvasive ventilation/high-flow oxygen but not in patients requiring mechanical ventilation. Favorable outcomes with baricitinib were observed during the Alpha and Omicron periods. By day 28, there were no differences in the changes of respiratory status for the treatment groups in either cohort. Also, no differences were seen in mortality, disposition, development of deep vein thrombosis/pulmonary embolism, or bloodstream infections. Conclusions Baricitinib treatment was associated with more favorable respiratory improvement at day 7 when compared with tocilizumab, but no differences were observed up to day 28.
Collapse
Affiliation(s)
- Subin Sunny
- Division of Infectious Diseases, Department of Medicine, NYC Health + Hospitals/Kings County, Brooklyn, New York, USA
| | - Ami Tran
- Division of Infectious Diseases, Department of Medicine, NYC Health + Hospitals/Kings County, Brooklyn, New York, USA
| | - Jennifer Lee
- Division of Infectious Diseases, Department of Medicine, NYC Health + Hospitals/Kings County, Brooklyn, New York, USA
| | - Marie Abdallah
- Division of Infectious Diseases, Department of Medicine, NYC Health + Hospitals/Kings County, Brooklyn, New York, USA
| | - Nimra Chaudhry
- Department of Pharmacy, NYC Health + Hospitals/Queens, Queens, New York, USA
| | - John Quale
- Division of Infectious Diseases, Department of Medicine, NYC Health + Hospitals/Kings County, Brooklyn, New York, USA
| |
Collapse
|
160
|
Pilgram L, Appel KS, Ruethrich MM, Koll CEM, Vehreschild MJGT, de Miranda SMN, Hower M, Hellwig K, Hanses F, Wille K, Haselberger M, Spinner CD, Vom Dahl J, Hertenstein B, Westhoff T, Vehreschild JJ, Jensen BEO, Stecher M. Use and effectiveness of remdesivir for the treatment of patients with covid-19 using data from the Lean European Open Survey on SARS-CoV-2 infected patients (LEOSS): a multicentre cohort study. Infection 2023; 51:1033-1049. [PMID: 36763285 PMCID: PMC9913009 DOI: 10.1007/s15010-023-01994-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
OBJECTIVES The use of remdesivir (RDV) as the first drug approved for coronavirus disease 2019 (COVID-19) remains controversial. Based on the Lean European Open Survey on severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infected patients (LEOSS), we aim to contribute timing-focused complementary real-world insights to its evaluation. METHODS SARS-CoV-2 infected patients between January 2020 and December 2021 treated with RDV were matched 1:1 to controls considering sociodemographics, comorbidities and clinical status. Multiple imputations were used to account for missing data. Effects on fatal outcome were estimated using uni- and multivariable Cox regression models. RESULTS We included 9,687 patients. For those starting RDV administration in the complicated phase, Cox regression for fatal outcome showed an adjusted hazard ratio (aHR) of 0.59 (95%CI 0.41-0.83). Positive trends could be obtained for further scenarios: an aHR of 0.51 (95%CI 0.16-1.68) when RDV was initiated in uncomplicated and of 0.76 (95% CI 0.55-1.04) in a critical phase of disease. Patients receiving RDV with concomitant steroids exhibited a further reduction in aHR in both, the complicated (aHR 0.50, 95%CI 0.29-0.88) and critical phase (aHR 0.63, 95%CI 0.39-1.02). CONCLUSION Our study results elucidate that RDV use, in particular when initiated in the complicated phase and accompanied by steroids is associated with improved mortality. However, given the limitations of non-randomized trials in estimating the magnitude of the benefit of an intervention, further randomized trials focusing on the timing of therapy initiation seem warranted.
Collapse
Affiliation(s)
- Lisa Pilgram
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Digital Clinician Scientist Program, Berlin, Germany.
- Department of Internal Medicine, Hematology and Oncology, Goethe University Frankfurt, Frankfurt, Germany.
| | - Katharina S Appel
- Department of Internal Medicine, Hematology and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Maria M Ruethrich
- Department of Internal Medicine II, University Hospital Jena, Jena, Germany
| | - Carolin E M Koll
- Faculty of Medicine and Department I of Internal Medicine, University of Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Maria J G T Vehreschild
- Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt, Germany
| | - Susana M Nunes de Miranda
- Faculty of Medicine and Department I of Internal Medicine, University of Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | - Martin Hower
- Department of Pneumology, Infectiology, Internal Medicine and Intensive Care, Klinikum Dortmund GmbH, Hospital of University Witten/Herdecke, Dortmund, Germany
- Hospital of University Witten, Herdecke, Germany
| | - Kerstin Hellwig
- Department of Neurology, St. Josef-Hospital Bochum, Ruhr University Bochum, Bochum, Germany
| | - Frank Hanses
- Emergency Department, University Hospital Regensburg, Regensburg, Germany
- Department for Infectious Diseases and Infection Control, University Hospital Regensburg, Regensburg, Germany
| | - Kai Wille
- University of Bochum, University Clinic for Hematology, Oncology, Hemostaseology and Palliative Care, Minden, Germany
| | | | - Christoph D Spinner
- Department of Internal Medicine II, School of Medicine, Technical University of Munich, University Hospital Rechts Der Isar, Munich, Germany
| | - Juergen Vom Dahl
- Department of Cardiology, Hospital Maria Hilf GmbH Moenchengladbach, Moenchengladbach, Germany
| | - Bernd Hertenstein
- Department of Internal Medicine I, Hospital Bremen-Mitte, Bremen, Germany
| | - Timm Westhoff
- Department of Internal Medicine I, Marien Hospital Herne Ruhr University Bochum, Herne, Germany
| | - J Janne Vehreschild
- Faculty of Medicine and Department I of Internal Medicine, University of Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Department of Internal Medicine, Hematology and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Björn-Erik Ole Jensen
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Melanie Stecher
- Faculty of Medicine and Department I of Internal Medicine, University of Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| |
Collapse
|
161
|
Singh SJ, Baldwin MM, Daynes E, Evans RA, Greening NJ, Jenkins RG, Lone NI, McAuley H, Mehta P, Newman J, Novotny P, Smith DJF, Stanel S, Toshner M, Brightling CE. Respiratory sequelae of COVID-19: pulmonary and extrapulmonary origins, and approaches to clinical care and rehabilitation. THE LANCET. RESPIRATORY MEDICINE 2023; 11:709-725. [PMID: 37216955 PMCID: PMC10198676 DOI: 10.1016/s2213-2600(23)00159-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023]
Abstract
Although the exact prevalence of post-COVID-19 condition (also known as long COVID) is unknown, more than a third of patients with COVID-19 develop symptoms that persist for more than 3 months after SARS-CoV-2 infection. These sequelae are highly heterogeneous in nature and adversely affect multiple biological systems, although breathlessness is a frequently cited symptom. Specific pulmonary sequelae, including pulmonary fibrosis and thromboembolic disease, need careful assessment and might require particular investigations and treatments. COVID-19 outcomes in people with pre-existing respiratory conditions vary according to the nature and severity of the respiratory disease and how well it is controlled. Extrapulmonary complications such as reduced exercise tolerance and frailty might contribute to breathlessness in post-COVID-19 condition. Non-pharmacological therapeutic options, including adapted pulmonary rehabilitation programmes and physiotherapy techniques for breathing management, might help to attenuate breathlessness in people with post-COVID-19 condition. Further research is needed to understand the origins and course of respiratory symptoms and to develop effective therapeutic and rehabilitative strategies.
Collapse
Affiliation(s)
- Sally J Singh
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK.
| | - Molly M Baldwin
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Enya Daynes
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Rachael A Evans
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Neil J Greening
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - R Gisli Jenkins
- Imperial College London National Heart and Lung Institute, London, UK
| | - Nazir I Lone
- Department of Anaesthesia, Critical Care and Pain Medicine, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Hamish McAuley
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | - Puja Mehta
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, UK
| | - Joseph Newman
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Petr Novotny
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| | | | - Stefan Stanel
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Mark Toshner
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre-Respiratory and Infectious Diseases, Leicester, UK
| |
Collapse
|
162
|
Anesi GL, Degnan K, Dutcher L, Saw S, Maguire C, Binkley A, Patel S, Athans V, Barton TD, Binkley S, Candeloro CL, Herman DJ, Kasbekar N, Kennedy L, Millstein JH, Meyer NJ, Talati NJ, Patel H, Pegues DA, Sayre PJ, Tebas P, Terico AT, Murphy KM, O’Donnell JA, White M, Hamilton KW. The Penn Medicine COVID-19 Therapeutics Committee-Reflections on a Model for Rapid Evidence Review and Dynamic Practice Recommendations During a Public Health Emergency. Open Forum Infect Dis 2023; 10:ofad428. [PMID: 37663091 PMCID: PMC10468749 DOI: 10.1093/ofid/ofad428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
The Penn Medicine COVID-19 Therapeutics Committee-an interspecialty, clinician-pharmacist, and specialist-front line primary care collaboration-has served as a forum for rapid evidence review and the production of dynamic practice recommendations during the 3-year coronavirus disease 2019 public health emergency. We describe the process by which the committee went about its work and how it navigated specific challenging scenarios. Our target audiences are clinicians, hospital leaders, public health officials, and researchers invested in preparedness for inevitable future threats. Our objectives are to discuss the logistics and challenges of forming an effective committee, undertaking a rapid evidence review process, aligning evidence-based guidelines with operational realities, and iteratively revising recommendations in response to changing pandemic data. We specifically discuss the arc of evidence for corticosteroids; the noble beginnings and dangerous misinformation end of hydroxychloroquine and ivermectin; monoclonal antibodies and emerging viral variants; and patient screening and safety processes for tocilizumab, baricitinib, and nirmatrelvir-ritonavir.
Collapse
Affiliation(s)
- George L Anesi
- Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen Degnan
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Dutcher
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stephen Saw
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Christina Maguire
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Amanda Binkley
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Sonal Patel
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Vasilios Athans
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Todd D Barton
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shawn Binkley
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Christina L Candeloro
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - David J Herman
- Division of Infectious Diseases, Penn Medicine Princeton Medical Center, University of Pennsylvania Health System, Princeton, New Jersey, USA
| | - Nishaminy Kasbekar
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Leigh Kennedy
- Division of Infectious Diseases, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Jeffrey H Millstein
- Regional Physician Practices of Penn Medicine, Woodbury Heights, New Jersey, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Naasha J Talati
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hinal Patel
- Department of Pharmacy, Penn Medicine Princeton Medical Center, University of Pennsylvania Health System, Princeton, New Jersey, USA
| | - David A Pegues
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Patrick J Sayre
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Pablo Tebas
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Adrienne T Terico
- Department of Pharmacy, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Kathleen M Murphy
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Judith A O’Donnell
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Melissa White
- Department of Pharmacy, Penn Medicine Lancaster General Health, University of Pennsylvania Health System, Lancaster, Pennsylvania, USA
| | - Keith W Hamilton
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
163
|
Ware LB, Soleymanlou N, McAuley DF, Estrada V, Diaz GA, Lacamera P, Kaste R, Choi W, Gupta A, Welte T. TRPC6 inhibitor (BI 764198) to reduce risk and severity of ARDS due to COVID-19: a phase II randomised controlled trial. Thorax 2023; 78:816-824. [PMID: 37024277 PMCID: PMC10359525 DOI: 10.1136/thorax-2022-219668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/31/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Despite the availability of COVID-19 vaccinations, there remains a need to investigate treatments to reduce the risk or severity of potentially fatal complications of COVID-19, such as acute respiratory distress syndrome (ARDS). This study evaluated the efficacy and safety of the transient receptor potential channel C6 (TRPC6) inhibitor, BI 764198, in reducing the risk and/or severity of ARDS in patients hospitalised for COVID-19 and requiring non-invasive, supplemental oxygen support (oxygen by mask or nasal prongs, oxygen by non-invasive ventilation or high-flow nasal oxygen). METHODS Multicentre, double-blind, randomised phase II trial comparing once-daily oral BI 764198 (n=65) with placebo (n=64) for 28 days (+2-month follow-up). PRIMARY ENDPOINT proportion of patients alive and free of mechanical ventilation at day 29. Secondary endpoints: proportion of patients alive and discharged without oxygen (day 29); occurrence of either in-hospital mortality, intensive care unit admission or mechanical ventilation (day 29); time to first response (clinical improvement/recovery); ventilator-free days (day 29); and mortality (days 15, 29, 60 and 90). RESULTS No difference was observed for the primary endpoint: BI 764198 (83.1%) versus placebo (87.5%) (estimated risk difference -5.39%; 95% CI -16.08 to 5.30; p=0.323). For secondary endpoints, a longer time to first response (rate ratio 0.67; 95% CI 0.46 to 0.99; p=0.045) and longer hospitalisation (+3.41 days; 95% CI 0.49 to 6.34; p=0.023) for BI 764198 versus placebo was observed; no other significant differences were observed. On-treatment adverse events were similar between trial arms and more fatal events were reported for BI 764198 (n=7) versus placebo (n=2). Treatment was stopped early based on an interim observation of a lack of efficacy and an imbalance of fatal events (Data Monitoring Committee recommendation). CONCLUSIONS TRPC6 inhibition was not effective in reducing the risk and/or severity of ARDS in patients with COVID-19 requiring non-invasive, supplemental oxygen support. TRIAL REGISTRATION NUMBER NCT04604184.
Collapse
Affiliation(s)
- Lorraine B Ware
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Nima Soleymanlou
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Danny Francis McAuley
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - Vicente Estrada
- Hospital Clínico San Carlos, IdISSC; CIBERINFE, Madrid, Spain
| | - George A Diaz
- Section of Infectious Diseases, Providence Regional Medical Center Everett, Everett, Washington, USA
| | - Peter Lacamera
- Division of Pulmonary and Critical Care Medicine, St Elizabeth's Medical Center, Boston, Massachusetts, USA
| | - Renee Kaste
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Wansuk Choi
- TA Cardio-Metabolism & Respiratory, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Abhya Gupta
- TA Inflammation Medicine, Boehringer Ingelheim International GmbH, Biberach an der Riss, Germany
| | - Tobias Welte
- Department of Pneumology, Hannover Medical School, Hannover, Niedersachsen, Germany
| |
Collapse
|
164
|
Karolyi M, Gruebl A, Omid S, Saak M, Pawelka E, Hoepler W, Kelani H, Kuran A, Laferl H, Ott C, Pereyra D, Santol J, Seitz T, Traugott M, Assinger A, Wenisch C, Zoufaly A. Tocilizumab vs. baricitinib in hospitalized severe COVID-19 patients: results from a real-world cohort. Infection 2023; 51:851-858. [PMID: 36083403 PMCID: PMC9461450 DOI: 10.1007/s15010-022-01915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tocilizumab and baricitinib are recommended treatment options for hospitalized COVID-19 patients requiring oxygen support. Literature about its efficacy and safety in a head-to-head comparison is scarce. METHODS Hospitalized COVID-19 patients requiring oxygen were treated with tocilizumab or baricitinib additionally to dexamethasone. Tocilizumab was available from February till the 19th of September 2021 and baricitinib from 21st of September. The primary outcome was in-hospital mortality. Secondary outcome parameters were progression to mechanical ventilation (MV), length-of-stay (LOS) and potential side effects. RESULTS 159 patients (tocilizumab 68, baricitinib 91) with a mean age of 60.5 years, 64% male were included in the study. Tocilizumab patients were admitted 1 day earlier, were in a higher WHO category at the time of inclusion and had a higher CRP level on admission and treatment initiation. Patients receiving Tocilizumab were treated with remdesivir more often and only patients in the baricitinib group were treated with monoclonal antibodies. Other characteristics did not differ significantly. In-hospital mortality (18% vs. 11%, p = 0.229), progression to MV (19% vs. 11%, p = 0.173) and LOS (13 vs. 12 days, p = 0.114) did not differ between groups. Side effects were equally distributed between groups, except ALAT elevation which was significantly more often observed in the tocilizumab group (43% vs. 25%, p = 0.021). CONCLUSIONS In-hospital mortality, progression to MV and LOS were not significantly different in patients treated with tocilizumab or baricitinib additionally to standard of care. Both drugs seem equally effective but further head-to-head trials are needed.
Collapse
Affiliation(s)
- Mario Karolyi
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria.
| | - Andreas Gruebl
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Sara Omid
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Magdalena Saak
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Erich Pawelka
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Wolfgang Hoepler
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Hasan Kelani
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Avelino Kuran
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Hermann Laferl
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Clemens Ott
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - David Pereyra
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jonas Santol
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Surgery, HPB Center, Viennese Health Network, Klinik Favoriten and Sigmund Freud Private University, Vienna, Austria
| | - Tamara Seitz
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Marianna Traugott
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christoph Wenisch
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
| | - Alexander Zoufaly
- Department for Infectious Diseases and Tropical Medicine, Klinik Favoriten, Kundratstraße 3, 1100, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| |
Collapse
|
165
|
Zhang T, Tian W, Wei S, Lu X, An J, He S, Zhao J, Gao Z, Li L, Lian K, Zhou Q, Zhang H, Wang L, Su L, Kang H, Niu T, Zhao A, Pan J, Cai Q, Xu Z, Chen W, Jing H, Li P, Zhao W, Cao Y, Mi J, Chen T, Chen Y, Zou P, Lukacs-Kornek V, Kurts C, Li J, Liu X, Mei Q, Zhang Y, Wei J. Multidisciplinary recommendations for the management of CAR-T recipients in the post-COVID-19 pandemic era. Exp Hematol Oncol 2023; 12:66. [PMID: 37501090 PMCID: PMC10375673 DOI: 10.1186/s40164-023-00426-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) posed an unprecedented challenge on public health systems. Despite the measures put in place to contain it, COVID-19 is likely to continue experiencing sporadic outbreaks for some time, and individuals will remain susceptible to recurrent infections. Chimeric antigen receptor (CAR)-T recipients are characterized by durable B-cell aplasia, hypogammaglobulinemia and loss of T-cell diversity, which lead to an increased proportion of severe/critical cases and a high mortality rate after COVID-19 infection. Thus, treatment decisions have become much more complex and require greater caution when considering CAR T-cell immunotherapy. Hence, we reviewed the current understanding of COVID-19 and reported clinical experience in the management of COVID-19 and CAR-T therapy. After a panel discussion, we proposed a rational procedure pertaining to CAR-T recipients with the aim of maximizing the benefit of CAR-T therapy in the post COVID-19 pandemic era.
Collapse
Affiliation(s)
- Tingting Zhang
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Weiwei Tian
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Respiratory and Critical Care Medicine, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Xinyi Lu
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Jing An
- School of Public Health, Shanxi Medical University, Taiyuan, 030000, Shanxi, China
| | - Shaolong He
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Jie Zhao
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Zhilin Gao
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Li Li
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Ke Lian
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China
| | - Qiang Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Cardiovascular Medicine, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Huilai Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Liang Wang
- Department of Hematology, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
| | - Liping Su
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Huicong Kang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Neurology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Pan
- State Key Laboratory of Experimental Hematology, Boren Biotherapy Translational Laboratory, Boren Clinical Translational Center, Beijing GoBroad Boren Hospital, Beijing, 100070, China
| | - Qingqing Cai
- Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Zhenshu Xu
- Hematology Department, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, 350001, Fujian, China
| | - Wenming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hongmei Jing
- Department of Hematology, Peking University Third Hospital, Beijing, 100191, China
| | - Peng Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, Guangdong, China
| | - Wanhong Zhao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Jianqing Mi
- Shanghai Institute of Hematology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tao Chen
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yuan Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Geriatrics, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Ping Zou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, 53111, Bonn, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, 53111, Bonn, Germany
| | - Jian Li
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, 53111, Bonn, Germany
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Department of Respiratory and Critical Care Medicine, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China.
| | - Qi Mei
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China.
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Jia Wei
- Department of Hematology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, Shanxi, China.
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, Shanxi, China.
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
166
|
Perreault G, Ching C, Nobel YR. COVID-19 in patients with liver disease and liver transplant: clinical implications, prevention, and management. Therap Adv Gastroenterol 2023; 16:17562848231188586. [PMID: 37521085 PMCID: PMC10372508 DOI: 10.1177/17562848231188586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/02/2023] [Indexed: 08/01/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has had enormous implications for the care of patients with chronic liver disease (CLD), cirrhosis, and liver transplant (LT). Clinical outcomes of COVID-19 vary in patients with CLD and cirrhosis compared to healthy controls, and in patients with LT compared to patients without LT. Several special considerations apply to the approach to vaccination and treatment in patients with CLD and LT. The practice of liver transplantation has also been heavily impacted by the pandemic, including persistent reductions in living donor LT and increases in LT for an indication of alcohol-related liver disease. Recent medical society guidelines strive to standardize severe acute respiratory syndrome coronavirus 2 testing in donors and recipients and the approach to transplantation after recovered from COVID-19 infection, but certain controversies remain.
Collapse
Affiliation(s)
- Gabriel Perreault
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Charlotte Ching
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
167
|
O’Halloran JA, Ko ER, Anstrom KJ, Kedar E, McCarthy MW, Panettieri RA, Maillo M, Nunez PS, Lachiewicz AM, Gonzalez C, Smith PB, de Tai SMT, Khan A, Lora AJM, Salathe M, Capo G, Gonzalez DR, Patterson TF, Palma C, Ariza H, Lima MP, Blamoun J, Nannini EC, Sprinz E, Mykietiuk A, Alicic R, Rauseo AM, Wolfe CR, Witting B, Wang JP, Parra-Rodriguez L, Der T, Willsey K, Wen J, Silverstein A, O’Brien SM, Al-Khalidi HR, Maldonado MA, Melsheimer R, Ferguson WG, McNulty SE, Zakroysky P, Halabi S, Benjamin DK, Butler S, Atkinson JC, Adam SJ, Chang S, LaVange L, Proschan M, Bozzette SA, Powderly WG. Abatacept, Cenicriviroc, or Infliximab for Treatment of Adults Hospitalized With COVID-19 Pneumonia: A Randomized Clinical Trial. JAMA 2023; 330:328-339. [PMID: 37428480 PMCID: PMC10334296 DOI: 10.1001/jama.2023.11043] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023]
Abstract
Importance Immune dysregulation contributes to poorer outcomes in COVID-19. Objective To investigate whether abatacept, cenicriviroc, or infliximab provides benefit when added to standard care for COVID-19 pneumonia. Design, Setting, and Participants Randomized, double-masked, placebo-controlled clinical trial using a master protocol to investigate immunomodulators added to standard care for treatment of participants hospitalized with COVID-19 pneumonia. The results of 3 substudies are reported from 95 hospitals at 85 clinical research sites in the US and Latin America. Hospitalized patients 18 years or older with confirmed SARS-CoV-2 infection within 14 days and evidence of pulmonary involvement underwent randomization between October 2020 and December 2021. Interventions Single infusion of abatacept (10 mg/kg; maximum dose, 1000 mg) or infliximab (5 mg/kg) or a 28-day oral course of cenicriviroc (300-mg loading dose followed by 150 mg twice per day). Main Outcomes and Measures The primary outcome was time to recovery by day 28 evaluated using an 8-point ordinal scale (higher scores indicate better health). Recovery was defined as the first day the participant scored at least 6 on the ordinal scale. Results Of the 1971 participants randomized across the 3 substudies, the mean (SD) age was 54.8 (14.6) years and 1218 (61.8%) were men. The primary end point of time to recovery from COVID-19 pneumonia was not significantly different for abatacept (recovery rate ratio [RRR], 1.12 [95% CI, 0.98-1.28]; P = .09), cenicriviroc (RRR, 1.01 [95% CI, 0.86-1.18]; P = .94), or infliximab (RRR, 1.12 [95% CI, 0.99-1.28]; P = .08) compared with placebo. All-cause 28-day mortality was 11.0% for abatacept vs 15.1% for placebo (odds ratio [OR], 0.62 [95% CI, 0.41-0.94]), 13.8% for cenicriviroc vs 11.9% for placebo (OR, 1.18 [95% CI 0.72-1.94]), and 10.1% for infliximab vs 14.5% for placebo (OR, 0.59 [95% CI, 0.39-0.90]). Safety outcomes were comparable between active treatment and placebo, including secondary infections, in all 3 substudies. Conclusions and Relevance Time to recovery from COVID-19 pneumonia among hospitalized participants was not significantly different for abatacept, cenicriviroc, or infliximab vs placebo. Trial Registration ClinicalTrials.gov Identifier: NCT04593940.
Collapse
Affiliation(s)
| | - Emily R. Ko
- Duke University Health System, Durham, North Carolina
| | | | | | | | | | | | | | | | - Cynthia Gonzalez
- National Center for Advancing Translational Sciences, Bethesda, Maryland
| | - P. Brian Smith
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Akram Khan
- Oregon Health and Science University, Portland
| | | | | | | | | | | | - Christopher Palma
- University of Rochester School of Medicine and Dentistry, Rochester, New York
| | | | | | | | | | - Eduardo Sprinz
- Hospital de Clinicas de Porto Alegre HCPA, Porto Alegre, Brazil
| | | | - Radica Alicic
- Providence Medical Research Center, Spokane, Washington
| | | | | | | | | | | | - Tatyana Der
- Duke University Health System, Durham, North Carolina
| | | | - Jun Wen
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Adam Silverstein
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Sean M. O’Brien
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Hussein R. Al-Khalidi
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | | | | | | | - Steven E. McNulty
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Pearl Zakroysky
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Susan Halabi
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Daniel K. Benjamin
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Sandra Butler
- Technical Resources International (TRI), Bethesda, Maryland
| | - Jane C. Atkinson
- National Center for Advancing Translational Sciences, Bethesda, Maryland
| | - Stacey J. Adam
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Soju Chang
- National Center for Advancing Translational Sciences, Bethesda, Maryland
| | | | - Michael Proschan
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Samuel A. Bozzette
- National Center for Advancing Translational Sciences, Bethesda, Maryland
| | | |
Collapse
|
168
|
Garcia-Carretero R, Vazquez-Gomez O, Ordoñez-Garcia M, Garrido-Peño N, Gil-Prieto R, Gil-de-Miguel A. Differences in Trends in Admissions and Outcomes among Patients from a Secondary Hospital in Madrid during the COVID-19 Pandemic: A Hospital-Based Epidemiological Analysis (2020-2022). Viruses 2023; 15:1616. [PMID: 37515302 PMCID: PMC10384448 DOI: 10.3390/v15071616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Spain had some of Europe's highest incidence and mortality rates for coronavirus disease 2019 (COVID-19). This study highlights the impact of the COVID-19 pandemic on daily health care in terms of incidence, critical patients, and mortality. We describe the characteristics and clinical outcomes of patients, comparing variables over the different waves. We performed a descriptive, retrospective study using the historical records of patients hospitalized with COVID-19. We describe demographic characteristics, admissions, and occupancy. Time series allowed us to visualize and analyze trends and patterns, and identify several waves during the 27-month period. A total of 3315 patients had been hospitalized with confirmed COVID-19. One-third of these patients were hospitalized during the first weeks of the pandemic. We observed that 4.6% of all hospitalizations had been admitted to the intensive care unit, and we identified a mortality rate of 9.4% among hospitalized patients. Arithmetic- and semi-logarithmic-scale charts showed how admissions and deaths rose sharply during the first weeks, increasing by 10 every few days. We described a single hospital's response and experiences during the pandemic. This research highlights certain demographic profiles in a population and emphasizes the importance of identifying waves when performing research on COVID-19. Our results can extend the analysis of the impact of COVID-19 and can be applied in other contexts, and can be considered when further analyzing the clinical, epidemiological, or demographic characteristics of populations with COVID-19. Our findings suggest that the pandemic should be analyzed not as a whole but rather in different waves.
Collapse
Affiliation(s)
- Rafael Garcia-Carretero
- Department of Internal Medicine, Mostoles University Hospital, 28935 Móstoles, Madrid, Spain
| | - Oscar Vazquez-Gomez
- Department of Internal Medicine, Mostoles University Hospital, 28935 Móstoles, Madrid, Spain
| | - María Ordoñez-Garcia
- Department of Hematology, Mostoles University Hospital, 28935 Móstoles, Madrid, Spain
| | - Noelia Garrido-Peño
- Department of Pharmacy, Mostoles University Hospital, 28935 Móstoles, Madrid, Spain
| | - Ruth Gil-Prieto
- Department of Preventive Medicine and Public Health, Rey Juan Carlos University, 28922 Alcorcón, Madrid, Spain
| | - Angel Gil-de-Miguel
- Department of Preventive Medicine and Public Health, Rey Juan Carlos University, 28922 Alcorcón, Madrid, Spain
| |
Collapse
|
169
|
Belperio J, Nguyen T, Lombardi DA, Bogus M, Moskalenko V, Singh D, Haumann B, Bourdet DL, Kaufman E, Pfeifer ND, Thompson CG, Woo J, Moran EJ, Saggar R. Efficacy and safety of an inhaled pan-Janus kinase inhibitor, nezulcitinib, in hospitalised patients with COVID-19: results from a phase 2 clinical trial. BMJ Open Respir Res 2023; 10:e001627. [PMID: 37460276 DOI: 10.1136/bmjresp-2023-001627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND The inhaled lung-selective pan-Janus kinase inhibitor nezulcitinib had favourable safety and potential efficacy signals in part 1 of a phase 2 trial in patients with severe COVID-19, supporting progression to part 2. METHODS Part 2 was a randomised, double-blind phase 2 study (NCT04402866). Hospitalised patients aged 18-80 years with confirmed symptomatic COVID-19 requiring supplemental oxygen (excluding baseline invasive mechanical ventilation) were randomised 1:1 to nebulised nezulcitinib 3 mg or placebo for up to 7 days with background standard-of-care therapy (including corticosteroids). Efficacy endpoints included respiratory failure-free (RFF) days through day 28 as the primary endpoint. Secondary endpoints included safety and change from baseline oxygen saturation (SaO2)/fraction of inspired oxygen (FiO2) ratio on day 7, and 28-day mortality rate was a prespecified exploratory endpoint. RESULTS Between June 2020 and April 2021, 205 patients were treated (nezulcitinib, 103; placebo, 102). There was no statistically significant difference between nezulcitinib versus placebo in the primary endpoint (RFF days; median, 21.0 vs 21.0; p=0.6137) or secondary efficacy endpoints. Nezulcitinib was generally well tolerated with a favourable safety profile. CONCLUSIONS Although the prespecified primary, secondary and exploratory efficacy endpoints, including RFF through day 28, change from baseline SaO2/FiO2 ratio on day 7, and 28-day mortality rate, were not met, nezulcitinib was generally well tolerated and had a favourable safety profile. Further studies are required to determine if treatment with nezulcitinib confers clinical benefit in specific inflammatory biomarker-defined populations of patients with COVID-19.
Collapse
Affiliation(s)
- John Belperio
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | - Tuan Nguyen
- Theravance Biopharma Inc, South San Francisco, California, USA
| | | | - Maxim Bogus
- Arensia Exploratory Medicine SRL, Chișinău, Moldova (the Republic of)
- Timofei Mosneaga Republican Hospital, Chișinău, Moldova (the Republic of)
| | - Valentyn Moskalenko
- Arensia Exploratory Medicine, LLC, Kyiv City Clinical Hospital #12, Oleksandrivska Kyiv City Clinical Hospital, Kyiv, Ukraine
- Brovary Multidisciplinary Clinical Hospital, Brovary, Ukraine
| | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - David L Bourdet
- Theravance Biopharma Inc, South San Francisco, California, USA
| | - Elad Kaufman
- Theravance Biopharma Inc, South San Francisco, California, USA
| | | | | | - Jacky Woo
- Theravance Biopharma Inc, South San Francisco, California, USA
| | - Edmund J Moran
- Theravance Biopharma Inc, South San Francisco, California, USA
| | - Rajeev Saggar
- Theravance Biopharma Inc, South San Francisco, California, USA
| |
Collapse
|
170
|
Tanaka H, Chubachi S, Namkoong H, Sato Y, Asakura T, Lee H, Azekawa S, Otake S, Nakagawara K, Fukushima T, Watase M, Sakurai K, Kusumoto T, Kondo Y, Masaki K, Kamata H, Ishii M, Kaneko Y, Hasegawa N, Ueda S, Sasaki M, Izumo T, Inomata M, Miyazawa N, Kimura Y, Suzuki Y, Harada N, Ichikawa M, Takata T, Ishikura H, Yoshiyama T, Kokuto H, Murakami K, Sano H, Ueda T, Kuwahara N, Fujiwara A, Ogura T, Inoue T, Asami T, Mutoh Y, Nakachi I, Baba R, Nishi K, Tani M, Kagyo J, Hashiguchi M, Oguma T, Asano K, Nishikawa M, Watanabe H, Okada Y, Koike R, Kitagawa Y, Kimura A, Imoto S, Miyano S, Ogawa S, Kanai T, Fukunaga K. Propensity-Score Matched Analysis of the Effectiveness of Baricitinib in Patients With Coronavirus Disease 2019 (COVID-19) Using Nationwide Real-World Data: An Observational Matched Cohort Study From the Japan COVID-19 Task Force. Open Forum Infect Dis 2023; 10:ofad311. [PMID: 37441355 PMCID: PMC10334380 DOI: 10.1093/ofid/ofad311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Background To determine the effectiveness of baricitinib in patients with coronavirus disease 2019 (COVID-19), investigate whether baricitinib prevents the need for invasive mechanical ventilation and identify patient subgroups that would benefit from baricitinib. Methods This observational matched-cohort study was conducted by the Japan COVID-19 Task Force, a nationwide multicenter consortium. Patients with COVID-19 aged ≥18 years were identified from 70 hospitals in Japan. Among patients with confirmed COVID-19 from February 2020 to September 2021, those receiving baricitinib were propensity-score matched with controls. Results Among 3309 patients, 144 propensity score-matched pairs were identified. Thirteen (9.0%) patients in the baricitinib group and 27 (18.8%) in the control group required invasive mechanical ventilation during the disease course (odds ratio, 0.43). Although the baricitinib group had more severe disease, there were no significant differences in the intensive care unit admission rates (odds ratio, 1.16) and mortality rates (odds ratio, 0.74) between groups. In subgroup analyses, baricitinib was associated with a significant reduction in the need for invasive mechanical ventilation in patients requiring oxygen support (odds ratio, 0.28), with rapid shadow spread on chest radiography (odds ratio, 0.11), or treated with remdesivir (odds ratio, 0.27), systemic corticosteroids (odds ratio, 0.31), or anticoagulants (odds ratio, 0.17). Conclusions Baricitinib is effective at preventing the need for invasive mechanical ventilation in patients with COVID-19.
Collapse
Affiliation(s)
- Hiromu Tanaka
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Chubachi
- Correspondence: Shotaro Chubachi, MD, PhD, Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan (); Ho Namkoong, MD, PhD, Department of Infectious Diseases, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan ()
| | - Ho Namkoong
- Correspondence: Shotaro Chubachi, MD, PhD, Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan (); Ho Namkoong, MD, PhD, Department of Infectious Diseases, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan ()
| | - Yasunori Sato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ho Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shiro Otake
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Nakagawara
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Fukushima
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mayuko Watase
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kaori Sakurai
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Kusumoto
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yasushi Kondo
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Katsunori Masaki
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Soichiro Ueda
- JCHO (Japan Community Health Care Organization) Saitama Medical Center, Internal Medicine, Saitama, Japan
| | - Mamoru Sasaki
- JCHO (Japan Community Health Care Organization) Saitama Medical Center, Internal Medicine, Saitama, Japan
| | | | | | - Naoki Miyazawa
- Department of Respiratory Medicine, Saiseikai Yokohamashi Nanbu Hospital, Yokohama, Japan
| | - Yasuhiro Kimura
- Department of Respiratory Medicine, Saiseikai Yokohamashi Nanbu Hospital, Yokohama, Japan
| | - Yusuke Suzuki
- Department of Respiratory Medicine, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Norihiro Harada
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, Japan
| | - Masako Ichikawa
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, Japan
| | - Tohru Takata
- Department of Infection Control, Fukuoka University Hospital, Fukuoka, Japan
| | - Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | | | | | - Koji Murakami
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirohito Sano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Ueda
- Department of Respiratory Medicine, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Naota Kuwahara
- Internal Medicine, Internal Medicine Center, Showa University Koto Toyosu Hospital, Tokyo, Japan
| | - Akiko Fujiwara
- Internal Medicine, Internal Medicine Center, Showa University Koto Toyosu Hospital, Tokyo, Japan
| | - Takashi Ogura
- Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Takashi Inoue
- Internal Medicine, Sano Kosei General Hospital, Sano, Japan
| | - Takahiro Asami
- Internal Medicine, Sano Kosei General Hospital, Sano, Japan
| | - Yoshikazu Mutoh
- Department of Infectious Diseases, Tosei General Hospital, Seto, Japan
| | | | - Rie Baba
- Department of Infectious Diseases, Tosei General Hospital, Seto, Japan
| | - Koichi Nishi
- Ishikawa Prefectural Central Hospital, Kanazawa, Japan
| | - Mayuko Tani
- Ishikawa Prefectural Central Hospital, Kanazawa, Japan
| | | | | | - Tsuyoshi Oguma
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Masanori Nishikawa
- Department of Respiratory Medicine, Fujisawa City Hospital, Fujisawa, Japan
| | - Hiroki Watanabe
- Department of Respiratory Medicine, Fujisawa City Hospital, Fujisawa, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Ryuji Koike
- Medical Innovation Promotion Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
171
|
Tokareva K, Reid P, Yang V, Liew D, Peterson AC, Baraff A, Giles J, Singh N. JAK inhibitors and black box warnings: what is the future for JAK inhibitors? Expert Rev Clin Immunol 2023; 19:1385-1397. [PMID: 37596779 PMCID: PMC10615860 DOI: 10.1080/1744666x.2023.2249237] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
INTRODUCTION Janus kinase inhibitors (JAKi) have dramatically improved the treatment of various autoimmune and myeloproliferative disorders. Recently, concern has arisen regarding their safety in patients with rheumatoid arthritis. AREAS COVERED Here, we provide a comprehensive summary of the major current and emerging JAKi and their indications, address recent studies on comparative safety, and provide insight into their future and use. We emphasize that the application of the research findings on a case-by-case basis should consider a patient's age, comorbidities, disease for which JAKi is being considered, disease activity, the JAKi target(s), alternate treatment options available for the patient, and the planned duration of JAKi. EXPERT OPINION Rheumatologists are used to prescribing therapies in which a risk-to-benefit assessment is required as well as to screening and monitoring the safety of medications. Thus, rheumatologists are already practiced in applying specific criteria to effectively screen and monitor patients who are candidates for JAKi therapy. Ongoing research will help to clarify any mechanisms underlying differential safety signals between JAK and other therapies, what the balance between risk and efficacy is, who the susceptible subpopulations are, and whether safety signals are shared between different JAKis and across indications.
Collapse
Affiliation(s)
- Kate Tokareva
- Medical Student, University of Washington, Seattle, WA, USA
| | - Pankti Reid
- Division of Rheumatology and Committee on Clinical Pharmacology and Pharmacogenomics, Department of Medicine, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Victor Yang
- Department of Rheumatology, Austin Health, Melbourne, Victoria, Australia
| | - David Liew
- Department of Rheumatology, Austin Health, Melbourne, Victoria, Australia
- Department of Clinical Pharmacology and Therapeutics, Austin Health, Melbourne, Victoria, Australia
| | | | - Aaron Baraff
- VA Puget Sound Healthcare System, Seattle, WA, USA
| | - Jon Giles
- Columbia University, New York, NY, USA
| | - Namrata Singh
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
172
|
Patel NM, Collotta D, Aimaretti E, Ferreira Alves G, Kröller S, Coldewey SM, Collino M, Thiemermann C. Inhibition of the JAK/STAT Pathway With Baricitinib Reduces the Multiple Organ Dysfunction Caused by Hemorrhagic Shock in Rats. Ann Surg 2023; 278:e137-e146. [PMID: 35837955 PMCID: PMC10249600 DOI: 10.1097/sla.0000000000005571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to investigate (a) the effects of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway inhibitor (baricitinib) on the multiple organ dysfunction syndrome (MODS) in a rat model of hemorrhagic shock (HS) and (b) whether treatment with baricitinib attenuates the activation of JAK/STAT, NF-κB, and NLRP3 caused by HS. BACKGROUND Posttraumatic MODS, which is in part due to excessive systemic inflammation, is associated with high morbidity and mortality. The JAK/STAT pathway is a regulator of numerous growth factor and cytokine receptors and, hence, is considered a potential master regulator of many inflammatory signaling processes. However, its role in trauma-hemorrhage is unknown. METHODS An acute HS rat model was performed to determine the effect of baricitinib on MODS. The activation of JAK/STAT, NF-κB, and NLRP3 pathways were analyzed by western blotting in the kidney and liver. RESULTS We demonstrate here for the first time that treatment with baricitinib (during resuscitation following severe hemorrhage) attenuates the organ injury and dysfunction and the activation of JAK/STAT, NF-κB, and NLRP3 pathways caused by HS in the rat. CONCLUSIONS Our results point to a role of the JAK/STAT pathway in the pathophysiology of the organ injury and dysfunction caused by trauma/hemorrhage and indicate that JAK inhibitors, such as baricitinib, may be repurposed for the treatment of the MODS after trauma and/or hemorrhage.
Collapse
Affiliation(s)
- Nikita M. Patel
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Debora Collotta
- Department of Neurosciences “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Eleonora Aimaretti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Sarah Kröller
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
173
|
Zhang W, Ling L, Li J, Li Y, Liu Y. Coronavirus disease 2019 and acute cerebrovascular events: a comprehensive overview. Front Neurol 2023; 14:1216978. [PMID: 37448747 PMCID: PMC10337831 DOI: 10.3389/fneur.2023.1216978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Since the Corona Virus Disease 2019 (COVID-19) pandemic, there has been increasing evidence that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with acute cerebrovascular events such as cerebral infarction, cerebral hemorrhage, and cerebral venous thrombosis. Although the mechanism of cerebrovascular complications among COVID-19 patients has not been adequately elucidated, the hypercoagulable state, excessive inflammation and ACE-2-associated alterations in the renin-angiotensin-aldosterone system after SARS-CoV-2 infection probably play an essential role. In this overview, we discuss the possible mechanisms underlying the SARS-CoV-2 infection leading to acute cerebrovascular events and review the characteristics of COVID-19-related acute cerebrovascular events cases and treatment options available worldwide.
Collapse
Affiliation(s)
- Wanzhou Zhang
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Li Ling
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Jie Li
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yudi Li
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yajie Liu
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
174
|
Massart N, Fillatre P, Lemaitre F, Mari A, Tron C. Pharmacokinetics of baricitinib in critically ill COVID-19 patients. Clin Biochem 2023:110601. [PMID: 37353137 PMCID: PMC10284613 DOI: 10.1016/j.clinbiochem.2023.110601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND The use of the selective Janus Kinase 1/2 inhibitor baricitinib has shown a survival benefit in mechanically ventilated COVID-19 patients but this is not without adverse drug reactions. Although critically ill patients are at risk of altered drug exposure, data on baricitinib pharmacokinetics (PK) are scarce. This study describes real-life baricitinib plasma exposure in critically ill COVID-19 patients. METHODS This retrospective observational study was conducted in critically ill patients with COVID-19 treated with baricitinib 4 mg/day. Plasma concentrations were measured at predose (C0), 1 h (C1) and 3 h (C3) after the drug intake. PK and area under the curve (AUC) were estimated using non-compartmental pharmacokinetic analysis. RESULTS Seven patients contributed to 22 baricitinib plasma concentration measurements after a median [range] of 3 days [2-3] of treatment. Median baricitinib plasma concentrations were 2.2 ng/mL [1.4-8.0], 24.0 ng/mL [4.9-37.3] and 14.1 ng/mL [8.3-15.1] for trough (C0), C1 and C3 concentrations respectively. The median AUC 0-24h was 188.8 ng.h/mL [141.3-236.3]. No difference was observed in C0 and C1 when comparing patients according to body mass index < or > 30. The patient with the lowest glomerular filtration rate (74 mL/min) had the highest baricitinib trough concentration. Overall, 2 patients had liver function test perturbation and both of them had atypical PK with delayed time to reach maximum concentration. CONCLUSION High inter-patient variability and relatively low baricitinib trough concentrations and AUC were observed in critically ill COVID-19 patients receiving the usual dosage of 4 mg/day. This preliminary study encourages further exploration of the concentration-effect relationship of baricitinib in this clinical context.
Collapse
Affiliation(s)
- Nicolas Massart
- Service de Réanimation, CH de St BRIEUC, 10, rue Marcel Proust, 22000 Saint-Brieuc, France
| | - Pierre Fillatre
- Service de Réanimation, CH de St BRIEUC, 10, rue Marcel Proust, 22000 Saint-Brieuc, France
| | - Florian Lemaitre
- Laboratoire de Pharmacologie, CHU de rennes, 2, rue Henri le, Guilloux 35000 rennes, France
| | - Arnaud Mari
- Service de Réanimation, CH de St BRIEUC, 10, rue Marcel Proust, 22000 Saint-Brieuc, France
| | - Camille Tron
- Laboratoire de Pharmacologie, CHU de rennes, 2, rue Henri le, Guilloux 35000 rennes, France.
| |
Collapse
|
175
|
Nay MA, Hindre R, Perrin C, Clément J, Plantier L, Sève A, Druelle S, Morrier M, Lainé JB, Colombain L, Corvaisier G, Bizien N, Pouget-Abadie X, Bigot A, Jamard S, Nyamankolly E, Planquette B, Fossat G, Boulain T. Prone position versus usual care in hypoxemic COVID-19 patients in medical wards: a randomised controlled trial. Crit Care 2023; 27:240. [PMID: 37330512 PMCID: PMC10276908 DOI: 10.1186/s13054-023-04529-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Benefit of early awake prone positioning for COVID-19 patients hospitalised in medical wards and who need oxygen therapy remains to be demonstrated. The question was considered at the time of COVID-19 pandemic to avoid overloading the intensive care units. We aimed to determine whether prone position plus usual care could reduce the rate of non-invasive ventilation (NIV) or intubation or death as compared to usual care alone. METHODS In this multicentre randomised clinical trial, 268 patients were randomly assigned to awake prone position plus usual care (N = 135) or usual care alone (N = 132). The primary outcome was the proportion of patients who underwent NIV or intubation or died within 28 days. Main secondary outcomes included the rates of NIV, of intubation or death, within 28 days. RESULTS Median time spent each day in the prone position within 72 h of randomisation was 90 min (IQR 30-133). The proportion of NIV or intubation or death within 28 days was 14.1% (19/135) in the prone position group and 12.9% (17/132) in the usual care group [odds ratio adjusted for stratification (aOR) 0.43; 95% confidence interval (CI) 0.14-1.35]. The probability of intubation, or intubation or death (secondary outcomes) was lower in the prone position group than in the usual care group (aOR 0.11; 95% CI 0.01-0.89 and aOR 0.09; 95% CI 0.01-0.76, respectively) in the whole study population and in the prespecified subgroup of patients with SpO2 ≥ 95% on inclusion (aOR 0.11; 95% CI 0.01-0.90, and aOR 0.09; 95% CI 0.03-0.27, respectively). CONCLUSIONS Awake prone position plus usual care in COVID-19 patients in medical wards did not decrease the composite outcome of need for NIV or intubation or death. Trial registration ClinicalTrials.gov Identifier: NCT04363463 . Registered 27 April 2020.
Collapse
Affiliation(s)
- Mai-Anh Nay
- Medical Intensive Care Unit, Centre Hospitalier Régional d'Orléans, 14, Avenue de l'hôpital, 45067, Orléans Cedex 2, France.
| | - Raphaël Hindre
- Department of Respiratory Medicine, Hôpital Européen Georges-Pompidou, Paris, France
- Innovative Therapies in Hemostasis, INSERM UMR S 1140, Biosurgical Research Lab (Carpentier Foundation), Université de Paris, Paris, France
| | - Christophe Perrin
- Department of Pneumology and Pneumo-Covid Unit, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Jérémy Clément
- Department of Internal Medicine and General Medicine, Centre Hospitalier Simone Veil, Blois, France
| | - Laurent Plantier
- Department of Pneumology and Respiratory Functional Testing, Bretonneau Hospital, CHRU de Tours, CEPR/INSERM UMR1100, University of Tours, Tours, France
| | - Aymeric Sève
- Department of Infectious and Tropical Diseases, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Sylvie Druelle
- Department of Pneumology, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Marine Morrier
- Department of Infectious Diseases, Centre Hospitalier Departmental de la Vendée, La Roche Sur Yon, France
| | - Jean-Baptiste Lainé
- Department of Infectious and Tropical Diseases, Le Mans Hospital, Le Mans, France
| | - Léa Colombain
- Department of Infectious and Tropical Diseases, Perpignan Hospital Centre, Perpignan, France
| | - Grégory Corvaisier
- Department of Internal Medicine, Centre Hospitalier Bretagne Atlantique, Vannes, France
| | - Nicolas Bizien
- Department of Pneumology, Centre Hospitalier Intercommunal de Cornouaille, Quimper, France
| | - Xavier Pouget-Abadie
- Department of Internal Medicine and Infectious Diseases, Groupement Hospitalier la Rochelle Ré Aunis, La Rochelle, France
| | - Adrien Bigot
- Department of Internal Medicine, Bretonneau Hospital, Tours, France
| | - Simon Jamard
- Department of Infectious Diseases, Bretonneau Hospital, University of Tours, Tours, France
| | - Elsa Nyamankolly
- Department of Internal Medicine and Infectious Diseases, Hospital Dax Côte D'argent, Dax, France
| | - Benjamin Planquette
- Department of Respiratory Medicine, Hôpital Européen Georges-Pompidou, Paris, France
- Innovative Therapies in Hemostasis, INSERM UMR S 1140, Biosurgical Research Lab (Carpentier Foundation), Université de Paris, Paris, France
| | - Guillaume Fossat
- Medical Intensive Care Unit, Centre Hospitalier Régional d'Orléans, 14, Avenue de l'hôpital, 45067, Orléans Cedex 2, France
| | - Thierry Boulain
- Medical Intensive Care Unit, Centre Hospitalier Régional d'Orléans, 14, Avenue de l'hôpital, 45067, Orléans Cedex 2, France
| |
Collapse
|
176
|
Anton DB, Galvez Bulhões Pedreira J, Zvirtes ML, Laufer SA, Ducati RG, Goettert M, Saraiva Macedo Timmers LF. Targeting SARS-CoV-2 Main Protease (MPro) with Kinase Inhibitors: A Promising Approach for Discovering Antiviral and Anti-inflammatory Molecules against SARS-CoV-2. J Chem Inf Model 2023. [PMID: 37329322 DOI: 10.1021/acs.jcim.3c00324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infected over 688 million people worldwide, causing public health concern and approximately 6.8 million deaths due to COVID-19. COVID-19, especially severe cases, is characterized by exacerbated lung inflammation with an increase of pro-inflammatory cytokines. In addition to antiviral drugs, there is a need for anti-inflammatory therapies to treat all phases of COVID-19. One of the most attractive drug targets for COVID-19 is the SARS-CoV-2 main protease (MPro), an enzyme responsible for cleaving polyproteins formed after the translation of viral RNA, which is essential for viral replication. MPro inhibitors, therefore, have the potential to stop viral replication and act as antiviral drugs. Considering that several kinase inhibitors are known for their action in inflammatory pathways, this could also be investigated toward a potential anti-inflammatory treatment for COVID-19. Therefore, the use of kinase inhibitors against SARS-CoV-2 MPro may be a promising strategy to find molecules with dual activity─antiviral and anti-inflammatory. Considering this, the potential of six kinase inhibitors against SARS-CoV-2 MPro were evaluated in silico and in vitro, including Baricitinib, Tofacitinib, Ruxolitinib, BIRB-796, Skepinone-L, and Sorafenib. To assess the inhibitory potential of the kinase inhibitors, a continuous fluorescent-based enzyme activity assay was optimized with SARS-CoV-2 MPro and MCA-AVLQSGFR-K(Dnp)-K-NH2 (substrate). BIRB-796 and Baricitinib were identified as inhibitors of SARS-CoV-2 MPro, presenting IC50 values of 7.99 and 25.31 μM, respectively. As they are also known for their anti-inflammatory action, both are prototype compounds with the potential to present antiviral and anti-inflammatory activity against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Débora Bublitz Anton
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
| | - Júlia Galvez Bulhões Pedreira
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen 72076, Germany
| | - Maria Luiza Zvirtes
- Department of Medicine, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
| | - Stefan A Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen 72076, Germany
| | - Rodrigo Gay Ducati
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
- Department of Medicine, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
| | - Márcia Goettert
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen 72076, Germany
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
| | - Luis Fernando Saraiva Macedo Timmers
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
- Department of Medicine, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado CEP 95914-014, Brazil
| |
Collapse
|
177
|
Foidart JM, Simon K, Utian WH, Mauvais-Jarvis F, Douxfils J, Dixon G, Barrington P. Estetrol Is Safe and Well Tolerated during Treatment of Hospitalized Men and Women with Moderate COVID-19 in a Randomized, Double-Blind Study. J Clin Med 2023; 12:3928. [PMID: 37373625 DOI: 10.3390/jcm12123928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Epidemiological data suggest that the severe acute respiratory syndrome coronavirus 2 infection rate is higher in women than in men, but the death rate is lower, while women (>50 years) on menopausal hormone therapy (MHT) have a higher survival rate than those not on MHT. Classical oral estrogen enhances the synthesis of coagulation markers and may increase the risk of thromboembolic events that are common in coronavirus disease 2019 (COVID-19). The favorable hemostatic profile of estetrol (E4) might be suitable for use in women who are receiving estrogen treatment and contract COVID-19. A multicenter, randomized, double-blind, placebo-controlled, phase 2 study (NCT04801836) investigated the efficacy, safety, and tolerability of E4 versus placebo in hospitalized patients with moderate COVID-19. Eligible postmenopausal women and men (aged ≥ 18 years old) were randomized to E4 15 mg or placebo, once daily for 21 days, in addition to the standard of care (SoC). The primary efficacy endpoint of improvement in COVID-19 (percentage of patients recovered at day 28) between the placebo and E4 arms was not met. E4 was well tolerated, with no safety signals or thromboembolic events, suggesting that postmenopausal women can safely continue E4-based therapy in cases of moderate COVID-19 managed with SoC.
Collapse
Affiliation(s)
- Jean Michel Foidart
- Mithra Pharmaceuticals, 4000 Liège, Belgium
- Department of Obstetrics and Gynecology, University of Liège, 4000 Liège, Belgium
| | - Krzysztof Simon
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51149 Wrocław, Poland
| | - Wulf H Utian
- Department of Reproductive Biology, Case Western Reserve Medical School, Cleveland, OH 44106, USA
| | - Franck Mauvais-Jarvis
- Department of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jonathan Douxfils
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center, Faculty of Medicine, University of Namur, 5000 Namur, Belgium
- QUALIblood s.a., 5000 Namur, Belgium
| | | | | |
Collapse
|
178
|
Jiang SL, Hu ZY, Wang WJ, Hu L, Li L, Kou SB, Shi JH. Investigation on the binding behavior of human α1-acid glycoprotein with Janus Kinase inhibitor baricitinib: Multi-spectroscopic and molecular simulation methodologies. Int J Biol Macromol 2023:125096. [PMID: 37285878 DOI: 10.1016/j.ijbiomac.2023.125096] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
Baricitinib is a Janus Kinase (JAK) inhibitor that is primarily used to treat moderately to severely active rheumatoid arthritis in adults and has recently been reported for the treatment of patients with severe COVID-19. This paper describes the investigation of the binding behavior of baricitinib to human α1-acid glycoprotein (HAG) employing a variety of spectroscopic techniques, molecular docking and dynamics simulations. Baricitinib can quench the fluorescence from amino acids in HAG through a mix of dynamic and static quenching, according to steady-state fluorescence and UV spectra observations, but it is mainly static quenching at low concentration. The binding constant (Kb) of baricitinib to HAG at 298 K was at the level of 104 M-1, indicating a moderate affinity of baricitinib to HAG. Hydrogen bonding and hydrophobic interactions conducted the main effect, according to thermodynamic characteristics, competition studies between ANS and sucrose, and molecular dynamics simulations. For the change in HAG conformation, the results of multiple spectra showed that baricitinib was able to alter the secondary structure of HAG as well as increase the polarity of the microenvironment around the Trp amino acid. Furthermore, the binding behavior of baricitinib to HAG was investigated by molecular docking and molecular dynamics simulations, which validated experimental results. Also explored is the influence of K+, Co2+, Ni2+, Ca2+, Fe3+, Zn2+, Mg2+ and Cu2+plasma on binding affinity.
Collapse
Affiliation(s)
- Shao-Liang Jiang
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China.
| | - Zhe-Ying Hu
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wan-Jun Wang
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Lu Hu
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Li Li
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Song-Bo Kou
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jie-Hua Shi
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
179
|
Fadanni GP, Calixto JB. Recent progress and prospects for anti-cytokine therapy in preclinical and clinical acute lung injury. Cytokine Growth Factor Rev 2023; 71-72:13-25. [PMID: 37481378 DOI: 10.1016/j.cytogfr.2023.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/24/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous cause of respiratory failure that has a rapid onset, a high mortality rate, and for which there is no effective pharmacological treatment. Current evidence supports a critical role of excessive inflammation in ARDS, resulting in several cytokines, cytokine receptors, and proteins within their downstream signalling pathways being putative therapeutic targets. However, unsuccessful trials of anti-inflammatory drugs have thus far hindered progress in the field. In recent years, the prospects of precision medicine and therapeutic targeting of cytokines coevolving into effective treatments have gained notoriety. There is an optimistic and growing understanding of ARDS subphenotypes as well as advances in treatment strategies and clinical trial design. Furthermore, large trials of anti-cytokine drugs in patients with COVID-19 have provided an unprecedented amount of information that could pave the way for therapeutic breakthroughs. While current clinical and nonclinical ARDS research suggest relatively limited potential in monotherapy with anti-cytokine drugs, combination therapy has emerged as an appealing strategy and may provide new perspectives on finding safe and effective treatments. Accurate evaluation of these drugs, however, also relies on well-founded experimental research and the implementation of biomarker-guided stratification in future trials. In this review, we provide an overview of anti-cytokine therapy for acute lung injury and ARDS, highlighting the current preclinical and clinical evidence for targeting the main cytokines individually and the therapeutic prospects for combination therapy.
Collapse
Affiliation(s)
- Guilherme Pasetto Fadanni
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| | - João Batista Calixto
- Centre of Innovation and Preclinical Studies (CIEnP), Florianópolis, Santa Catarina, Brazil; Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
180
|
McManus D, Davis MW, Ortiz A, Britto-Leon C, Dela Cruz CS, Topal JE. Immunomodulatory Agents for Coronavirus Disease-2019 Pneumonia. Clin Chest Med 2023; 44:299-319. [PMID: 37085221 PMCID: PMC9678826 DOI: 10.1016/j.ccm.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Morbidity and mortality from COVID-19 is due to severe inflammation and end-organ damage caused by a hyperinflammatory response. Multiple immunomodulatory agents to attenuate this response have been studied. Corticosteroids, specifically dexamethasone, have been shown to reduce mortality in hospitalized patients who require supplemental oxygen. Interleukin-6 antagonist, tocilizimab, and Janus kinase inhibitors have also been shown to reduce mortality. However, patients who have severe pulmonary end-organ damage requiring mechanical ventilation or extracorporeal membrane oxygenation appear not to benefit from immunomodulatory therapies. This highlights the importance of appropriate timing to initiate immunomodulatory therapies in the management of severe COVID-19 disease.
Collapse
Affiliation(s)
- Dayna McManus
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| | - Matthew W Davis
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA
| | - Alex Ortiz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Clemente Britto-Leon
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Charles S Dela Cruz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Jeffrey E Topal
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| |
Collapse
|
181
|
Li G, Hilgenfeld R, Whitley R, De Clercq E. Therapeutic strategies for COVID-19: progress and lessons learned. Nat Rev Drug Discov 2023; 22:449-475. [PMID: 37076602 PMCID: PMC10113999 DOI: 10.1038/s41573-023-00672-y] [Citation(s) in RCA: 320] [Impact Index Per Article: 160.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 04/21/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has stimulated tremendous efforts to develop therapeutic strategies that target severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and/or human proteins to control viral infection, encompassing hundreds of potential drugs and thousands of patients in clinical trials. So far, a few small-molecule antiviral drugs (nirmatrelvir-ritonavir, remdesivir and molnupiravir) and 11 monoclonal antibodies have been marketed for the treatment of COVID-19, mostly requiring administration within 10 days of symptom onset. In addition, hospitalized patients with severe or critical COVID-19 may benefit from treatment with previously approved immunomodulatory drugs, including glucocorticoids such as dexamethasone, cytokine antagonists such as tocilizumab and Janus kinase inhibitors such as baricitinib. Here, we summarize progress with COVID-19 drug discovery, based on accumulated findings since the pandemic began and a comprehensive list of clinical and preclinical inhibitors with anti-coronavirus activities. We also discuss the lessons learned from COVID-19 and other infectious diseases with regard to drug repurposing strategies, pan-coronavirus drug targets, in vitro assays and animal models, and platform trial design for the development of therapeutics to tackle COVID-19, long COVID and pathogenic coronaviruses in future outbreaks.
Collapse
Affiliation(s)
- Guangdi Li
- Xiangya School of Public Health, Central South University; Hunan Children's Hospital, Changsha, China.
| | - Rolf Hilgenfeld
- Institute of Molecular Medicine & German Center for Infection Research (DZIF), University of Lübeck, Lübeck, Germany.
| | - Richard Whitley
- Department of Paediatrics, Microbiology, Medicine and Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Erik De Clercq
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
182
|
Sacchi A, Giannessi F, Sabatini A, Percario ZA, Affabris E. SARS-CoV-2 Evasion of the Interferon System: Can We Restore Its Effectiveness? Int J Mol Sci 2023; 24:ijms24119353. [PMID: 37298304 DOI: 10.3390/ijms24119353] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Type I and III Interferons (IFNs) are the first lines of defense in microbial infections. They critically block early animal virus infection, replication, spread, and tropism to promote the adaptive immune response. Type I IFNs induce a systemic response that impacts nearly every cell in the host, while type III IFNs' susceptibility is restricted to anatomic barriers and selected immune cells. Both IFN types are critical cytokines for the antiviral response against epithelium-tropic viruses being effectors of innate immunity and regulators of the development of the adaptive immune response. Indeed, the innate antiviral immune response is essential to limit virus replication at the early stages of infection, thus reducing viral spread and pathogenesis. However, many animal viruses have evolved strategies to evade the antiviral immune response. The Coronaviridae are viruses with the largest genome among the RNA viruses. Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) pandemic. The virus has evolved numerous strategies to contrast the IFN system immunity. We intend to describe the virus-mediated evasion of the IFN responses by going through the main phases: First, the molecular mechanisms involved; second, the role of the genetic background of IFN production during SARS-CoV-2 infection; and third, the potential novel approaches to contrast viral pathogenesis by restoring endogenous type I and III IFNs production and sensitivity at the sites of infection.
Collapse
Affiliation(s)
- Alessandra Sacchi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Flavia Giannessi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Andrea Sabatini
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Zulema Antonia Percario
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Elisabetta Affabris
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| |
Collapse
|
183
|
Völkel S, Tarawneh TS, Sacher L, Bhagwat AM, Karim I, Mack HID, Wiesmann T, Beutel B, Hoyer J, Keller C, Renz H, Burchert A, Neubauer A, Graumann J, Skevaki C, Mack EKM. Serum proteomics hint at an early T-cell response and modulation of SARS-CoV-2-related pathogenic pathways in COVID-19-ARDS treated with Ruxolitinib. Front Med (Lausanne) 2023; 10:1176427. [PMID: 37293294 PMCID: PMC10244732 DOI: 10.3389/fmed.2023.1176427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 06/10/2023] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) in corona virus disease 19 (COVID-19) is triggered by hyperinflammation, thus providing a rationale for immunosuppressive treatments. The Janus kinase inhibitor Ruxolitinib (Ruxo) has shown efficacy in severe and critical COVID-19. In this study, we hypothesized that Ruxo's mode of action in this condition is reflected by changes in the peripheral blood proteome. Methods This study included 11 COVID-19 patients, who were treated at our center's Intensive Care Unit (ICU). All patients received standard-of-care treatment and n = 8 patients with ARDS received Ruxo in addition. Blood samples were collected before (day 0) and on days 1, 6, and 10 of Ruxo treatment or, respectively, ICU admission. Serum proteomes were analyzed by mass spectrometry (MS) and cytometric bead array. Results Linear modeling of MS data yielded 27 significantly differentially regulated proteins on day 1, 69 on day 6 and 72 on day 10. Only five factors (IGLV10-54, PSMB1, PGLYRP1, APOA5, WARS1) were regulated both concordantly and significantly over time. Overrepresentation analysis revealed biological processes involving T-cells only on day 1, while a humoral immune response and complement activation were detected at day 6 and day 10. Pathway enrichment analysis identified the NRF2-pathway early under Ruxo treatment and Network map of SARS-CoV-2 signaling and Statin inhibition of cholesterol production at later time points. Conclusion Our results indicate that the mechanism of action of Ruxo in COVID-19-ARDS can be related to both known effects of this drug as a modulator of T-cells and the SARS-CoV-2-infection.
Collapse
Affiliation(s)
- Sara Völkel
- Institute of Laboratory Medicine, Philipps-University Marburg, Marburg, Germany
| | - Thomas S. Tarawneh
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| | - Laura Sacher
- Institute of Laboratory Medicine, Philipps-University Marburg, Marburg, Germany
| | - Aditya M. Bhagwat
- Institute of Translational Proteomics, Philipps-University Marburg, Marburg, Germany
| | - Ihab Karim
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| | - Hildegard I. D. Mack
- Institute for Biomedical Aging Research, Leopold-Franzens-Universität Innsbruck, Innsbruck, Austria
| | - Thomas Wiesmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Diakonie-Klinikum Schwäbisch Hall, Schwäbisch Hall, Germany
| | - Björn Beutel
- Department of Pulmonary and Critical Care Medicine, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
- German Center for Lung Research (DZL), Member of the Universities of Gießen and Marburg Lung Center, Gießen, Germany
| | - Joachim Hoyer
- Department of Nephrology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| | - Christian Keller
- Institute of Virology, Philipps-University Marburg, Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps-University Marburg, Marburg, Germany
- German Center for Lung Research (DZL), Member of the Universities of Gießen and Marburg Lung Center, Gießen, Germany
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Philipps-University Marburg, Marburg, Germany
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps-University Marburg, Marburg, Germany
- German Center for Lung Research (DZL), Member of the Universities of Gießen and Marburg Lung Center, Gießen, Germany
| | - Elisabeth K. M. Mack
- Department of Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
184
|
Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023; 8:204. [PMID: 37208335 DOI: 10.1038/s41392-023-01468-7] [Citation(s) in RCA: 202] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved mechanism of transmembrane signal transduction that enables cells to communicate with the exterior environment. Various cytokines, interferons, growth factors, and other specific molecules activate JAK-STAT signaling to drive a series of physiological and pathological processes, including proliferation, metabolism, immune response, inflammation, and malignancy. Dysregulated JAK-STAT signaling and related genetic mutations are strongly associated with immune activation and cancer progression. Insights into the structures and functions of the JAK-STAT pathway have led to the development and approval of diverse drugs for the clinical treatment of diseases. Currently, drugs have been developed to mainly target the JAK-STAT pathway and are commonly divided into three subtypes: cytokine or receptor antibodies, JAK inhibitors, and STAT inhibitors. And novel agents also continue to be developed and tested in preclinical and clinical studies. The effectiveness and safety of each kind of drug also warrant further scientific trials before put into being clinical applications. Here, we review the current understanding of the fundamental composition and function of the JAK-STAT signaling pathway. We also discuss advancements in the understanding of JAK-STAT-related pathogenic mechanisms; targeted JAK-STAT therapies for various diseases, especially immune disorders, and cancers; newly developed JAK inhibitors; and current challenges and directions in the field.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
185
|
La Rocca G, Ferro F, Sambataro G, Elefante E, Fonzetti S, Fulvio G, Navarro IC, Mosca M, Baldini C. Primary-Sjögren's-Syndrome-Related Interstitial Lung Disease: A Clinical Review Discussing Current Controversies. J Clin Med 2023; 12:3428. [PMID: 37240535 PMCID: PMC10218845 DOI: 10.3390/jcm12103428] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Lung involvement, especially interstitial lung disease, is a potentially severe extra-glandular manifestation of Primary Sjogren's Syndrome (pSS-ILD). ILD can manifest either as a late complication of pSS or anticipate sicca symptoms, likely reflecting two different patho-physiological entities. Presence of lung involvement in pSS subjects can remain subclinical for a long time; therefore, patients should be actively screened, and lung ultrasound is currently being investigated as a potential low cost, radiation-free, easily repeatable screening tool for detection of ILD. In contrast, rheumatologic evaluation, serology testing, and minor salivary gland biopsy are crucial for the recognition of pSS in apparently idiopathic ILD patients. Whether the HRCT pattern influences prognosis and treatment response in pSS-ILD is not clear; a UIP pattern associated with a worse prognosis in some studies, but not in others. Many aspects of pSS-ILD, including its actual prevalence, association with specific clinical-serological characteristics, and prognosis, are still debated by the current literature, likely due to poor phenotypic stratification of patients in clinical studies. In the present review, we critically discuss these and other clinically relevant "hot topics" in pSS-ILD. More specifically, after a focused discussion, we compiled a list of questions regarding pSS-ILD that, in our opinion, are not easily answered by the available literature. We subsequently tried to formulate adequate answers on the basis of an extensive literature search and our clinical experience. At the same, we highlighted different issues that require further investigation.
Collapse
Affiliation(s)
- Gaetano La Rocca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Francesco Ferro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Gianluca Sambataro
- Department of Clinical and Experimental Medicine, Regional Referral Centre for Rare Lung Diseases, A.O.U. Policlinico “G. Rodolico-San Marco”, University of Catania, Via Santa Sofia 78, 95124 Catania, Italy;
- Artroreuma S.R.L., Rheumatology Outpatient Clinic Associated with the National Health System, Corso S. Vito 53, 95030 Catania, Italy
| | - Elena Elefante
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Silvia Fonzetti
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Giovanni Fulvio
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Inmaculada C. Navarro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy; (F.F.); (E.E.); (S.F.); (G.F.); (I.C.N.); (M.M.); (C.B.)
| |
Collapse
|
186
|
Phan A, Avila H, MacKay JA. Biomimetic SARS-CoV-2 Spike Protein Nanoparticles. Biomacromolecules 2023; 24:2030-2041. [PMID: 37001147 PMCID: PMC10084924 DOI: 10.1021/acs.biomac.2c01465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/21/2023] [Indexed: 04/03/2023]
Abstract
COVID-19 is an infectious respiratory disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus contains a crucial coat protein that engages with target cells via a receptor binding domain (RBD) on its spike protein. To better study the RBD and its therapeutic opportunities, we genetically engineered a simple fusion with a thermo-responsive elastin-like polypeptide (ELP). These fusions express in Escherichia coli at a high yield in the soluble fraction and were easily purified using ELP-mediated phase separation (79 mg/L culture). Interestingly, they assembled peptide-based nanoparticles (Rh = 71.4 nm), which was attributed to oligomerization of RBDs (25.3 kDa) counterbalanced by steric stabilization by a soluble ELP (73.4 kDa). To investigate their biophysical properties, we explored the size, shape, and binding affinity for the human angiotensin-converting enzyme 2 (hACE2) and cellular uptake. Biomimetic nanoparticles such as these may enable future strategies to target the same cells, tissues, and cell-surface receptors as those harnessed by SARS-CoV-2.
Collapse
Affiliation(s)
- Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
| | - Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
- Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1450 San Pablo Street, Los Angeles, CA 90033, United States
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA 90089, United States
| |
Collapse
|
187
|
Lam V, Hu KM. Management of Coronavirus Disease-2019 Infection in Pregnancy. Emerg Med Clin North Am 2023; 41:307-322. [PMID: 37024166 PMCID: PMC9755010 DOI: 10.1016/j.emc.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the majority of pregnant patients who contract severe acute respiratory syndrome coronavirus 2 will have a mild course of illness, pregnant patients with coronavirus disease-2019 are more likely than their nonpregnant counterparts to develop a severe illness with an increased risk of poor maternal and fetal outcomes. Although the extent of research in this specific patient population remains limited, there are tenets of care with which physicians and other providers must be familiar to increase the chances of better outcomes for the two patients in their care.
Collapse
Affiliation(s)
- Vivian Lam
- Department of Internal Medicine, Section of Critical Care Medicine, Advocate Christ Medical Center, 4440 West 95th Street, Suite AIP, Oak Lawn, IL 60453, USA
| | - Kami M Hu
- Department of Internal Medicine, Section of Critical Care Medicine, Advocate Christ Medical Center, 4440 West 95th Street, Suite AIP, Oak Lawn, IL 60453, USA; Division of Pulmonary & Critical Care, Department of Internal Medicine, University of Maryland, School of Medicine, 110 South Paca Street, 6th Floor Suite 200, Baltimore, MD 21201, USA.
| |
Collapse
|
188
|
Kumar NR, Karanam VC, Kumar S, Kumar SD. Convalescent Plasma Therapy in Late-State, Severe COVID-19 Infection. South Med J 2023; 116:427-433. [PMID: 37137479 PMCID: PMC10143395 DOI: 10.14423/smj.0000000000001546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Current evidence favors plasma to be effective against coronavirus disease 2019 (COVID-19) in critically ill patients in the early stages of infection. We investigated the safety and efficacy of convalescent plasma in specifically late-stage (designated as after 2 weeks of hospital admission) severe COVID-19 infection. We also conducted a literature review on the late-stage use of plasma in COVID-19. METHODS This case series examined eight COVID-19 patients admitted to the intensive care unit (ICU) who met criteria for severe or life-threatening complications. Each patient received one dose (200 mL) of plasma. Clinical information was gathered in intervals of 1 day pretransfusion and 1 hour, 3 days, and 7 days posttransfusion. The primary outcome was effectiveness of plasma transfusion, measured by clinical improvement, laboratory parameters, and all-cause mortality. RESULTS Eight ICU patients received plasma late in the course of COVID-19 infection, on average at 16.13 days postadmission. On the day before transfusion, the averaged initial Sequential Organ Failure Assessment (SOFA) score, PaO2:FiO2 ratio, Glasgow Coma Scale (GCS), and lymphocyte count were 6.5, 228.03, 8.63, and 1.19, respectively. Three days after plasma treatment, the group averages for the SOFA score (4.86), PaO2:FiO2 ratio (302.73), GCS (9.29), and lymphocyte count (1.75) improved. Although the mean GCS improved to 10.14 by posttransfusion day 7, the other means marginally worsened with an SOFA score of 5.43, a PaO2:FiO2 ratio of 280.44, and a lymphocyte count of 1.71. Clinical improvement was noted in six patients who were discharged from the ICU. CONCLUSIONS This case series provides evidence that convalescent plasma may be safe and effective in late-stage, severe COVID-19 infection. Results showed clinical improvement posttransfusion as well as decreased all-cause mortality in comparison to pretransfusion predicted mortality. Randomized controlled trials are needed to conclusively determine benefits, dosage, and timing of treatment.
Collapse
Affiliation(s)
- Neil R Kumar
- From Internal Medicine, University of Miami/Jackson Memorial Hospital, Miami, Florida
| | - Veena C Karanam
- the University of Miami Miller School of Medicine, Miami, Florida
| | - Shari Kumar
- Columbia University College of Dental Medicine, New York, New York
| | - Sunil D Kumar
- the Pulmonary/Critical Care, Broward Health Medical Center, Ft Lauderdale, Florida
| |
Collapse
|
189
|
Sweeney DA, Tuyishimire B, Ahuja N, Beigel JH, Beresnev T, Cantos VD, Castro JG, Cohen SH, Cross K, Dodd LE, Erdmann N, Fung M, Ghazaryan V, George SL, Grimes KA, Hynes NA, Julian KG, Kandiah S, Kim HJ, Levine CB, Lindholm DA, Lye DC, Maves RC, Oh MD, Paules C, Rapaka RR, Short WR, Tomashek KM, Wolfe CR, Kalil AC. Baricitinib Treatment of Coronavirus Disease 2019 Is Associated With a Reduction in Secondary Infections. Open Forum Infect Dis 2023; 10:ofad205. [PMID: 37206623 PMCID: PMC10191442 DOI: 10.1093/ofid/ofad205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
We performed a secondary analysis of the National Institutes of Health-sponsored Adaptive COVID-19 Treatment Trial (ACTT-2) randomized controlled trial and found that baricitinib was associated with a 50% reduction in secondary infections after controlling for baseline and postrandomization patient characteristics. This finding provides a novel mechanism of benefit for baricitinib and supports the safety profile of this immunomodulator for the treatment of coronavirus disease 2019.
Collapse
Affiliation(s)
- Daniel A Sweeney
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Neera Ahuja
- Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - John H Beigel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tatiana Beresnev
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jose G Castro
- Department of Medicine, University of Miami, Miami, FL, USA
| | - Stuart H Cohen
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | | | - Lori E Dodd
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nathan Erdmann
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Monica Fung
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Varduhi Ghazaryan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah L George
- Department of Medicine, Saint Louis University and St. Louis VA Medical Center, Saint Louis, MO, USA
| | - Kevin A Grimes
- Department of Medicine, Houston Methodist, Houston, TX, USA
| | - Noreen A Hynes
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen G Julian
- Department of Medicine, Pennsylvania State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | | | - Hannah Jang Kim
- Community Health Systems Department, University of California San Francisco, San Francisco, CA, USA
- Department of Nursing, Kaiser Permanente National Patient Care Services, Oakland, CA, USA
| | - Corri B Levine
- Department of Internal Medicine Galveston, University of Texas Medical Branch, TX, USA
| | - David A Lindholm
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Medicine, Brooke Army Medical Center, Joint Base San Antonio-Ft Sam Houston, TX, USA
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
- Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Ryan C Maves
- Departments of Internal Medicine and Anesthesiology, Wake Forest University, Winston-Salem, NC, USA
| | - Myoung-don Oh
- Department of Internal Medicine, Seoul National University Hospital College of Medicine, Seoul, Korea
| | - Catharine Paules
- Department of Medicine, Pennsylvania State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Rekha R Rapaka
- Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Willam R Short
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kay M Tomashek
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Andre C Kalil
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
190
|
Morishita M, Hojo M. Treatment options for patients with severe COVID-19. Glob Health Med 2023; 5:99-105. [PMID: 37128231 PMCID: PMC10130548 DOI: 10.35772/ghm.2023.01024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/03/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has affected the world for over 3 years. Treatment options have improved substantially during this period, including antiviral drugs, antibody drugs, immune-based agents, and vaccination. While these improvements have reduced mortality rates in patients with COVID-19, some patients still develop severe illness. In this review, we aimed to provide an overview of treatments for patients with severe COVID-19 from study reports and clinical experience. We discussed the treatments from two perspectives: respiratory care and drug treatments. In the respiratory care section, we discussed the usefulness of high-flow nasal cannula therapy and non-invasive ventilation as an alternative to invasive ventilation. In the drug treatments section, we focused on three classes for severe COVID-19 treatment: antiviral drugs, immune-based agents, and anticoagulation therapy. We did not discuss antibody drugs and vaccination, as they are not used for severe COVID-19 treatment.
Collapse
Affiliation(s)
| | - Masayuki Hojo
- Address correspondence to:Masayuki Hojo, Department of Respiratory Medicine, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan.
| |
Collapse
|
191
|
Koh JY, Ko JH, Lim SY, Bae S, Huh K, Cho SY, Kang CI, Chung DR, Chung CR, Kim SH, Peck KR, Lee JS. Triple immune modulator therapy for aberrant hyperinflammatory responses in severe COVID-19. Clin Immunol 2023; 251:109628. [PMID: 37119951 PMCID: PMC10139747 DOI: 10.1016/j.clim.2023.109628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
A dysregulated hyperinflammatory response is a key pathogenesis of severe COVID-19, but optimal immune modulator treatment has not been established. To evaluate the clinical effectiveness of double (glucocorticoids and tocilizumab) and triple (plus baricitinib) immune modulator therapy for severe COVID-19, a retrospective cohort study was conducted. For the immunologic investigation, a single-cell RNA sequencing analysis was performed in serially collected PBMCs and neutrophil specimens. Triple immune modulator therapy was a significant factor in a multivariable analysis for 30-day recovery. In the scRNA-seq analysis, type I and II IFN response-related pathways were suppressed by GC, and the IL-6-associated signature was additionally downregulated by TOC. Adding BAR to GC and TOC distinctly downregulated the ISGF3 cluster. Adding BAR also regulated the pathologically activated monocyte and neutrophil subpopulation induced by aberrant IFN signals. Triple immune modulator therapy in severe COVID-19 improved 30-day recovery through additional regulation of the aberrant hyperinflammatory immune response.
Collapse
Affiliation(s)
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - So Yun Lim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seongman Bae
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun Young Cho
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-In Kang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Doo Ryeon Chung
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Chi Ryang Chung
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Jeong Seok Lee
- Genome Insight, Inc., San Diego, La Jolla, CA, USA; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
192
|
Rezapour A, Behroozi Z, Nasirzadeh M, Rezaeian M, Barzegar M, Tashakori-Miyanroudi M, Sayyad A, Souresrafil A. Cost-effectiveness of remdesivir for the treatment of hospitalized patients with COVID-19: a systematic review. Infect Dis Poverty 2023; 12:39. [PMID: 37081575 PMCID: PMC10116457 DOI: 10.1186/s40249-023-01092-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/04/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Remdesivir is being studied and used to treat coronavirus disease 2019 (COVID-19). This study aimed to systematically identify, critically evaluate, and summarize the findings of the studies on the cost-effectiveness of remdesivir in the treatment of hospitalized patients with COVID-19. METHODS In this systematic review, PubMed, EMBASE, Web of Science, SCOPUS, and the Cochrane Library were searched for studies published between 2019 and 2022. We included all full economic evaluations of remdesivir for the treatment of hospitalized patients with COVID-19. Data were summarized in a structured and narrative manner. RESULTS Out of 616 articles obtained in this literature search, 12 studies were included in the final analysis. The mean score of the Quality of Health Economic Studies (QHES) for the studies was 87.66 (high quality). All studies were conducted in high-income countries (eight studies in the USA and one study in England), except for three studies from middle-to-high-income countries (China, South Africa, and Turkey). Six studies conducted their economic analysis in terms of a health system perspective; five studies conducted their economic analysis from a payer perspective; three studies from the perspective of a health care provider. The results of five studies showed that remdesivir was cost-effective compared to standard treatment. Furthermore, the therapeutic strategy of combining remdesivir with baricitinib was cost-effective compared to remdesivir alone. CONCLUSIONS Based on the results of the present study, remdesivir appears to be cost-effective in comparison with the standard of care in China, Turkey, and South Africa. Studies conducted in the United States show conflicting results, and combining remdesivir with baricitinib is cost-effective compared with remdesivir alone. However, the cost-effectiveness of remdesivir in low-income countries remains unknown. Thus, more studies in different countries are required to determine the cost-effectiveness of this drug.
Collapse
Affiliation(s)
- Aziz Rezapour
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mostafa Nasirzadeh
- Department of Health Education and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohsen Rezaeian
- Department of Epidemiology and Biostatistics, School of Medicine, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Barzegar
- Department of English Language Teaching, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Tashakori-Miyanroudi
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdollah Sayyad
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Aghdas Souresrafil
- Department of Health Services and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
193
|
Hirasawa Y, Terada J, Shionoya Y, Fujikawa A, Isaka Y, Takeshita Y, Kinouchi T, Koshikawa K, Tajima H, Kinoshita T, Tada Y, Tatsumi K, Tsushima K. Combination therapy with predicted body weight-based dexamethasone, remdesivir, and baricitinib in patients with COVID-19 pneumonia: A single-center retrospective cohort study during 5th wave in Japan. Respir Investig 2023; 61:438-444. [PMID: 37119744 PMCID: PMC10110979 DOI: 10.1016/j.resinv.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Dexamethasone, remdesivir, and baricitinib reduce mortality in patients with coronavirus disease 2019 (COVID-19). A single-arm study using combination therapy with all three drugs reported low mortality in patients with severe COVID-19. In this clinical setting, whether dexamethasone administered as a fixed dose of 6 mg has sufficient inflammatory modulation effects of reducing lung injury has been debated. METHODS This single-center retrospective study was conducted to compare the treatment strategies/management in different time periods. A total of 152 patients admitted with COVID-19 pneumonia who required oxygen therapy were included in this study. A predicted body weight (PBW)-based dose of dexamethasone with remdesivir and baricitinib was administered between May and June 2021. After this period, patients were administered a fixed dose of dexamethasone at 6.6 mg/day between July and August 2021. The additional respiratory support frequency of high-flow nasal cannula, noninvasive ventilation, and mechanical ventilation was analyzed. Moreover, the Kaplan-Meier method was used to analyze the duration of oxygen therapy and the 30-day discharge alive rate, and they were compared using the log-rank test. RESULTS Intervention and prognostic comparisons were performed in 64 patients with PBW-based and 88 with fixed-dose groups. The frequency of infection or additional respiratory support did not differ statistically. The cumulative incidence of being discharged alive or oxygen-free rate within 30 days did not differ between the groups. CONCLUSIONS In patients with COVID-19 pneumonia who required oxygen therapy, combination therapy with PBW-based dexamethasone, remdesivir, and baricitinib might not shorten the hospital stay's length or oxygen therapy's duration.
Collapse
Affiliation(s)
- Yasutaka Hirasawa
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan.
| | - Jiro Terada
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Yu Shionoya
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Atsushi Fujikawa
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Yuri Isaka
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan; Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Yuichiro Takeshita
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Toru Kinouchi
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan; Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Ken Koshikawa
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Hiroshi Tajima
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Taku Kinoshita
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Yuji Tada
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan
| | - Koichiro Tatsumi
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan; Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Kenji Tsushima
- Department of Pulmonary Medicine, International University of Health and Welfare, Narita Hospital, Hatakeda 852, Narita, Chiba, 286-0124, Japan.
| |
Collapse
|
194
|
Sun J, Yang ZD, Xie X, Li L, Zeng HS, Gong B, Xu JQ, Wu JH, Qu BB, Song GW. Clinical application of SARS-CoV-2 antibody detection and monoclonal antibody therapies against COVID-19. World J Clin Cases 2023; 11:2168-2180. [PMID: 37122515 PMCID: PMC10131020 DOI: 10.12998/wjcc.v11.i10.2168] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/17/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The purpose of this study was to investigate the clinical application of severe acute respiratory distress syndrome coronavirus-2 (SARS-CoV-2) specific antibody detection and anti-SARS-CoV-2 specific monoclonal antibodies (mAbs) in the treatment of coronavirus infectious disease 2019 (COVID-19). The dynamic changes of SARS-CoV-2 specific antibodies during COVID-19 were studied. Immunoglobulin M (IgM) appeared earlier and lasted for a short time, while immunoglobulin G (IgG) appeared later and lasted longer. IgM tests can be used for early diagnosis of COVID-19, and IgG tests can be used for late diagnosis of COVID-19 and identification of asymptomatic infected persons. The combination of antibody testing and nucleic acid testing, which complement each other, can improve the diagnosis rate of COVID-19. Monoclonal anti-SARS-CoV-2 specific antibodies can be used to treat hospitalized severe and critically ill patients and non-hospitalized mild to moderate COVID-19 patients. COVID-19 convalescent plasma, highly concentrated immunoglobulin, and anti-SARS-CoV-2 specific mAbs are examples of anti-SARS-CoV-2 antibody products. Due to the continuous emergence of mutated strains of the novel coronavirus, especially omicron, its immune escape ability and infectivity are enhanced, making the effects of authorized products reduced or invalid. Therefore, the optimal application of anti-SARS-CoV-2 antibody products (especially anti-SARS-CoV-2 specific mAbs) is more effective in the treatment of COVID-19 and more conducive to patient recovery.
Collapse
Affiliation(s)
- Jin Sun
- Medical Innovation Research Office, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
| | - Zhen-Dong Yang
- Department of Respiratory, Beijing Jindu Children Hospital, Beijing 102208, China
- Innovative Medicine Working Committee of the Chinese Society of Water Resources and Electric Power Medical Science and Technology, Beijing 100053, China
- Department of Pediatrics, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
| | - Xiong Xie
- Department of Pediatrics, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
| | - Li Li
- Department of Intensive Care, First Clinical Medical College of Three Gorges University, Yichang 443000, Hubei Province, China
| | - Hua-Song Zeng
- Department of Allergy Immunology and Rheumatology, Guangzhou Children's Hospital, Women's and Children's Medical Center Affiliated with Guangzhou Medical University, Guangzhou 510000, Guangdong Province, China
| | - Bo Gong
- Central Laboratory, Shanghai Changning District Maternal and Child Health, Maternal and Child Health Hospital Affiliated with Shanghai East China Normal University, Shanghai 210000, China
| | - Jian-Qiang Xu
- Department of Respiratory and Critical Care Medicine, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
| | - Ji-Hong Wu
- School of Clinical Medicine, Beijing Tsinghua Chang Gung Hospital Affiliated to Tsinghua University, Beijing 102218, China
| | - Bei-Bei Qu
- Medical Innovation Research Office, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
| | - Guo-Wei Song
- Department of Emergency, Children's Hospital Affiliated with Beijing Capital Institute of Pediatrics, Beijing 100020, China
| |
Collapse
|
195
|
Cable J, Sun J, Cheon IS, Vaughan AE, Castro IA, Stein SR, López CB, Gostic KM, Openshaw PJM, Ellebedy AH, Wack A, Hutchinson E, Thomas MM, Langlois RA, Lingwood D, Baker SF, Folkins M, Foxman EF, Ward AB, Schwemmle M, Russell AB, Chiu C, Ganti K, Subbarao K, Sheahan TP, Penaloza-MacMaster P, Eddens T. Respiratory viruses: New frontiers-a Keystone Symposia report. Ann N Y Acad Sci 2023; 1522:60-73. [PMID: 36722473 PMCID: PMC10580159 DOI: 10.1111/nyas.14958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Respiratory viruses are a common cause of morbidity and mortality around the world. Viruses like influenza, RSV, and most recently SARS-CoV-2 can rapidly spread through a population, causing acute infection and, in vulnerable populations, severe or chronic disease. Developing effective treatment and prevention strategies often becomes a race against ever-evolving viruses that develop resistance, leaving therapy efficacy either short-lived or relevant for specific viral strains. On June 29 to July 2, 2022, researchers met for the Keystone symposium "Respiratory Viruses: New Frontiers." Researchers presented new insights into viral biology and virus-host interactions to understand the mechanisms of disease and identify novel treatment and prevention approaches that are effective, durable, and broad.
Collapse
Affiliation(s)
| | - Jie Sun
- Division of Pulmonary and Critical Medicine, Department of Medicine; Department of Immunology; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Carter Immunology Center and Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - In Su Cheon
- Division of Pulmonary and Critical Medicine, Department of Medicine; Department of Immunology; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Carter Immunology Center and Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew E Vaughan
- University of Pennsylvania School of Veterinary Medicine, Biomedical Sciences, Philadelphia, Pennsylvania, USA
| | - Italo A Castro
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, São Paulo, Brazil
| | - Sydney R Stein
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center and Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Carolina B López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St Louis, Missouri, USA
| | - Katelyn M Gostic
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
| | | | - Ali H Ellebedy
- Department of Pathology and Immunology; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs; and Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, USA
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | | | | | - Ryan A Langlois
- Center for Immunology and Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, USA
| | - Steven F Baker
- Lovelace Biomedical Research Institute, Albuquerque, New Mexico, USA
| | - Melanie Folkins
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Ellen F Foxman
- Department of Laboratory Medicine and Department of Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alistair B Russell
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ketaki Ganti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kanta Subbarao
- Department of Microbiology and Immunology, WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, School of Medicine, Northwestern University Feinberg, Chicago, Illinois, USA
| | - Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
196
|
Aomar-Millán IF, de Victoria-Carazo JM, Fernández Reyes D, Torres-Parejo Ú, Pérez Fernández L, Martínez-Diz S, Ceballos Torres A, López Gómez J, Bizzarri F, Raya Álvarez E, Salvatierra J. Characteristics and clinical outcome in 312 patients with moderate to severe pneumonia due to SARS-COV-2 and hyperinflammation treated with anakinra and corticosteroids: A retrospective cohort study. PLoS One 2023; 18:e0283529. [PMID: 36961847 PMCID: PMC10038301 DOI: 10.1371/journal.pone.0283529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/12/2023] [Indexed: 03/25/2023] Open
Abstract
OBJECTIVE To assess the clinical outcome (death and/or Intensive Care Unit (ICU) admission) based on the time from hospital admission to the administration of anakinra and the possible usefulness of a "simplified" SCOPE score to stratify the risk of worse prognosis in our cohort of patients with moderate/severe SARS-CoV-2 pneumonia, both vaccinated and unvaccinated, that received anakinra and corticosteroids. In addition, the clinical, analytical, and imaging characteristics of patients at admission are described. METHODS Retrospective cohort study of 312 patients admitted to Hospital Clínico San Cecilio in Granada for moderate/severe pneumonia caused by SARS-CoV-2 that received anakinra and corticosteroids between March 2020 and January 2022. Clinical and analytical data were collected as well as the patient outcome at 30 and 60 days after admission. Three treatment groups were established according to the time from hospital admission to administration of anakinra: early (1st-2nd day), intermediate (3rd-5th day), and late (after the 5th day). RESULTS The median age was 67.4 years (IQR 22-97 years) and 204 (65.4%) were male. The most common comorbidity was hypertension (58%). The median time from the start of symptoms to anakinra administration was 6 days (IQR 5-10) and the SaFi (SaO2/FiO2) was 228 (IQR 71-471). The cure rate was higher in the early-onset anakinra group versus the late-onset group (73% vs 56.6%). The latter had a higher percentage of deaths (27.4%) and a greater number of patients remained hospitalized for a month (16%). On admission, the patients had elevated C-reactive protein (CRP), ferritin, and D-dimer values and decreased total lymphocytes. Analytical improvement was observed at both 72 hours and one month after treatment. 42 (13.5%) required ICU admission, and 23 (7.3%) orotracheal intubation. At 60 days, 221 (70.8%) were discharged, 87 (27.8%) had died and 4 (1.4%) remained hospitalized. The mean dose of anakinra was 1000 mg (100-2600 mg) with differences found between the dose administered and the clinical outcome. There were no differences in the primary outcome based on vaccination. A simplified SCOPE score at the start of anakinra administration was lower in patients with better clinical evolution. CONCLUSIONS Early treatment with anakinra and corticosteroids was associated with a better outcome regardless of vaccination status. A simplified SCOPE was found to be a good prognostic tool.
Collapse
Affiliation(s)
- Ismael Francisco Aomar-Millán
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
| | | | - Daniel Fernández Reyes
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Úrsula Torres-Parejo
- Department of Statistics, Faculty of Health Sciences, University of Granada, Granada, Spain
| | | | - Silvia Martínez-Diz
- Department of Preventive Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Angel Ceballos Torres
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Jairo López Gómez
- Department of Internal Medicine, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Francesco Bizzarri
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Enrique Raya Álvarez
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Juan Salvatierra
- Biosanitary Research Institute of Granada, ibsGRANADA, Granada, Spain
- Department of Rheumatology, Hospital Universitario Clínico San Cecilio, Granada, Spain
| |
Collapse
|
197
|
Wang T, Zhai Y, Xue H, Zhou W, Ding Y, Nie H. Regulation of Epithelial Sodium Transport by SARS-CoV-2 Is Closely Related with Fibrinolytic System-Associated Proteins. Biomolecules 2023; 13:biom13040578. [PMID: 37189326 DOI: 10.3390/biom13040578] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
Dyspnea and progressive hypoxemia are the main clinical features of patients with coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Pulmonary pathology shows diffuse alveolar damage with edema, hemorrhage, and the deposition of fibrinogens in the alveolar space, which are consistent with the Berlin Acute Respiratory Distress Syndrome Criteria. The epithelial sodium channel (ENaC) is a key channel protein in alveolar ion transport and the rate-limiting step for pulmonary edema fluid clearance, the dysregulation of which is associated with acute lung injury/acute respiratory distress syndrome. The main protein of the fibrinolysis system, plasmin, can bind to the furin site of γ-ENaC and induce it to an activation state, facilitating pulmonary fluid reabsorption. Intriguingly, the unique feature of SARS-CoV-2 from other β-coronaviruses is that the spike protein of the former has the same furin site (RRAR) with ENaC, suggesting that a potential competition exists between SARS-CoV-2 and ENaC for the cleavage by plasmin. Extensive pulmonary microthrombosis caused by disorders of the coagulation and fibrinolysis system has also been seen in COVID-19 patients. To some extent, high plasmin (ogen) is a common risk factor for SARS-CoV-2 infection since an increased cleavage by plasmin accelerates virus invasion. This review elaborates on the closely related relationship between SARS-CoV-2 and ENaC for fibrinolysis system-related proteins, aiming to clarify the regulation of ENaC under SARS-CoV-2 infection and provide a novel reference for the treatment of COVID-19 from the view of sodium transport regulation in the lung epithelium.
Collapse
Affiliation(s)
- Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hao Xue
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
198
|
COVID-19 Management Strategies in Solid Organ Transplant Recipients. Infect Dis Clin North Am 2023:S0891-5520(23)00024-7. [PMID: 37142512 PMCID: PMC10028355 DOI: 10.1016/j.idc.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Solid organ transplant recipients are at high risk of severe coronavirus disease-2019 (COVID-19). If left untreated, it results in high rates of hospitalization, intensive care unit admission and death. Early diagnosis of COVID-19 is essential to ensure the early administration of therapeutics. Treatment of mild-to-moderate COVID-19 with remdesivir, ritonavir-boosted nirmatrelvir, or an anti-spike neutralizing monoclonal antibody may prevent progression to severe and critical COVID-19. Among patients with severe and critical COVID-19, treatment with intravenous remdesivir and immunomodulation is recommended. This review article discusses strategies in the management of solid organ transplant recipients with COVID-19.
Collapse
|
199
|
Hogan JI, Duerr R, Heguy A. Reply to Yan. Clin Infect Dis 2023; 76:1155-1156. [PMID: 36346103 DOI: 10.1093/cid/ciac878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- John I Hogan
- Department of Medicine, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Ralf Duerr
- Department of Microbiology, NYU Grossman School of Medicine, New York, USA
| | - Adriana Heguy
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| |
Collapse
|
200
|
Scaramuzzo G, Nucera F, Asmundo A, Messina R, Mari M, Montanaro F, Johansen MD, Monaco F, Fadda G, Tuccari G, Hansbro NG, Hansbro PM, Hansel TT, Adcock IM, David A, Kirkham P, Caramori G, Volta CA, Spadaro S. Cellular and molecular features of COVID-19 associated ARDS: therapeutic relevance. J Inflamm (Lond) 2023; 20:11. [PMID: 36941580 PMCID: PMC10027286 DOI: 10.1186/s12950-023-00333-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/08/2023] [Indexed: 03/23/2023] Open
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can be asymptomatic or cause a disease (COVID-19) characterized by different levels of severity. The main cause of severe COVID-19 and death is represented by acute (or acute on chronic) respiratory failure and acute respiratory distress syndrome (ARDS), often requiring hospital admission and ventilator support.The molecular pathogenesis of COVID-19-related ARDS (by now termed c-ARDS) is still poorly understood. In this review we will discuss the genetic susceptibility to COVID-19, the pathogenesis and the local and systemic biomarkers correlated with c-ARDS and the therapeutic options that target the cell signalling pathways of c-ARDS.
Collapse
Affiliation(s)
- Gaetano Scaramuzzo
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Emergency, Section of Intensive Care and Anesthesia, Azienda Ospedaliera-Universitaria Sant’Anna, Ferrara, Italy
| | - Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Alessio Asmundo
- Medicina Legale, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Roberto Messina
- Intensive Care Unit, Dipartimento di Patologia Umana e dell’Età Evolutiva Gaetano Barresi, Università di Messina, Messina, Italy
| | - Matilde Mari
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Emergency, Section of Intensive Care and Anesthesia, Azienda Ospedaliera-Universitaria Sant’Anna, Ferrara, Italy
| | - Federica Montanaro
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Emergency, Section of Intensive Care and Anesthesia, Azienda Ospedaliera-Universitaria Sant’Anna, Ferrara, Italy
| | - Matt D. Johansen
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Francesco Monaco
- Chirurgia Toracica, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Guido Fadda
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Giovanni Tuccari
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Nicole G. Hansbro
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Philip M. Hansbro
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Trevor T. Hansel
- Medical Research Council and Asthma, UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Ian M. Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Antonio David
- Intensive Care Unit, Dipartimento di Patologia Umana e dell’Età Evolutiva Gaetano Barresi, Università di Messina, Messina, Italy
| | - Paul Kirkham
- Department of Biomedical Sciences, Faculty of Sciences and Engineering, University of Wolverhampton, West Midlands, Wolverhampton, UK
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Carlo Alberto Volta
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Emergency, Section of Intensive Care and Anesthesia, Azienda Ospedaliera-Universitaria Sant’Anna, Ferrara, Italy
| | - Savino Spadaro
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Department of Emergency, Section of Intensive Care and Anesthesia, Azienda Ospedaliera-Universitaria Sant’Anna, Ferrara, Italy
| |
Collapse
|