101
|
Lin Z, Niu J, Xu Y, Qin L, Ding J, Zhou L. Clinical efficacy and adverse events of baricitinib treatment for coronavirus disease-2019 (COVID-19): A systematic review and meta-analysis. J Med Virol 2021; 94:1523-1534. [PMID: 34846766 PMCID: PMC9015594 DOI: 10.1002/jmv.27482] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/08/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022]
Abstract
The benefits of baricitinib in coronavirus disease-2019 are inadequately defined. We performed a systematic review and meta-analysis of studies of baricitinib to determine its clinical efficacy and adverse events in patients with COVID-19. Databases were searched from their inception to September 5, 2021. The primary outcome was the coefficient of mortality. We also compared secondary indicators and adverse events between baricitinib treatment and placebo or other treatments. Twelve studies of 3564 patients were included and assessed qualitatively (modified Jadad and Newcastle-Ottawa Scale scores). Baricitinib effectively improved the mortality rate (relative risk of mortality = 0.56; 95% confidence interval: 0.46-0.69; p < 0.001; I2 = 2%), and this result was unchanged by subgroup analysis. Baricitinib improved intensive care unit admission, the requirement for invasive mechanical ventilation, and improved the oxygenation index. Data from these studies also showed that baricitinib slightly reduced the risk of adverse events. Regarding the choice of the drug dosage of baricitinib, the high-dose group appeared to have additional benefits for clinical efficacy. Our study shows that baricitinib may be a promising, safe, and effective anti-severe acute respiratory syndrome-coronavirus-2 drug candidate, with the advantages of low cost, easy production, and convenient storage.
Collapse
Affiliation(s)
- Zhiwei Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianyi Niu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifan Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijie Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiabin Ding
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Luqian Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
102
|
Tang XD, Ji TT, Dong JR, Feng H, Chen FQ, Chen X, Zhao HY, Chen DK, Ma WT. Pathogenesis and Treatment of Cytokine Storm Induced by Infectious Diseases. Int J Mol Sci 2021; 22:13009. [PMID: 34884813 PMCID: PMC8658039 DOI: 10.3390/ijms222313009] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokine storm is a phenomenon characterized by strong elevated circulating cytokines that most often occur after an overreactive immune system is activated by an acute systemic infection. A variety of cells participate in cytokine storm induction and progression, with profiles of cytokines released during cytokine storm varying from disease to disease. This review focuses on pathophysiological mechanisms underlying cytokine storm induction and progression induced by pathogenic invasive infectious diseases. Strategies for targeted treatment of various types of infection-induced cytokine storms are described from both host and pathogen perspectives. In summary, current studies indicate that cytokine storm-targeted therapies can effectively alleviate tissue damage while promoting the clearance of invading pathogens. Based on this premise, "multi-omics" immune system profiling should facilitate the development of more effective therapeutic strategies to alleviate cytokine storms caused by various diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| | - Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| |
Collapse
|
103
|
Ezeonwumelu IJ, Garcia-Vidal E, Ballana E. JAK-STAT Pathway: A Novel Target to Tackle Viral Infections. Viruses 2021; 13:v13122379. [PMID: 34960648 PMCID: PMC8704679 DOI: 10.3390/v13122379] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Modulation of the antiviral innate immune response has been proposed as a putative cellular target for the development of novel pan-viral therapeutic strategies. The Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway is especially relevant due to its essential role in the regulation of local and systemic inflammation in response to viral infections, being, therefore, a putative therapeutic target. Here, we review the extraordinary diversity of strategies that viruses have evolved to interfere with JAK-STAT signaling, stressing the relevance of this pathway as a putative antiviral target. Moreover, due to the recent remarkable progress on the development of novel JAK inhibitors (JAKi), the current knowledge on its efficacy against distinct viral infections is also discussed. JAKi have a proven efficacy against a broad spectrum of disorders and exhibit safety profiles comparable to biologics, therefore representing good candidates for drug repurposing strategies, including viral infections.
Collapse
|
104
|
Gopcsa L, Bobek I, Bekő G, Lakatos B, Molnár E, Réti M, Reményi P, Sinkó J, Szlávik J, Tatai G, Vályi-Nagy I. Common points of therapeutic intervention in COVID-19 and in allogeneic hematopoietic stem cell transplantation associated severe cytokine release syndrome. Acta Microbiol Immunol Hung 2021; 68:240-255. [PMID: 34797216 DOI: 10.1556/030.2021.01620] [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: 10/03/2021] [Accepted: 10/22/2021] [Indexed: 11/19/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) and coronavirus disease 2019 (COVID-19) infection can both lead to severe cytokine release syndrome (sCRS) resulting in critical illness and death. In this single institution, preliminary comparative case-series study we compared clinical and laboratory co-variates as well as response to tocilizumab (TCZ)-based therapy of 15 allogeneic-HSCT- and 17 COVID-19-associated sCRS patients. Reaction to a TCZ plus posttransplant cyclophosphamide (PTCY) consolidation therapy in the allogeneic-HSCT-associated sCRS group yielded significantly inferior long-term outcome as compared to TCZ-based therapy in the COVID-19-associated group (P = 0.003). We report that a TCZ followed by consolidation therapy with a Janus kinase/signal transducer and activator of transcription (JAK/STAT) inhibitor given to 4 out of 8 critically ill COVID-19 patients resulted in their complete recovery. Non-selective JAK/STAT inhibitors influencing the action of several cytokines exhibit a broader effect than TCZ alone in calming down sCRS. Serum levels of cytokines and chemokines show similar changes in allogeneic-HSCT- and COVID-19-associated sCRS with marked elevation of interleukin-6 (IL-6), regulated upon activation normal T-cell expressed and secreted (RANTES), monocyte chemoattractant protein-1 (MCP-1) and interferon γ-induced protein 10 kDa (IP-10) levels. In addition, levels of IL-5, IL-10, IL-15 were also elevated in allogeneic-HSCT-associated sCRS. Our multi-cytokine expression data indicate that the pathophysiology of allogeneic-HSCT and COVID-19-associated sCRS are similar therefore the same clinical grading system and TCZ-based treatment approaches can be applied. TCZ with JAK/STAT inhibitor consolidation therapy might be highly effective in COVID-19 sCRS patients.
Collapse
Affiliation(s)
- László Gopcsa
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Ilona Bobek
- 2Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Intensive Care Unit, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Gabriella Bekő
- 3Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Central Laboratory, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Botond Lakatos
- 4Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Infectious Diseases, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Eszter Molnár
- 2Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Intensive Care Unit, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Marienn Réti
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Péter Reményi
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - János Sinkó
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - János Szlávik
- 4Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Infectious Diseases, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - Gábor Tatai
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| | - István Vályi-Nagy
- 1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Albert Florian ut 5-7, H-1097, Budapest, Hungary
| |
Collapse
|
105
|
What have we learned from the first to the second wave of COVID-19 pandemic? An international survey from the ESCMID Study Group for Infection in the Elderly (ESGIE) group. Eur J Clin Microbiol Infect Dis 2021; 41:281-288. [PMID: 34775534 PMCID: PMC8590524 DOI: 10.1007/s10096-021-04377-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022]
Abstract
The purpose of this survey is to explore changes in the management of COVID-19 during the first versus the second wave, with particular emphasis on therapies, antibiotic prescriptions, and elderly care. An internet-based questionnaire survey was distributed to European Society of Clinical Microbiology and Infectious Diseases (ESCMID) members. Therapeutic approach to patients with mild-to-moderate (PiO2/FiO2 200–350) and severe (PiO2/FiO2 < 200) COVID-19, antibiotic use, and reasons for excluding patients from the intensive care unit (ICU) were investigated. A total of 463 from 21 countries participated in the study. Most representatives were infectious disease specialists (68.3%). During the second wave of pandemic, physicians abandoned the use of hydroxychloroquine, lopinavir/ritonavir, and azithromycin in favor of dexamethasone, low-molecular weight heparin (LMWH), and remdesivir in mild-to-moderate COVID-19. In critically ill patients, we detected an increased use of high-dose steroids (51%) and a decrease in tocilizumab use. The use of antibiotics at hospital admission decreased but remained high in the second wave. Age was reported to be a main consideration for exclusion of patients from ICU care by 25% of responders; a third reported that elderly were not candidates for ICU admission in their center. The decision to exclude patients from ICU care was based on the individual decision of an intensivist in 59.6% of cases. The approach of physicians to COVID-19 changed over time following evidence accumulation and guidelines. Antibiotic use at hospital admission and decision to exclude patients from ICU care remain critical aspects that should be better investigated and harmonized among clinicians.
Collapse
|
106
|
Alfaro-Martínez JJ, Solís García Del Pozo J, Lamas Oliveira C, Paterna Mellinas G, García-Castillo S, Molina Cifuentes M, Pinés Corrales P. Hospital capacity and admission rate may be a factor of importance to mortality in COVID-19. Infect Dis (Lond) 2021; 54:238-240. [PMID: 34751087 DOI: 10.1080/23744235.2021.2001565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
| | | | - Cristina Lamas Oliveira
- Department of Endocrinology, Albacete University Hospital Complex C/ Hermanos Falcó, 34, Albacete 02006, Spain
| | - Gema Paterna Mellinas
- Department of Geriatrics, Albacete University Hospital Complex C/ Hermanos Falcó, 34, Albacete 02006, Spain
| | - Sergio García-Castillo
- Department of Pneumonology, Albacete University Hospital Complex C/ Hermanos Falcó, 34, Albacete 02006, Spain
| | - María Molina Cifuentes
- Department of Internal Medicine, Albacete University Hospital Complex C/ Hermanos Falcó, 34, Albacete 02006, Spain
| | - Pedro Pinés Corrales
- Department of Endocrinology, Albacete University Hospital Complex C/ Hermanos Falcó, 34, Albacete 02006, Spain
| |
Collapse
|
107
|
Brüssow H. Host-modifying drugs against COVID-19: some successes, but not yet the breakthrough. Environ Microbiol 2021; 23:7257-7270. [PMID: 34729893 PMCID: PMC8652514 DOI: 10.1111/1462-2920.15828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
After reviewing antiviral drugs (Brüssow Environmental Microbiology 2021) the present review summarizes the results of clinical trials with host‐modifying drugs in COVID‐19 patients. Clinical benefits were observed with different immunomodulators. The variable outcomes of trials with the interleukin 6 receptor inhibitor tocilizumab demonstrated that treatment benefits might only be present in specific subgroups of patients or in specific infection stages. A meta‐analysis of trials with the interleukin 1 receptor antagonist anakinra showed a survival benefit only in patients with hyperinflammation. The Janus kinase inhibitor baricitinib is an anti‐inflammatory treatment that showed a clinical benefit in hospitalized patients who do not yet need supplementary oxygen. In contrast, the corticosteroid dexamethasone showed mortality reducing effects that were limited to patients on ventilation or in need of supplementary oxygen. Therapeutic dose of anticoagulation met the criteria for inferiority in severe cases, but showed a small survival benefit in non‐severe COVID‐19 patients. Large trials with colchicine showed a small or no survival benefit. Azithromycin, an antibiotic with immunomodulatory activity, showed no effects in numerous clinical trials. The trials showed a clear need for new drugs instead of repurposed drugs and drugs that specifically target the SARS‐CoV‐2 virus or the pathology developing in COVID‐19 patients.
Collapse
Affiliation(s)
- Harald Brüssow
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| |
Collapse
|
108
|
Tejada Cifuentes F, Lloret Callejo Á, Tirado Pérez MJ, Romero Candel G. Perfil de efectividad y seguridad de la colchicina en pacientes con diagnóstico de COVID-19 en un área sanitaria. REVISTA CLÍNICA DE MEDICINA DE FAMILIA 2021. [DOI: 10.55783/rcmf.140304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Objetivo: describir la evolución y la seguridad de la utilización de colchicina en pacientes no ingresados con diagnóstico de COVID-19 leve-moderado en la Gerencia de Atención Integrada (GAI) de Albacete.
Métodos: estudio observacional retrospectivo. De los 389 participantes incluidos en el primer reclutamiento, se seleccionaron 315 con datos válidos. La variable principal del estudio ha sido el fallecimiento o ingreso hospitalario en pacientes con diagnóstico de COVID-19 y tratamiento con colchicina. Se registraron variables sociodemográficas, clínicas y tratamientos y comorbilidades concomitantes.
Resultados: fallecieron 6 (1,90%) pacientes y 49 (15,5%) requirieron ingreso hospitalario. A un 58,4% se les prescribió un antibiótico, siendo la azitromicina el más utilizado y el responsable en un 32,7% de las posibles interacciones. Un 34,5% y un 43% de pacientes recibieron heparinas de bajo peso molecular (HBPM) y corticosteroides respectivamente. En el 42,3% de pacientes no se tuvo en cuenta el valor del aclaramiento de creatinina al dosificar la colchicina. La edad elevada muestra una relación estadísticamente significativa con la gravedad de la clínica (68,5 versus 58,9) y con la variable recaída (ingreso + urgencias) (63,25 versus 58,54).
Conclusiones: en nuestra muestra, la utilización de colchicina en pacientes ambulatorios no ha modificado el curso de la enfermedad en pacientes diagnosticados de COVID-19.
Palabras clave: colchicina, COVID-19, coronavirus, tratamiento.
Collapse
Affiliation(s)
- Francisco Tejada Cifuentes
- Servicio de Farmacia de Atención Primaria. Gerencia de Atención Integrada de Albacete. Albacete (España)
| | - Ángeles Lloret Callejo
- Servicio de Farmacia de Atención Primaria. Gerencia de Atención Integrada de Albacete. Albacete (España)
| | - María José Tirado Pérez
- Servicio de Farmacia de Atención Primaria. Gerencia de Atención Integrada de Albacete. Albacete (España)
| | - Gregorio Romero Candel
- Servicio de Farmacia. Hospital de Hellín. Gerencia de Atención Integrada de Hellín. Albacete (España)
| |
Collapse
|
109
|
Mak KK, Balijepalli MK, Pichika MR. Success stories of AI in drug discovery - where do things stand? Expert Opin Drug Discov 2021; 17:79-92. [PMID: 34553659 DOI: 10.1080/17460441.2022.1985108] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Artificial intelligence (AI) in drug discovery and development (DDD) has gained more traction in the past few years. Many scientific reviews have already been made available in this area. Thus, in this review, the authors have focused on the success stories of AI-driven drug candidates and the scientometric analysis of the literature in this field. AREA COVERED The authors explore the literature to compile the success stories of AI-driven drug candidates that are currently being assessed in clinical trials or have investigational new drug (IND) status. The authors also provide the reader with their expert perspectives for future developments and their opinions on the field. EXPERT OPINION Partnerships between AI companies and the pharma industry are booming. The early signs of the impact of AI on DDD are encouraging, and the pharma industry is hoping for breakthroughs. AI can be a promising technology to unveil the greatest successes, but it has yet to be proven as AI is still at the embryonic stage.
Collapse
Affiliation(s)
- Kit-Kay Mak
- School of Postgraduate Studies and Research, International Medical University, Bukit Jalil, Malaysia.,Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| | | | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| |
Collapse
|
110
|
Stamatelopoulos K, Georgiopoulos G, Baker KF, Tiseo G, Delialis D, Lazaridis C, Barbieri G, Masi S, Vlachogiannis NI, Sopova K, Mengozzi A, Ghiadoni L, Schim van der Loeff I, Hanrath AT, Ajdini B, Vlachopoulos C, Dimopoulos MA, Duncan CJA, Falcone M, Stellos K. Estimated pulse wave velocity improves risk stratification for all-cause mortality in patients with COVID-19. Sci Rep 2021; 11:20239. [PMID: 34642385 PMCID: PMC8511157 DOI: 10.1038/s41598-021-99050-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
Accurate risk stratification in COVID-19 patients consists a major clinical need to guide therapeutic strategies. We sought to evaluate the prognostic role of estimated pulse wave velocity (ePWV), a marker of arterial stiffness which reflects overall arterial integrity and aging, in risk stratification of hospitalized patients with COVID-19. This retrospective, longitudinal cohort study, analyzed a total population of 1671 subjects consisting of 737 hospitalized COVID-19 patients consecutively recruited from two tertiary centers (Newcastle cohort: n = 471 and Pisa cohort: n = 266) and a non-COVID control cohort (n = 934). Arterial stiffness was calculated using validated formulae for ePWV. ePWV progressively increased across the control group, COVID-19 survivors and deceased patients (adjusted mean increase per group 1.89 m/s, P < 0.001). Using a machine learning approach, ePWV provided incremental prognostic value and improved reclassification for mortality over the core model including age, sex and comorbidities [AUC (core model + ePWV vs. core model) = 0.864 vs. 0.755]. ePWV provided similar prognostic value when pulse pressure or hs-Troponin were added to the core model or over its components including age and mean blood pressure (p < 0.05 for all). The optimal prognostic ePWV value was 13.0 m/s. ePWV conferred additive discrimination (AUC: 0.817 versus 0.779, P < 0.001) and reclassification value (NRI = 0.381, P < 0.001) over the 4C Mortality score, a validated score for predicting mortality in COVID-19 and the Charlson comorbidity index. We suggest that calculation of ePWV, a readily applicable estimation of arterial stiffness, may serve as an additional clinical tool to refine risk stratification of hospitalized patients with COVID-19 beyond established risk factors and scores.
Collapse
Affiliation(s)
- Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
- Biosciences Institute, International Centre for Life, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK.
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, King's College, London, UK
| | - Kenneth F Baker
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Giusy Tiseo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Charalampos Lazaridis
- Biosciences Institute, International Centre for Life, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
- RVI and Freeman Hospitals, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Greta Barbieri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nikolaos I Vlachogiannis
- Biosciences Institute, International Centre for Life, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Kateryna Sopova
- Biosciences Institute, International Centre for Life, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
- RVI and Freeman Hospitals, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lorenzo Ghiadoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ina Schim van der Loeff
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Aidan T Hanrath
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Bajram Ajdini
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Charalambos Vlachopoulos
- First Department of Cardiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Christopher J A Duncan
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- RVI and Freeman Hospitals, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marco Falcone
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Konstantinos Stellos
- Biosciences Institute, International Centre for Life, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK.
- RVI and Freeman Hospitals, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
111
|
Smet A, Breugelmans T, Michiels J, Lamote K, Arras W, De Man JG, Heyndrickx L, Hauner A, Huizing M, Malhotra-Kumar S, Lammens M, Hotterbeekx A, Kumar-Singh S, Verstraeten A, Loeys B, Verhoeven V, Jacobs R, Dams K, Coenen S, Ariën KK, Jorens PG, De Winter BY. A dynamic mucin mRNA signature associates with COVID-19 disease presentation and severity. JCI Insight 2021; 6:e151777. [PMID: 34448730 PMCID: PMC8525642 DOI: 10.1172/jci.insight.151777] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/25/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUNDSARS-CoV-2 infection induces mucin overexpression, further promoting disease. Given that mucins are critical components of innate immunity, unraveling their expression profiles that dictate the course of disease could greatly enhance our understanding and management of COVID-19.METHODSUsing validated RT-PCR assays, we assessed mucin mRNA expression in the blood of patients with symptomatic COVID-19 compared with symptomatic patients without COVID-19 and healthy controls and correlated the data with clinical outcome parameters. Additionally, we analyzed mucin expression in mucus and lung tissue from patients with COVID-19 and investigated the effect of drugs for COVID-19 treatment on SARS-CoV-2-induced mucin expression in pulmonary epithelial cells.RESULTSWe identified a dynamic blood mucin mRNA signature that clearly distinguished patients with symptomatic COVID-19 from patients without COVID-19 based on expression of MUC1, MUC2, MUC4, MUC6, MUC13, MUC16, and MUC20 (AUCROC of 91.8%; sensitivity and specificity of 90.6% and 93.3%, respectively) and that discriminated between mild and critical COVID-19 based on the expression of MUC16, MUC20, and MUC21 (AUCROC of 89.1%; sensitivity and specificity of 90.0% and 85.7%, respectively). Differences in the transcriptional landscape of mucins in critical cases compared with mild cases identified associations with COVID-19 symptoms, respiratory support, organ failure, secondary infections, and mortality. Furthermore, we identified different mucins in the mucus and lung tissue of critically ill COVID-19 patients and showed the ability of baricitinib, tocilizumab, favipiravir, and remdesivir to suppress expression of SARS-CoV-2-induced mucins.CONCLUSIONThis multifaceted blood mucin mRNA signature showed the potential role of mucin profiling in diagnosing, estimating severity, and guiding treatment options in patients with COVID-19.FUNDINGThe Antwerp University Research and the Research Foundation Flanders COVID-19 funds.
Collapse
Affiliation(s)
- Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Tom Breugelmans
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Johan Michiels
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Kevin Lamote
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Wout Arras
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Joris G. De Man
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Anne Hauner
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Manon Huizing
- Biobank Antwerpen, Antwerp University Hospital, Edegem, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Martin Lammens
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - An Hotterbeekx
- Laboratory of Cell Biology and Histology, Molecular Pathology Group, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Samir Kumar-Singh
- Laboratory of Cell Biology and Histology, Molecular Pathology Group, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Aline Verstraeten
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bart Loeys
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Veronique Verhoeven
- Department of Family Medicine and Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Rita Jacobs
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Critical Care Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Karolien Dams
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Critical Care Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Samuel Coenen
- Department of Family Medicine and Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Kevin K. Ariën
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Philippe G. Jorens
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Critical Care Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Benedicte Y. De Winter
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, and
- Infla-med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
112
|
Kalyanaraman B. Reactive oxygen species, proinflammatory and immunosuppressive mediators induced in COVID-19: overlapping biology with cancer. RSC Chem Biol 2021; 2:1402-1414. [PMID: 34704045 PMCID: PMC8496060 DOI: 10.1039/d1cb00042j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
This review analyzes the published literature linking the different mechanisms focused on oxidative stress and inflammation that contribute to COVID-19 disease severity. The objective is to bring together potential proinflammatory mechanisms of COVID-19 pathogenesis and address mitigation strategies using naturally occurring compounds and FDA-approved drugs. Outstanding questions addressed include the following: What is the mechanistic basis for linking enhanced vulnerability in COVID-19 to increased oxidative damage and proinflammatory mediators (e.g., cytokines), especially in high-risk people? Can we repurpose anti-inflammatory and immunomodulatory agents to mitigate inflammation in COVID-19 patients? How does 2-deoxy-d-glucose function as an anti-COVID drug? COVID-19, cancer biology, and immunotherapy share many mechanistic similarities. Repurposing drugs that already have been FDA-approved for mitigating inflammation and immunosuppression in cancer may be a way to counteract disease severity, progression, and chronic inflammation in COVID-19. What are the long-term effects of reactive oxygen species-inducing immune cells and sustained inflammation in so-called long-haulers (long COVID) after recovery from COVID-19? Can we use mitochondria-targeted agents prophylactically to prevent inflammation and boost immunity in long-haulers? Addressing the oxidative chemical biology of COVID-19 and the mechanistic commonalities with cancer may provide new insights potentially leading to appropriate clinical trials and new treatments.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of Biophysics, Cancer Center, Center for Disease Prevention Research, Medical College of Wisconsin 8701 Watertown Plank Road Milwaukee WI 53226 USA
| |
Collapse
|
113
|
Li Y, Zhao W, Liu J, Chen Z, Lv Q, Zhang Z. Immunotherapy Summary for Cytokine Storm in COVID-19. Front Pharmacol 2021; 12:731847. [PMID: 34603047 PMCID: PMC8484328 DOI: 10.3389/fphar.2021.731847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has ravaged the world, resulting in an alarming number of infections and deaths, and the number continues to increase. The pathogenesis caused by the novel coronavirus was found to be a disruption of the pro-inflammatory/anti-inflammatory response. Due to the lack of effective treatments, different strategies and treatment methods are still being researched, with the use of vaccines to make the body immune becoming the most effective means of prevention. Antiviral drugs and respiratory support are often used clinically as needed, but are not yet sufficient to alleviate the cytokine storm (CS) and systemic inflammatory response syndrome. How to neutralize the cytokine storm and inhibit excessive immune cell activation becomes the key to treating neocoronavirus pneumonia. Immunotherapy through the application of hormones and monoclonal antibodies can alleviate the immune imbalance, but the clinical effectiveness and side effects remain controversial. This article reviews the pathogenesis of neocoronavirus pneumonia and discusses the immunomodulatory therapies currently applied to COVID-19. We aim to give some conceptual thought to the prevention and immunotherapy of neocoronavirus pneumonia.
Collapse
Affiliation(s)
- Yaqun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjie Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhua Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zichao Chen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingtao Lv
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
114
|
Pérez-Alba E, Nuzzolo-Shihadeh L, Aguirre-García GM, Espinosa-Mora J, Lecona-Garcia JD, Flores-Pérez RO, Mendoza-Garza M, Camacho-Ortiz A. Baricitinib plus dexamethasone compared to dexamethasone for the treatment of severe COVID-19 pneumonia: A retrospective analysis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 54:787-793. [PMID: 34294593 PMCID: PMC8253716 DOI: 10.1016/j.jmii.2021.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We aimed to analyze clinical outcomes from patients with severe COVID-19 pneumonia that received either baricitinib plus dexamethasone or dexamethasone monotherapy. METHODOLOGY We performed a retrospective comparative study. Data from hospitalized patients with severe COVID-19 pneumonia (saturation <93%, bilateral pulmonary infiltrates) that were treated with baricitinib plus dexamethasone or dexamethasone were collected. Our primary objective was to compare overall mortality and secondly to compare progression to mechanical ventilation and over infection rates. RESULTS A total of 793 patients were assessed for inclusion criteria, 596 were excluded and 197 were analyzed for primary outcome: 123 in the baricitinib plus dexamethasone group and 74 in the dexamethasone monotherapy group. The mean age was 59.9 years (SD ± 14.5) and 62.1% (123/197) were male. 42.9% (85/197) of the cases required ICU admission and 25.8% (51/197) underwent invasive mechanical ventilation (IMV). Overall thirty-day mortality was 27.9% (55/197); Mortality was significantly lower in the baricitinib plus dexamethasone group compared to the dexamethasone monotherapy group (20.3% vs 40.5%, P = <.05). There was no difference in hospital acquired infections between both groups. CONCLUSION Thirty-day mortality was significantly lower in patients with COVID-19 pneumonia treated with baricitinib plus dexamethasone versus dexamethasone monotherapy. No difference was observed in progression to invasive mechanical ventilation and hospital acquired infections.
Collapse
Affiliation(s)
- Eduardo Pérez-Alba
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico; Coordinación de Epidemiología, Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Laura Nuzzolo-Shihadeh
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Gloria Mayela Aguirre-García
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Jaime Espinosa-Mora
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Juan Diego Lecona-Garcia
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Romulo Omar Flores-Pérez
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Marisela Mendoza-Garza
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Adrián Camacho-Ortiz
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico; Coordinación de Epidemiología, Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Mexico.
| |
Collapse
|
115
|
Shi R, Feng Z, Zhang X. Integrative Multi-omics Landscape of Non-structural Protein 3 of Severe Acute Respiratory Syndrome Coronaviruses. GENOMICS, PROTEOMICS & BIOINFORMATICS 2021; 19:707-726. [PMID: 34774773 PMCID: PMC8578027 DOI: 10.1016/j.gpb.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/26/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022]
Abstract
The coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is currently a global pandemic. Extensive investigations have been performed to study the clinical and cellular effects of SARS-CoV-2 infection. Mass spectrometry-based proteomics studies have revealed the cellular changes due to the infection and identified a plethora of interactors for all SARS-CoV-2 components, except for the longest non-structural protein 3 (NSP3). Here, we expressed the full-length NSP3 proteins of SARS-CoV and SARS-CoV-2 to investigate their unique and shared functions using multi-omics methods. We conducted interactome, phosphoproteome, ubiquitylome, transcriptome, and proteome analyses of NSP3-expressing cells. We found that NSP3 plays essential roles in cellular functions such as RNA metabolism and immune response (e.g., NF-κB signal transduction). Interestingly, we showed that SARS-CoV-2 NSP3 has both endoplasmic reticulum and mitochondrial localizations. In addition, SARS-CoV-2 NSP3 is more closely related to mitochondrial ribosomal proteins, whereas SARS-CoV NSP3 is related to the cytosolic ribosomal proteins. In summary, our integrative multi-omics study of NSP3 improves the understanding of the functions of NSP3 and offers potential targets for the development of anti-SARS strategies.
Collapse
Affiliation(s)
- Ruona Shi
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenhuan Feng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaofei Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Cell Lineage and Atlas, Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510530, China.
| |
Collapse
|
116
|
Abizanda P, Calbo Mayo JM, Mas Romero M, Cortés Zamora EB, Tabernero Sahuquillo MT, Romero Rizos L, Sánchez‐Jurado PM, Sánchez‐Nievas G, Campayo Escolano C, Ochoa Serrano A, Sánchez‐Flor Alfaro V, López Bru R, Gómez Ballesteros C, Caldevilla Bernardo D, Callejas González FJ, Andrés‐Pretel F, Lauschke VM, Stebbing J. Baricitinib reduces 30-day mortality in older adults with moderate-to-severe COVID-19 pneumonia. J Am Geriatr Soc 2021; 69:2752-2758. [PMID: 34235720 PMCID: PMC8447356 DOI: 10.1111/jgs.17357] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Older adults are at the highest risk of severe disease and death due to COVID-19. Randomized data have shown that baricitinib improves outcomes in these patients, but focused stratified analyses of geriatric cohorts are lacking. Our objective was to analyze the efficacy of baricitinib in older adults with COVID-19 moderate-to-severe pneumonia. METHODS This is a propensity score [PS]-matched retrospective cohort study. Patients from the COVID-AGE and Alba-Score cohorts, hospitalized for moderate-to-severe COVID-19 pneumonia, were categorized in two age brackets of age <70 years old (86 with baricitinib and 86 PS-matched controls) or ≥70 years old (78 on baricitinib and 78 PS-matched controls). Thirty-day mortality rates were analyzed with Kaplan-Meier and Cox proportional hazard models. RESULTS Mean age was 79.1 for those ≥70 years and 58.9 for those <70. Exactly 29.6% were female. Treatment with baricitinib resulted in a significant reduction in death from any cause by 48% in patients aged 70 or older, an 18.5% reduction in 30-day absolute mortality risk (n/N: 16/78 [20.5%] baricitinib, 30/78 [38.5%] in PS-matched controls, p < 0.001) and a lower 30-day adjusted fatality rate (HR 0.21; 95% CI 0.09-0.47; p < 0.001). Beneficial effects on mortality were also observed in the age group <70 (8.1% reduction in 30-day absolute mortality risk; HR 0.14; 95% CI 0.03-0.64; p = 0.011). CONCLUSIONS Baricitinib is associated with an absolute mortality risk reduction of 18.5% in adults older than 70 years hospitalized with COVID-19 pneumonia.
Collapse
Affiliation(s)
- Pedro Abizanda
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
- CIBERFESMinisterio de Economía y CompetitividadMadridSpain
- Facultad de MedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Juan María Calbo Mayo
- Department of Internal MedicineComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
| | - Marta Mas Romero
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
| | - Elisa Belén Cortés Zamora
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
- CIBERFESMinisterio de Economía y CompetitividadMadridSpain
| | | | - Luis Romero Rizos
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
- CIBERFESMinisterio de Economía y CompetitividadMadridSpain
- Facultad de MedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Pedro Manuel Sánchez‐Jurado
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
- CIBERFESMinisterio de Economía y CompetitividadMadridSpain
- Facultad de MedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Ginés Sánchez‐Nievas
- Department of RheumatologyComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
| | | | - Alba Ochoa Serrano
- Department of Internal MedicineComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
| | | | - Rita López Bru
- Department of GeriatricsComplejo Hospitalario Universitario de AlbaceteAlbaceteSpain
| | | | | | | | - Fernando Andrés‐Pretel
- Department of StatisticsFoundation of the National Paraplegics Hospital of ToledoToledoSpain
| | | | - Justin Stebbing
- Department of Surgery and Cancer, Imperial CollegeHammersmith Hospital, ICTEM BuildingLondonUK
| |
Collapse
|
117
|
Thoms BL, Gosselin J, Libman B, Littenberg B, Budd R. Efficacy of Combination Therapy With The JAK Inhibitor Baricitinib In The Treatment of COVID-19. RESEARCH SQUARE 2021. [PMID: 34518834 PMCID: PMC8437315 DOI: 10.21203/rs.3.rs-835734/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND: Coronavirus disease-19 (COVID-19), resulting from infection with SARS-CoV-2, spans a wide spectrum of illness. In severely ill patients, highly elevated serum levels of certain cytokines and considerable cytolytic T cell infiltrates in the lungs have been observed. These same patients may bear low to negligible viral burdens suggesting that an overactive immune response, often termed cytokine storm, contributes to the severity of COVID-19. We report the safety and efficacy of baricitinib combined with remdesivir and dexamethasone in 45 hospitalized patients with COVID-19 pneumonia at a tertiary academic medical center. METHODS: Retrospective review of 45 patients hospitalized with COVID-19 pneumonia. Patients received 7-day course of baricitinib, 5-day course of remdesivir and 10-day course of dexamethasone. Clinical status and biomarkers were obtained daily. Outcomes assessed include mortality, duration of hospitalization, presence of shock, need for supplemental oxygen, need for non-invasive ventilation, need for mechanical ventilation and development of thrombosis. RESULTS: Obesity and multiple medical comorbidities were associated with hospitalization in the setting of COVID-19. Treated patients demonstrated rapid declines of C-reactive protein (CRP), ferritin and D-dimer with gradual improvement in hemoglobin, platelet counts and clinical status. Only 2 of 45 (4.4%) treated patients required mechanical ventilation after initiating treatment and there were six deaths (13.3%).Only 2 of 45 (4.4%) treated patients required mechanical ventilation after initiating treatment. There were six deaths (13.3%) and these were associated with lower BMI. CONCLUSIONS: These findings support the utility of immunosuppression via JAK inhibition in moderate to severe COVID-19 pneumonia.
Collapse
|
118
|
Melikhov O, Kruglova T, Lytkina K, Melkonyan G, Prokhorovich E, Putsman G, Rodoman G, Vertkin A, Zagrebneva A, Stebbing J. Use of Janus kinase inhibitors in COVID-19: a prospective observational series in 522 individuals. Ann Rheum Dis 2021; 80:1245-1246. [PMID: 34099486 PMCID: PMC8372389 DOI: 10.1136/annrheumdis-2021-220049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/25/2021] [Indexed: 01/20/2023]
Affiliation(s)
- Oleg Melikhov
- Institute of Clinical Research, Moscow, Russian Federation
| | | | | | | | - Elena Prokhorovich
- Moscow State University of Medicine and Dentistry n.a. AI Yevdokimov, Moscow, Russian Federation
| | - Gleb Putsman
- City Clinical Hospital No 24, Moscow, Russian Federation
| | | | - Arkady Vertkin
- Moscow State University of Medicine and Dentistry n.a. AI Yevdokimov, Moscow, Russian Federation
| | | | | |
Collapse
|
119
|
Marconi VC, Ramanan AV, de Bono S, Kartman CE, Krishnan V, Liao R, Piruzeli MLB, Goldman JD, Alatorre-Alexander J, de Cassia Pellegrini R, Estrada V, Som M, Cardoso A, Chakladar S, Crowe B, Reis P, Zhang X, Adams DH, Ely EW. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): a randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. THE LANCET RESPIRATORY MEDICINE 2021; 9:1407-1418. [PMID: 34480861 PMCID: PMC8409066 DOI: 10.1016/s2213-2600(21)00331-3] [Citation(s) in RCA: 496] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Background Baricitinib is an oral selective Janus kinase 1/2 inhibitor with known anti-inflammatory properties. This study evaluates the efficacy and safety of baricitinib in combination with standard of care for the treatment of hospitalised adults with COVID-19. Methods In this phase 3, double-blind, randomised, placebo-controlled trial, participants were enrolled from 101 centres across 12 countries in Asia, Europe, North America, and South America. Hospitalised adults with COVID-19 receiving standard of care were randomly assigned (1:1) to receive once-daily baricitinib (4 mg) or matched placebo for up to 14 days. Standard of care included systemic corticosteroids, such as dexamethasone, and antivirals, including remdesivir. The composite primary endpoint was the proportion who progressed to high-flow oxygen, non-invasive ventilation, invasive mechanical ventilation, or death by day 28, assessed in the intention-to-treat population. All-cause mortality by day 28 was a key secondary endpoint, and all-cause mortality by day 60 was an exploratory endpoint; both were assessed in the intention-to-treat population. Safety analyses were done in the safety population defined as all randomly allocated participants who received at least one dose of study drug and who were not lost to follow-up before the first post-baseline visit. This study is registered with ClinicalTrials.gov, NCT04421027. Findings Between June 11, 2020, and Jan 15, 2021, 1525 participants were randomly assigned to the baricitinib group (n=764) or the placebo group (n=761). 1204 (79·3%) of 1518 participants with available data were receiving systemic corticosteroids at baseline, of whom 1099 (91·3%) were on dexamethasone; 287 (18·9%) participants were receiving remdesivir. Overall, 27·8% of participants receiving baricitinib and 30·5% receiving placebo progressed to meet the primary endpoint (odds ratio 0·85 [95% CI 0·67 to 1·08], p=0·18), with an absolute risk difference of −2·7 percentage points (95% CI −7·3 to 1·9). The 28-day all-cause mortality was 8% (n=62) for baricitinib and 13% (n=100) for placebo (hazard ratio [HR] 0·57 [95% CI 0·41–0·78]; nominal p=0·0018), a 38·2% relative reduction in mortality; one additional death was prevented per 20 baricitinib-treated participants. The 60-day all-cause mortality was 10% (n=79) for baricitinib and 15% (n=116) for placebo (HR 0·62 [95% CI 0·47–0·83]; p=0·0050). The frequencies of serious adverse events (110 [15%] of 750 in the baricitinib group vs 135 [18%] of 752 in the placebo group), serious infections (64 [9%] vs 74 [10%]), and venous thromboembolic events (20 [3%] vs 19 [3%]) were similar between the two groups. Interpretation Although there was no significant reduction in the frequency of disease progression overall, treatment with baricitinib in addition to standard of care (including dexamethasone) had a similar safety profile to that of standard of care alone, and was associated with reduced mortality in hospitalised adults with COVID-19. Funding Eli Lilly and Company. Translations For the French, Japanese, Portuguese, Russian and Spanish translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Vincent C Marconi
- Emory University School of Medicine, Rollins School of Public Health and the Emory Vaccine Center, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA.
| | - Athimalaipet V Ramanan
- Translational Health Sciences, University of Bristol, Bristol, UK; Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
| | | | | | | | - Ran Liao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Providence St Joseph Health, Seattle, WA, USA; Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Rita de Cassia Pellegrini
- Pesquisare/Santo André, Brazil and Hospital Beneficência Portuguesa de São Caetano do Sul / São Caetano do Sul, Brazil
| | - Vicente Estrada
- Hospital Clinico San Carlos-IdiSSC, Universidad Complutense, Madrid, Spain
| | - Mousumi Som
- Oklahoma State University Medicine, Internal Medicine-Houston Center, Tulsa, OK, USA
| | | | | | | | - Paulo Reis
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Xin Zhang
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - E Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship Center (CIBS), Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine at Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Veteran's Affairs Geriatric Research Education Clinical Center (GRECC), Nashville, TN, USA
| | | |
Collapse
|
120
|
Spini A, Giudice V, Brancaleone V, Morgese MG, De Francia S, Filippelli A, Ruggieri A, Ziche M, Ortona E, Cignarella A, Trabace L. Sex-tailored pharmacology and COVID-19: Next steps towards appropriateness and health equity. Pharmacol Res 2021; 173:105848. [PMID: 34454035 PMCID: PMC8387562 DOI: 10.1016/j.phrs.2021.105848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 12/12/2022]
Abstract
Making gender bias visible allows to fill the gaps in knowledge and understand health records and risks of women and men. The coronavirus disease 2019 (COVID-19) pandemic has shown a clear gender difference in health outcomes. The more severe symptoms and higher mortality in men as compared to women are likely due to sex and age differences in immune responses. Age-associated decline in sex steroid hormone levels may mediate proinflammatory reactions in older adults, thereby increasing their risk of adverse outcomes, whereas sex hormones and/or sex hormone receptor modulators may attenuate the inflammatory response and provide benefit to COVID-19 patients. While multiple pharmacological options including anticoagulants, glucocorticoids, antivirals, anti-inflammatory agents and traditional Chinese medicine preparations have been tested to treat COVID-19 patients with varied levels of evidence in terms of efficacy and safety, information on sex-targeted treatment strategies is currently limited. Women may have more benefit from COVID-19 vaccines than men, despite the occurrence of more frequent adverse effects, and long-term safety data with newly developed vectors are eagerly awaited. The prevalent inclusion of men in randomized clinical trials (RCTs) with subsequent extrapolation of results to women needs to be addressed, as reinforcing sex-neutral claims into COVID-19 research may insidiously lead to increased inequities in health care. The huge worldwide effort with over 3000 ongoing RCTs of pharmacological agents should focus on improving knowledge on sex, gender and age as pillars of individual variation in drug responses and enforce appropriateness.
Collapse
Affiliation(s)
- Andrea Spini
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, via Ateneo Lucano, 85100 Potenza, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Silvia De Francia
- Department of Clinical and Biological Sciences, S. Luigi Hospital, University of Turin, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Anna Ruggieri
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Ziche
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Elena Ortona
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy.
| |
Collapse
|
121
|
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has resulted in a pandemic that has had widespread effects on human activities. The clinical presentation of severe COVID-19 includes a broad spectrum of clinical disease, most notably acute respiratory distress syndrome, cytokine release syndrome (CRS), multiorgan failure, and death. Direct viral damage and uncontrolled inflammation have been suggested as contributory factors in COVID-19 disease severity. The COVID-19 pandemic has emphasized the critical role of an effective host immune response in controlling a virus infection and demonstrated the devastating effect of immune dysregulation. Understanding the nature of the immune response to SARS-CoV-2 pathogenesis is key to developing effective treatments for COVID-19. Here, we describe the nature of the dysregulated host immune response in COVID-19, identify potential mechanisms involved in CRS, and discuss potential strategies that can be used to manage immune dysregulation in COVID-19. Expected final online publication date for the Annual Review of Medicine, Volume 73 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;
| |
Collapse
|
122
|
Xu J, Xiao W, Liang X, Shi L, Zhang P, Wang Y, Wang Y, Yang H. A meta-analysis on the risk factors adjusted association between cardiovascular disease and COVID-19 severity. BMC Public Health 2021; 21:1533. [PMID: 34380456 PMCID: PMC8355578 DOI: 10.1186/s12889-021-11051-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 05/12/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD), one of the most common comorbidities of coronavirus disease 2019 (COVID-19), has been suspected to be associated with adverse outcomes in COVID-19 patients, but their correlation remains controversial. METHOD This is a quantitative meta-analysis on the basis of adjusted effect estimates. PubMed, Web of Science, MedRxiv, Scopus, Elsevier ScienceDirect, Cochrane Library and EMBASE were searched comprehensively to obtain a complete data source up to January 7, 2021. Pooled effects (hazard ratio (HR), odds ratio (OR)) and the 95% confidence intervals (CIs) were estimated to evaluate the risk of the adverse outcomes in COVID-19 patients with CVD. Heterogeneity was assessed by Cochran's Q-statistic, I2test, and meta-regression. In addition, we also provided the prediction interval, which was helpful for assessing whether the variation across studies was clinically significant. The robustness of the results was evaluated by sensitivity analysis. Publication bias was assessed by Begg's test, Egger's test, and trim-and-fill method. RESULT Our results revealed that COVID-19 patients with pre-existing CVD tended more to adverse outcomes on the basis of 203 eligible studies with 24,032,712 cases (pooled ORs = 1.41, 95% CIs: 1.32-1.51, prediction interval: 0.84-2.39; pooled HRs = 1.34, 95% CIs: 1.23-1.46, prediction interval: 0.82-2.21). Further subgroup analyses stratified by age, the proportion of males, study design, disease types, sample size, region and disease outcomes also showed that pre-existing CVD was significantly associated with adverse outcomes among COVID-19 patients. CONCLUSION Our findings demonstrated that pre-existing CVD was an independent risk factor associated with adverse outcomes among COVID-19 patients.
Collapse
Affiliation(s)
- Jie Xu
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Wenwei Xiao
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Xuan Liang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Li Shi
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Peihua Zhang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Ying Wang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
123
|
Gomaa AA, Mohamed HS, Abd-Ellatief RB, Gomaa MA. Boswellic acids/Boswellia serrata extract as a potential COVID-19 therapeutic agent in the elderly. Inflammopharmacology 2021; 29:1033-1048. [PMID: 34224069 PMCID: PMC8256410 DOI: 10.1007/s10787-021-00841-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/20/2021] [Indexed: 12/14/2022]
Abstract
The most severe cases of COVID-19, and the highest rates of death, are among the elderly. There is an urgent need to search for an agent to treat the disease and control its progression. Boswellia serrata is traditionally used to treat chronic inflammatory diseases of the lung. This review aims to highlight currently published research that has shown evidence of potential therapeutic effects of boswellic acids (BA) and B. serrata extract against COVID-19 and associated conditions. We reviewed the published information up to March 2021. Studies were collected through a search of online electronic databases (academic libraries such as PubMed, Scopus, Web of Science, and Egyptian Knowledge Bank). Several recent studies reported that BAs and B. serrata extract are safe agents and have multiple beneficial activities in treating similar symptoms experienced by patients with COVID-19. Because of the low oral bioavailability and improvement of buccal/oral cavity hygiene, traditional use by chewing B. serrata gum may be more beneficial than oral use. It is the cheapest option for a lot of poorer people. The promising effect of B. serrata and BA can be attributed to its antioxidant, anti-inflammatory, immunomodulatory, cardioprotective, anti-platelet aggregation, antibacterial, antifungal, and broad antiviral activity. B. serrata and BA act by multiple mechanisms. The most common mechanism may be through direct interaction with IκB kinases and inhibiting nuclear factor-κB-regulated gene expression. However, the most recent mechanism proposed that BA not only inhibited the formation of classical 5-lipoxygenase products but also produced anti-inflammatory LOX-isoform-selective modulators. In conclusion a small to moderate dose B. serrata extract may be useful in the enhancing adaptive immune response in mild to moderate symptoms of COVID-19. However, large doses of BA may be beneficial in suppressing uncontrolled activation of the innate immune response. More clinical results are required to determine with certainty whether there is sufficient evidence of the benefits against COVID-19.
Collapse
Affiliation(s)
- Adel A Gomaa
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Hamdy S Mohamed
- Department of Internal Medicine, Faculty of Medicine, Sohage University, Sohâg, Egypt
| | | | - Mohamed A Gomaa
- Department of Plastic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
124
|
Affiliation(s)
- Justin Stebbing
- From the Department of Surgery and Cancer, Imperial College, London (J.S.); the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (V.M.L.); and the Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- From the Department of Surgery and Cancer, Imperial College, London (J.S.); the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (V.M.L.); and the Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
125
|
Smith DP, Oechsle O, Rawling MJ, Savory E, Lacoste AMB, Richardson PJ. Expert-Augmented Computational Drug Repurposing Identified Baricitinib as a Treatment for COVID-19. Front Pharmacol 2021; 12:709856. [PMID: 34393789 PMCID: PMC8356560 DOI: 10.3389/fphar.2021.709856] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
The onset of the 2019 Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic necessitated the identification of approved drugs to treat the disease, before the development, approval and widespread administration of suitable vaccines. To identify such a drug, we used a visual analytics workflow where computational tools applied over an AI-enhanced biomedical knowledge graph were combined with human expertise. The workflow comprised rapid augmentation of knowledge graph information from recent literature using machine learning (ML) based extraction, with human-guided iterative queries of the graph. Using this workflow, we identified the rheumatoid arthritis drug baricitinib as both an antiviral and anti-inflammatory therapy. The effectiveness of baricitinib was substantiated by the recent publication of the data from the ACTT-2 randomised Phase 3 trial, followed by emergency approval for use by the FDA, and a report from the CoV-BARRIER trial confirming significant reductions in mortality with baricitinib compared to standard of care. Such methods that iteratively combine computational tools with human expertise hold promise for the identification of treatments for rare and neglected diseases and, beyond drug repurposing, in areas of biological research where relevant data may be lacking or hidden in the mass of available biomedical literature.
Collapse
Affiliation(s)
| | | | | | - Ed Savory
- BenevolentAI, London, United Kingdom
| | | | | |
Collapse
|
126
|
Gatti M, Turrini E, Raschi E, Sestili P, Fimognari C. Janus Kinase Inhibitors and Coronavirus Disease (COVID)-19: Rationale, Clinical Evidence and Safety Issues. Pharmaceuticals (Basel) 2021; 14:ph14080738. [PMID: 34451835 PMCID: PMC8401109 DOI: 10.3390/ph14080738] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
We are witnessing a paradigm shift in drug development and clinical practice to fight the novel coronavirus disease (COVID-19), and a number of clinical trials have been or are being testing various pharmacological approaches to counteract viral load and its complications such as cytokine storm. However, data on the effectiveness of antiviral and immune therapies are still inconclusive and inconsistent. As compared to other candidate drugs to treat COVID-19, Janus Kinase (JAK) inhibitors, including baricitinib and ruxolitinib, possess key pharmacological features for a potentially successful repurposing: convenient oral administration, favorable pharmacokinetic profile, multifunctional pharmacodynamics by exerting dual anti-inflammatory and anti-viral effects. Baricitinib, originally approved for rheumatoid arthritis, received Emergency Use Authorization in November 2020 by the Food and Drug Administration in combination with remdesivir for the treatment of COVID-19 in hospitalized patients ≥ 2 years old who require supplemental oxygen, invasive mechanical ventilation, or extracorporeal membrane oxygenation. By July 2021, the European Medicines Agency is also expected to issue the opinion on whether or not to extend its use in hospitalised patients from 10 years of age who require supplemental oxygen. Ruxolitinib, approved for myelofibrosis, was prescribed in patients with COVID-19 within an open-label Emergency Expanded Access Plan. This review will address key milestones in the discovery and use of JAK inhibitors in COVID-19, from artificial intelligence to current clinical evidence, including real world experience, and critically appraise emerging safety issues, namely infections, thrombosis, and liver injury. An outlook to ongoing studies (ClinicalTrials.gov) and unpublished pharmacovigilance data is also offered.
Collapse
Affiliation(s)
- Milo Gatti
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum—Università di Bologna, Via Irnerio 48, 40126 Bologna, Italy;
- SSD Clinical Pharmacology, IRCCS Azienda Ospedaliero Universitaria Sant’Orsola, 40126 Bologna, Italy
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum—Università di Bologna, C.so D’Augusto 237, 47921 Rimini, Italy;
| | - Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum—Università di Bologna, Via Irnerio 48, 40126 Bologna, Italy;
- Correspondence: (E.R.); (C.F.)
| | - Piero Sestili
- Department of Biomolecular Sciences (DISB), Università degli Studi di Urbino Carlo Bo, Via I Maggetti 26, 61029 Urbino, Italy;
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum—Università di Bologna, C.so D’Augusto 237, 47921 Rimini, Italy;
- Correspondence: (E.R.); (C.F.)
| |
Collapse
|
127
|
Clément F, Nougarède A, Combe S, Kermarrec F, Dey AK, Obeid P, Millet A, Navarro FP, Marche PN, Sulpice E, Gidrol X. Therapeutic siRNAs Targeting the JAK/STAT Signalling Pathway in Inflammatory Bowel Diseases. J Crohns Colitis 2021; 16:286-300. [PMID: 34286840 PMCID: PMC8864631 DOI: 10.1093/ecco-jcc/jjab129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel diseases are highly debilitating conditions that require constant monitoring and life-long medication. Current treatments are focused on systemic administration of immunomodulatory drugs, but they have a broad range of undesirable side-effects. RNA interference is a highly specific endogenous mechanism that regulates the expression of the gene at the transcript level, which can be repurposed using exogenous short interfering RNA [siRNA] to repress expression of the target gene. While siRNA therapeutics can offer an alternative to existing therapies, with a high specificity critical for chronically administrated drugs, evidence of their potency compared to chemical kinase inhibitors used in clinics is still lacking in alleviating an adverse inflammatory response. METHODS We provide a framework to select highly specific siRNA, with a focus on two kinases strongly involved in pro-inflammatory diseases, namely JAK1 and JAK3. Using western-blot, real-time quantitative PCR and large-scale analysis, we assessed the specificity profile of these siRNA drugs and compared their efficacy to the most recent and promising kinase inhibitors for Janus kinases [Jakinibs], tofacitinib and filgotinib. RESULTS siRNA drugs can reach higher efficiency and selectivity at lower doses [5 pM vs 1 µM] than Jakinibs. Moreover, JAK silencing lasted up to 11 days, even with 6 h pulse transfection. CONCLUSIONS The siRNA-based drugs developed hold the potential to develop more potent therapeutics for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Flora Clément
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France,Univ. Grenoble Alpes, INSERM U1209, CNRS UMR5309, IAB, La Tronche, France
| | - Adrien Nougarède
- Univ. Grenoble Alpes, CEA, Leti, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Stéphanie Combe
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France
| | | | - Arindam K Dey
- Univ. Grenoble Alpes, INSERM U1209, CNRS UMR5309, IAB, La Tronche, France
| | - Patricia Obeid
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France
| | - Arnaud Millet
- Univ. Grenoble Alpes, Inserm U1209, CNRS UMR5309, Team Mechanobiology, immunity and Cancer, Institute for Advanced Biosciences, La Tronche, France
| | - Fabrice P Navarro
- Univ. Grenoble Alpes, CEA, Leti, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Patrice N Marche
- Univ. Grenoble Alpes, INSERM U1209, CNRS UMR5309, IAB, La Tronche, France
| | - Eric Sulpice
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France
| | - Xavier Gidrol
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France,Corresponding author: Xavier Gidrol, Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, F-38000, Grenoble, France. Tel: +(33)4 38 78 22 36; Fax: +(33)4 38 78 59 17;
| |
Collapse
|
128
|
Current Status of Baricitinib as a Repurposed Therapy for COVID-19. Pharmaceuticals (Basel) 2021; 14:ph14070680. [PMID: 34358107 PMCID: PMC8308612 DOI: 10.3390/ph14070680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 12/23/2022] Open
Abstract
The emergence of the COVID-19 pandemic has mandated the instant (re)search for potential drug candidates. In response to the unprecedented situation, it was recognized early that repurposing of available drugs in the market could timely save lives, by skipping the lengthy phases of preclinical and initial safety studies. BenevolentAI’s large knowledge graph repository of structured medical information suggested baricitinib, a Janus-associated kinase inhibitor, as a potential repurposed medicine with a dual mechanism; hindering SARS-CoV2 entry and combatting the cytokine storm; the leading cause of mortality in COVID-19. However, the recently-published Adaptive COVID-19 Treatment Trial-2 (ACTT-2) positioned baricitinib only in combination with remdesivir for treatment of a specific category of COVID-19 patients, whereas the drug is not recommended to be used alone except in clinical trials. The increased pace of data output in all life sciences fields has changed our understanding of data processing and manipulation. For the purpose of drug design, development, or repurposing, the integration of different disciplines of life sciences is highly recommended to achieve the ultimate benefit of using new technologies to mine BIG data, however, the final say remains to be concluded after the drug is used in clinical practice. This review demonstrates different bioinformatics, chemical, pharmacological, and clinical aspects of baricitinib to highlight the repurposing journey of the drug and evaluates its placement in the current guidelines for COVID-19 treatment.
Collapse
|
129
|
van Eijk LE, Binkhorst M, Bourgonje AR, Offringa AK, Mulder DJ, Bos EM, Kolundzic N, Abdulle AE, van der Voort PHJ, Olde Rikkert MGM, van der Hoeven JG, den Dunnen WFA, Hillebrands J, van Goor H. COVID-19: immunopathology, pathophysiological mechanisms, and treatment options. J Pathol 2021; 254:307-331. [PMID: 33586189 PMCID: PMC8013908 DOI: 10.1002/path.5642] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to spread globally despite the worldwide implementation of preventive measures to combat the disease. Although most COVID-19 cases are characterised by a mild, self-limiting disease course, a considerable subset of patients develop a more severe condition, varying from pneumonia and acute respiratory distress syndrome (ARDS) to multi-organ failure (MOF). Progression of COVID-19 is thought to occur as a result of a complex interplay between multiple pathophysiological mechanisms, all of which may orchestrate SARS-CoV-2 infection and contribute to organ-specific tissue damage. In this respect, dissecting currently available knowledge of COVID-19 immunopathogenesis is crucially important, not only to improve our understanding of its pathophysiology but also to fuel the rationale of both novel and repurposed treatment modalities. Various immune-mediated pathways during SARS-CoV-2 infection are relevant in this context, which relate to innate immunity, adaptive immunity, and autoimmunity. Pathological findings in tissue specimens of patients with COVID-19 provide valuable information with regard to our understanding of pathophysiology as well as the development of evidence-based treatment regimens. This review provides an updated overview of the main pathological changes observed in COVID-19 within the most commonly affected organ systems, with special emphasis on immunopathology. Current management strategies for COVID-19 include supportive care and the use of repurposed or symptomatic drugs, such as dexamethasone, remdesivir, and anticoagulants. Ultimately, prevention is key to combat COVID-19, and this requires appropriate measures to attenuate its spread and, above all, the development and implementation of effective vaccines. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Larissa E van Eijk
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Mathijs Binkhorst
- Department of Paediatrics, Subdivision of NeonatologyRadboud University Medical Center Amalia Children's HospitalNijmegenThe Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Annette K Offringa
- Microbiology and System BiologyNetherlands Organisation for Applied Scientific ResearchZeistThe Netherlands
| | - Douwe J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Eelke M Bos
- Department of NeurosurgeryErasmus University Medical CenterRotterdamThe Netherlands
| | - Nikola Kolundzic
- Stem Cell Laboratory, Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Assisted Conception Unit, Guy's HospitalLondonUK
| | - Amaal E Abdulle
- Department of Internal Medicine, Division of Vascular Medicine, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Peter HJ van der Voort
- Department of Critical Care, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Marcel GM Olde Rikkert
- Department of Geriatric MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Wilfred FA den Dunnen
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Jan‐Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
130
|
Čikoš A, Dragojević S, Kubiček A. Degradation products of azetidine core G334089 - Isolation, structure elucidation and pathway. J Pharm Biomed Anal 2021; 203:114232. [PMID: 34246845 DOI: 10.1016/j.jpba.2021.114232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
An extensive forced degradation study using hydrolytic degradation conditions was performed on G334089, the S-enantiomer of the free fatty acid receptor 2 (FFA2) antagonist GLPG0974, to identify the degradation product structures and discern degradation pathways. Not all degradation products generated ions in the MS spectra, while several others were isomers, so more rigorous degradation conditions were applied to increase the degradant yield. Esterification of the degradants facilitated isolation via preparative HPLC and subsequent NMR and MS characterisation. The determined structures, retention times and fragmentation patterns were used to identify the original degradation products and postulate a degradation pathway. In addition to the expected amide bond hydrolysis, a second degradation mechanism involving azetidine activation through formation of an azetidinium ion was demonstrated.
Collapse
Affiliation(s)
- Ana Čikoš
- Fidelta Ltd, Prilaz Baruna Filipovića 29, 10000, Zagreb, Croatia.
| | | | - Adrijana Kubiček
- Fidelta Ltd, Prilaz Baruna Filipovića 29, 10000, Zagreb, Croatia
| |
Collapse
|
131
|
Najafi Fard S, Petrone L, Petruccioli E, Alonzi T, Matusali G, Colavita F, Castilletti C, Capobianchi MR, Goletti D. In Vitro Models for Studying Entry, Tissue Tropism, and Therapeutic Approaches of Highly Pathogenic Coronaviruses. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8856018. [PMID: 34239932 PMCID: PMC8221881 DOI: 10.1155/2021/8856018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/27/2021] [Accepted: 06/05/2021] [Indexed: 12/31/2022]
Abstract
Coronaviruses (CoVs) are enveloped nonsegmented positive-sense RNA viruses belonging to the family Coronaviridae that contain the largest genome among RNA viruses. Their genome encodes 4 major structural proteins, and among them, the Spike (S) protein plays a crucial role in determining the viral tropism. It mediates viral attachment to the host cell, fusion to the membranes, and cell entry using cellular proteases as activators. Several in vitro models have been developed to study the CoVs entry, pathogenesis, and possible therapeutic approaches. This article is aimed at summarizing the current knowledge about the use of relevant methodologies and cell lines permissive for CoV life cycle studies. The synthesis of this information can be useful for setting up specific experimental procedures. We also discuss different strategies for inhibiting the binding of the S protein to the cell receptors and the fusion process which may offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Francesca Colavita
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| |
Collapse
|
132
|
Putlyaeva LV, Lukyanov KA. Studying SARS-CoV-2 with Fluorescence Microscopy. Int J Mol Sci 2021; 22:6558. [PMID: 34207305 PMCID: PMC8234815 DOI: 10.3390/ijms22126558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 coronavirus deeply affected the world community. It gave a strong impetus to the development of not only approaches to diagnostics and therapy, but also fundamental research of the molecular biology of this virus. Fluorescence microscopy is a powerful technology enabling detailed investigation of virus-cell interactions in fixed and live samples with high specificity. While spatial resolution of conventional fluorescence microscopy is not sufficient to resolve all virus-related structures, super-resolution fluorescence microscopy can solve this problem. In this paper, we review the use of fluorescence microscopy to study SARS-CoV-2 and related viruses. The prospects for the application of the recently developed advanced methods of fluorescence labeling and microscopy-which in our opinion can provide important information about the molecular biology of SARS-CoV-2-are discussed.
Collapse
Affiliation(s)
| | - Konstantin A. Lukyanov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia;
| |
Collapse
|
133
|
Giri A, Srinivasan A, Sundar IK. COVID-19: Sleep, Circadian Rhythms and Immunity - Repurposing Drugs and Chronotherapeutics for SARS-CoV-2. Front Neurosci 2021; 15:674204. [PMID: 34220430 PMCID: PMC8249936 DOI: 10.3389/fnins.2021.674204] [Citation(s) in RCA: 4] [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: 02/28/2021] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has affected nearly 28 million people in the United States and has caused more than five hundred thousand deaths as of February 21, 2021. As the novel coronavirus continues to take its toll in the United States and all across the globe, particularly among the elderly (>65 years), clinicians and translational researchers are taking a closer look at the nexus of sleep, circadian rhythms and immunity that may contribute toward a more severe coronavirus disease-19 (COVID-19). SARS-CoV-2-induced multi-organ failure affects both central and peripheral organs, causing increased mortality in the elderly. However, whether differences in sleep, circadian rhythms, and immunity between older and younger individuals contribute to the age-related differences in systemic dysregulation of target organs observed in SARS-CoV-2 infection remain largely unknown. Current literature demonstrates the emerging role of sleep, circadian rhythms, and immunity in the development of chronic pulmonary diseases and respiratory infections in human and mouse models. The exact mechanism underlying acute respiratory distress syndrome (ARDS) and other cardiopulmonary complications in elderly patients in combination with associated comorbidities remain unclear. Nevertheless, understanding the critical role of sleep, circadian clock dysfunction in target organs, and immune status of patients with SARS-CoV-2 may provide novel insights into possible therapies. Chronotherapy is an emerging concept that is gaining attention in sleep medicine. Accumulating evidence suggests that nearly half of all physiological functions follow a strict daily rhythm. However, healthcare professionals rarely take implementing timed-administration of drugs into consideration. In this review, we summarize recent findings directly relating to the contributing roles of sleep, circadian rhythms and immune response in modulating infectious disease processes, and integrate chronotherapy in the discussion of the potential drugs that can be repurposed to improve the treatment and management of COVID-19.
Collapse
Affiliation(s)
| | | | - Isaac Kirubakaran Sundar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
134
|
Ramasamy S, Subbian S. Critical Determinants of Cytokine Storm and Type I Interferon Response in COVID-19 Pathogenesis. Clin Microbiol Rev 2021; 34:e00299-20. [PMID: 33980688 PMCID: PMC8142516 DOI: 10.1128/cmr.00299-20] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a rapidly evolving pandemic worldwide with at least 68 million COVID-19-positive cases and a mortality rate of about 2.2%, as of 10 December 2020. About 20% of COVID-19 patients exhibit moderate to severe symptoms. Severe COVID-19 manifests as acute respiratory distress syndrome (ARDS) with elevated plasma proinflammatory cytokines, including interleukin 1β (IL-1β), IL-6, tumor necrosis factor α (TNF-α), C-X-C motif chemokine ligand 10 (CXCL10/IP10), macrophage inflammatory protein 1 alpha (MIP-1α), and chemokine (C-C motif) ligand 2 (CCL2), with low levels of interferon type I (IFN-I) in the early stage and elevated levels of IFN-I during the advanced stage of COVID-19. Most of the severe and critically ill COVID-19 patients have had preexisting comorbidities, including hypertension, diabetes, cardiovascular diseases, and respiratory diseases. These conditions are known to perturb the levels of cytokines, chemokines, and angiotensin-converting enzyme 2 (ACE2), an essential receptor involved in SARS-CoV-2 entry into the host cells. ACE2 downregulation during SARS-CoV-2 infection activates the angiotensin II/angiotensin receptor (AT1R)-mediated hypercytokinemia and hyperinflammatory syndrome. However, several SARS-CoV-2 proteins, including open reading frame 3b (ORF3b), ORF6, ORF7, ORF8, and the nucleocapsid (N) protein, can inhibit IFN type I and II (IFN-I and -II) production. Thus, hyperinflammation, in combination with the lack of IFN responses against SARS-CoV-2 early on during infection, makes the patients succumb rapidly to COVID-19. Therefore, therapeutic approaches involving anti-cytokine/anti-cytokine-signaling and IFN therapy would favor the disease prognosis in COVID-19. This review describes critical host and viral factors underpinning the inflammatory "cytokine storm" induction and IFN antagonism during COVID-19 pathogenesis. Therapeutic approaches to reduce hyperinflammation and their limitations are also discussed.
Collapse
Affiliation(s)
- Santhamani Ramasamy
- Public Health Research Institute (PHRI) at New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI) at New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
135
|
Assadiasl S, Fatahi Y, Mosharmovahed B, Mohebbi B, Nicknam MH. Baricitinib: From Rheumatoid Arthritis to COVID-19. J Clin Pharmacol 2021; 61:1274-1285. [PMID: 33870531 PMCID: PMC8250677 DOI: 10.1002/jcph.1874] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/11/2021] [Indexed: 12/19/2022]
Abstract
Baricitinib is a JAK1/2 inhibitor that was first approved for treating moderate to severe rheumatoid arthritis (RA) but that later showed considerable efficacy in the control of exaggerated inflammatory responses that occur in a wide range of diseases. There is a growing body of evidence, obtained from clinical trials and case reports, demonstrating clinical and paraclinical improvement in patients following administration of baricitinib including RA, systemic lupus erythematosus, psoriasis, atopic dermatitis, alopecia areata, interferon‐mediated autoinflammatory diseases, graft‐versus‐host disease, diabetic kidney disease, and, recently, coronavirus disease‐19. However, despite overall encouraging results, many adverse effects have been observed in baricitinib‐treated patients, ranging from simple infections to increased risk of malignancies, particularly in long‐term use. The significant efficacy of baricitinib, versus the probable adverse effects, urge further investigation before establishing it as a part of standard therapeutic protocols. Here, we have provided a review of the studies that have used baricitinib for treating various inflammatory disorders and summarized the advantages and disadvantages of its administration.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Banafsheh Mosharmovahed
- Department of Chemical Engineering-Pharmaceutical Engineering, University of Tehran, Tehran, Iran
| | - Bahareh Mohebbi
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Nicknam
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
136
|
Hsu J, Liu CH, Wei JC. Suggested Considerations for the Treatment of Rheumatic Diseases in Adult Patients With COVID-19: Comment on the Article by Mikuls et al. Arthritis Rheumatol 2021; 73:2355. [PMID: 34105903 PMCID: PMC8239925 DOI: 10.1002/art.41889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Jeffrey Hsu
- University of Virginia School of Medicine, Charlottesville, VA
| | - Chin-Hsiu Liu
- China Medical University Hospital and China Medical University, Taichung, Taiwan
| | - James C Wei
- Chung Shan Medical University Hospital Chung Shan Medical University and China Medical University, Taichung, Taiwan
| |
Collapse
|
137
|
Zhou Y, Xu J, Hou Y, Leverenz JB, Kallianpur A, Mehra R, Liu Y, Yu H, Pieper AA, Jehi L, Cheng F. Network medicine links SARS-CoV-2/COVID-19 infection to brain microvascular injury and neuroinflammation in dementia-like cognitive impairment. Alzheimers Res Ther 2021; 13:110. [PMID: 34108016 PMCID: PMC8189279 DOI: 10.1186/s13195-021-00850-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dementia-like cognitive impairment is an increasingly reported complication of SARS-CoV-2 infection. However, the underlying mechanisms responsible for this complication remain unclear. A better understanding of causative processes by which COVID-19 may lead to cognitive impairment is essential for developing preventive and therapeutic interventions. METHODS In this study, we conducted a network-based, multimodal omics comparison of COVID-19 and neurologic complications. We constructed the SARS-CoV-2 virus-host interactome from protein-protein interaction assay and CRISPR-Cas9-based genetic assay results and compared network-based relationships therein with those of known neurological manifestations using network proximity measures. We also investigated the transcriptomic profiles (including single-cell/nuclei RNA-sequencing) of Alzheimer's disease (AD) marker genes from patients infected with COVID-19, as well as the prevalence of SARS-CoV-2 entry factors in the brains of AD patients not infected with SARS-CoV-2. RESULTS We found significant network-based relationships between COVID-19 and neuroinflammation and brain microvascular injury pathways and processes which are implicated in AD. We also detected aberrant expression of AD biomarkers in the cerebrospinal fluid and blood of patients with COVID-19. While transcriptomic analyses showed relatively low expression of SARS-CoV-2 entry factors in human brain, neuroinflammatory changes were pronounced. In addition, single-nucleus transcriptomic analyses showed that expression of SARS-CoV-2 host factors (BSG and FURIN) and antiviral defense genes (LY6E, IFITM2, IFITM3, and IFNAR1) was elevated in brain endothelial cells of AD patients and healthy controls relative to neurons and other cell types, suggesting a possible role for brain microvascular injury in COVID-19-mediated cognitive impairment. Overall, individuals with the AD risk allele APOE E4/E4 displayed reduced expression of antiviral defense genes compared to APOE E3/E3 individuals. CONCLUSION Our results suggest significant mechanistic overlap between AD and COVID-19, centered on neuroinflammation and microvascular injury. These results help improve our understanding of COVID-19-associated neurological manifestations and provide guidance for future development of preventive or treatment interventions, although causal relationship and mechanistic pathways between COVID-19 and AD need future investigations.
Collapse
Affiliation(s)
- Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - James B Leverenz
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Asha Kallianpur
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Reena Mehra
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Haiyuan Yu
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14850, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, 14850, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, NY, 10065, USA
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lara Jehi
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
138
|
Santoso CS, Li Z, Rottenberg JT, Liu X, Shen VX, Fuxman Bass JI. Therapeutic Targeting of Transcription Factors to Control the Cytokine Release Syndrome in COVID-19. Front Pharmacol 2021; 12:673485. [PMID: 34163359 PMCID: PMC8215608 DOI: 10.3389/fphar.2021.673485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Treatment of the cytokine release syndrome (CRS) has become an important part of rescuing hospitalized COVID-19 patients. Here, we systematically explored the transcriptional regulators of inflammatory cytokines involved in the COVID-19 CRS to identify candidate transcription factors (TFs) for therapeutic targeting using approved drugs. We integrated a resource of TF-cytokine gene interactions with single-cell RNA-seq expression data from bronchoalveolar lavage fluid cells of COVID-19 patients. We found 581 significantly correlated interactions, between 95 TFs and 16 cytokines upregulated in the COVID-19 patients, that may contribute to pathogenesis of the disease. Among these, we identified 19 TFs that are targets of FDA approved drugs. We investigated the potential therapeutic effect of 10 drugs and 25 drugs combinations on inflammatory cytokine production, which revealed two drugs that inhibited cytokine production and numerous combinations that show synergistic efficacy in downregulating cytokine production. Further studies of these candidate repurposable drugs could lead to a therapeutic regimen to treat the CRS in COVID-19 patients.
Collapse
Affiliation(s)
| | - Zhaorong Li
- Bioinformatics Program, Boston University, Boston, MA, United States
| | | | - Xing Liu
- Department of Biology, Boston University, Boston, MA, United States
| | - Vivian X. Shen
- Department of Biology, Boston University, Boston, MA, United States
| | - Juan I. Fuxman Bass
- Department of Biology, Boston University, Boston, MA, United States
- Bioinformatics Program, Boston University, Boston, MA, United States
| |
Collapse
|
139
|
Vallianou NG, Tsilingiris D, Christodoulatos GS, Karampela Ι, Dalamaga M. Anti-viral treatment for SARS-CoV-2 infection: A race against time amidst the ongoing pandemic. Metabol Open 2021; 10:100096. [PMID: 34056571 PMCID: PMC8143911 DOI: 10.1016/j.metop.2021.100096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Remdesivir (GS-5734), a drug initially developed to treat hepatitis C and Ebola virus disease, was the first approved treatment for severe coronavirus disease 2019 (COVID-19). However, apart from remdesivir, there is a paucity of other specific anti-viral agents against SARS-CoV-2 infection. In 2017, researchers had documented the anti-coronavirus potential of remdesivir in animal models. At the same time, trials performed during two Ebola outbreaks in Africa showed that the drug was safe. Although vaccines against SARS-CoV-2 infection have emerged at an enormously high speed, equivalent results from efforts towards the development of anti-viral drugs, which could have played a truly life-saving role in the current stage of the pandemic, have been stagnating. In this review, we will focus on the current treatment options for COVID-19 which mainly consist of repurposed agents or treatments conferring passive immunity (convalescent plasma or monoclonal antibodies). Additionally, potential specific anti-viral therapies under development will be reviewed, such as the decoy miniprotein CTC-445.2d, protease inhibitors, mainly against the Main protein Mpro, nucleoside analogs, such as molnupiravir and compounds blocking the replication transcription complex proteins, such as zotatifin and plitidepsin. These anti-viral agents seem to be very promising but still require meticulous clinical trial testing in order to establish their efficacy and safety. The continuous emergence of viral variants may pose a real challenge to the scientific community towards that end. In this context, the advent of nanobodies together with the potential administration of a combination of anti-viral drugs could serve as useful tools in the armamentarium against COVID-19.
Collapse
Affiliation(s)
- Natalia G. Vallianou
- First Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Dimitrios Tsilingiris
- First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 17 St Thomas street, 11527, Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Greece
| | - Ιrene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini St, Haidari, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Greece
| |
Collapse
|
140
|
Beeraka NM, Tulimilli SV, Karnik M, Sadhu SP, Pragada RR, Aliev G, Madhunapantula SV. The Current Status and Challenges in the Development of Vaccines and Drugs against Severe Acute Respiratory Syndrome-Corona Virus-2 (SARS-CoV-2). BIOMED RESEARCH INTERNATIONAL 2021; 2021:8160860. [PMID: 34159203 PMCID: PMC8168478 DOI: 10.1155/2021/8160860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection causes coronavirus disease-19 (COVID-19), which is characterized by clinical manifestations such as pneumonia, lymphopenia, severe acute respiratory distress, and cytokine storm. S glycoprotein of SARS-CoV-2 binds to angiotensin-converting enzyme II (ACE-II) to enter into the lungs through membrane proteases consequently inflicting the extensive viral load through rapid replication mechanisms. Despite several research efforts, challenges in COVID-19 management still persist at various levels that include (a) availability of a low cost and rapid self-screening test, (b) lack of an effective vaccine which works against multiple variants of SARS-CoV-2, and (c) lack of a potent drug that can reduce the complications of COVID-19. The development of vaccines against SARS-CoV-2 is a complicated process due to the emergence of mutant variants with greater virulence and their ability to invoke intricate lung pathophysiology. Moreover, the lack of a thorough understanding about the virus transmission mechanisms and complete pathogenesis of SARS-CoV-2 is making it hard for medical scientists to develop a better strategy to prevent the spread of the virus and design a clinically viable vaccine to protect individuals from being infected. A recent report has tested the hypothesis of T cell immunity and found effective when compared to the antibody response in agammaglobulinemic patients. Understanding SARS-CoV-2-induced changes such as "Th-2 immunopathological variations, mononuclear cell & eosinophil infiltration of the lung and antibody-dependent enhancement (ADE)" in COVID-19 patients provides key insights to develop potential therapeutic interventions for immediate clinical management. Therefore, in this review, we have described the details of rapid detection methods of SARS-CoV-2 using molecular and serological tests and addressed different therapeutic modalities used for the treatment of COVID-19 patients. In addition, the current challenges against the development of vaccines for SARS-CoV-2 are also briefly described in this article.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow 119991, Russia
| | - SubbaRao V. Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| | - Medha Karnik
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| | - Surya P. Sadhu
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, 530003 Andhra Pradesh, India
| | - Rajeswara Rao Pragada
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, 530003 Andhra Pradesh, India
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow 119991, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region 142432, Russia
- Research Institute of Human Morphology, 3Tsyurupy Street, Moscow 117418, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore, 570015 Karnataka, India
| |
Collapse
|
141
|
Spinelli FR, Meylan F, O'Shea JJ, Gadina M. JAK inhibitors: Ten years after. Eur J Immunol 2021; 51:1615-1627. [PMID: 33930196 DOI: 10.1002/eji.202048922] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
The European Journal of Immunology was launched 50 years ago, coinciding with the discovery of many cytokines and growth factors and the emergence of an entirely new field of research. Ultimately, our knowledge about the biological activity of these factors allowed us to better understand how the immune system functions in the context of inflammatory and autoimmune diseases leading to the development of targeted biologic therapies. The study of cytokine signal transduction led to the discovery of Janus kinases (JAK), and the consideration of therapeutically targeting JAKs to treat immune and inflammatory diseases. This year also marks the tenth anniversary of the approval of the first JAK inhibitor (jakinib) and now there are a total of nine approved jakinibs for treatment of rheumatologic, dermatologic, gastrointestinal, and neoplastic indications and most recently COVID-19. Here, we summarized the discoveries that led to development of first-generation jakinibs, discussed some of the newer, possibly more selective jakinibs, as well as jakinibs that also target other kinases. We also illustrated the rationale behind the application of these drugs in the treatment of COVID-19 cytokine storm. In this review, we will discuss the clinical success of jakinibs, the gaps in our understanding of their biological activities as well as challenges in regard to their clinical application.
Collapse
Affiliation(s)
- Francesca Romana Spinelli
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Università di Roma, Rome, Italy
| | - Françoise Meylan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Translational Immunology Section, Bethesda, MD, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Translational Immunology Section, Bethesda, MD, USA
| |
Collapse
|
142
|
Qu C, Fuhler GM, Pan Y. Could Histamine H1 Receptor Antagonists Be Used for Treating COVID-19? Int J Mol Sci 2021; 22:5672. [PMID: 34073529 PMCID: PMC8199351 DOI: 10.3390/ijms22115672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 has rapidly become a pandemic worldwide, causing extensive and long-term health issues. There is an urgent need to identify therapies that limit SARS-CoV-2 infection and improve the outcome of COVID-19 patients. Unbalanced lung inflammation is a common feature in severe COVID-19 patients; therefore, reducing lung inflammation can undoubtedly benefit the clinical manifestations. Histamine H1 receptor (H1 receptor) antagonists are widely prescribed medications to treat allergic diseases, while recently it has emerged that they show significant promise as anti-SARS-CoV-2 agents. Here, we briefly summarize the novel use of H1 receptor antagonists in combating SARS-CoV-2 infection. We also describe the potential antiviral mechanisms of H1 receptor antagonists on SARS-CoV-2. Finally, the opportunities and challenges of the use of H1 receptor antagonists in managing COVID-19 are discussed.
Collapse
Affiliation(s)
- Changbo Qu
- Tomas Lindahl Nobel Laureate Laboratory, Precision Medicine Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China;
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Gwenny M. Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, 3015 CN Rotterdam, The Netherlands;
| | - Yihang Pan
- Tomas Lindahl Nobel Laureate Laboratory, Precision Medicine Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China;
| |
Collapse
|
143
|
Abstract
COVID-19 is characterized by a wide range of clinical manifestations, from asymptomatic to extremely severe. At the onset of the pandemic, it became clear that old age and chronic illness were the major risk factors. However, they do not fully explain the variety of symptoms and complications of the SARS-COV-2 coronavirus infection. The research on genetic risk factors for COVID-19 is still at its early stages. A number of mutations and polymorphisms have been identified that affect the structure and stability of proteins factors of susceptibility to SARS-COV-2 infection, as well as a predisposition to the development of respiratory failure and the need for intensive care. Most of the identified genetic factors are related to the function of the immune system. On the other hand, the genetic polymorphism of the virus itself affects the COVID-19 spread and severity of its course . The genome of the virus accumulates mutations and evolves towards increasing contagiousness, replicative ability and evasion from the host's immune system. Genetic determinants of the COVID-19 infection are potential therapeutic targets. Studying them will provide information for the development of drugs and vaccines to combat the pandemic.
Collapse
|
144
|
Chong ZZ, Souayah N. SARS-CoV-2 Induced Neurological Manifestations Entangles Cytokine Storm That Implicates For Therapeutic Strategies. Curr Med Chem 2021; 29:2051-2074. [PMID: 33970839 DOI: 10.2174/0929867328666210506161543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 11/22/2022]
Abstract
The new coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can present with neurological symptoms and induce neurological complications. The involvement in both the central and peripheral nervous systems in COVID-19 patients has been associated with direct invasion of the virus and the induction of cytokine storm. This review discussed the pathways for the virus invasion into the nervous system and characterized the SARS-CoV-2 induced cytokine storm. In addition, the mechanisms underlying the immune responses and cytokine storm induction after SARS-CoV-2 infection were also discussed. Although some neurological symptoms are mild and disappear after recovery from infection, some severe neurological complications contribute to the mortality of COVID-19 patients. Therefore, the insight into the cause of SARS-CoV-2 induced cytokine storm in context with neurological complications will formulate the novel management of the disease and further identify new therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Zhao-Zhong Chong
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan, China
| | - Nizar Souayah
- Department of Neurology, Rutgers New Jersey Medical School, 90 Bergen Street Room Suite 8100, Newark, NJ 07101, United States
| |
Collapse
|
145
|
Bonaventura A, Vecchié A, Dagna L, Martinod K, Dixon DL, Van Tassell BW, Dentali F, Montecucco F, Massberg S, Levi M, Abbate A. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol 2021; 21:319-329. [PMID: 33824483 PMCID: PMC8023349 DOI: 10.1038/s41577-021-00536-9] [Citation(s) in RCA: 624] [Impact Index Per Article: 156.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a clinical syndrome caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients with severe disease show hyperactivation of the immune system, which can affect multiple organs besides the lungs. Here, we propose that SARS-CoV-2 infection induces a process known as immunothrombosis, in which activated neutrophils and monocytes interact with platelets and the coagulation cascade, leading to intravascular clot formation in small and larger vessels. Microthrombotic complications may contribute to acute respiratory distress syndrome (ARDS) and other organ dysfunctions. Therapeutic strategies aimed at reducing immunothrombosis may therefore be useful. Several antithrombotic and immunomodulating drugs have been proposed as candidates to treat patients with SARS-CoV-2 infection. The growing understanding of SARS-CoV-2 infection pathogenesis and how it contributes to critical illness and its complications may help to improve risk stratification and develop targeted therapies to reduce the acute and long-term consequences of this disease.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
- Department of Internal Medicine, ASST dei Sette Laghi, Varese, Italy.
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Internal Medicine, ASST dei Sette Laghi, Varese, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Dave L Dixon
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcome Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Benjamin W Van Tassell
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcome Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genova - Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Steffen Massberg
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marcel Levi
- Department of Medicine and Cardio-metabolic Programme - NIHR UCLH/UCL BRC, University College London Hospitals NHS Foundation Trust, London, UK
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
146
|
Youhanna S, Wright SC, Lauschke VM. Organotypic human ex vivo models for coronavirus disease 2019 research and drug development. Curr Opin Pharmacol 2021; 59:11-18. [PMID: 34029832 PMCID: PMC8075816 DOI: 10.1016/j.coph.2021.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023]
Abstract
Since the discovery of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019, intense research efforts on an unprecedented scale have focused on the study of viral entry mechanisms and adaptive immunity. While the identification of angiotensin-converting enzyme 2 (ACE2) and other co-receptors has elucidated the molecular and structural basis for viral entry, the pathobiological mechanisms of SARS-CoV-2 in human tissues are less understood. Recent advances in bioengineering have opened opportunities for the use of organotypic human tissue models to investigate host-virus interactions and test antiviral drug candidates in a physiological context. Although it is too early to accurately quantify the added value of these systems compared with conventional cell systems, it can be assumed that these advanced three-dimensional (3D) models contribute toward improved result translation. This mini-review summarizes recent work to study SARS-CoV-2 infection in human 3D tissue models with an emphasis on the pharmacological tools that have been developed to understand and prevent viral entry and replication.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Shane C Wright
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
147
|
Singh N, Villoutreix BO. Resources and computational strategies to advance small molecule SARS-CoV-2 discovery: Lessons from the pandemic and preparing for future health crises. Comput Struct Biotechnol J 2021; 19:2537-2548. [PMID: 33936562 PMCID: PMC8074526 DOI: 10.1016/j.csbj.2021.04.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need to identify new therapies that prevent SARS-CoV-2 infection and improve the outcome of COVID-19 patients. This pandemic has thus spurred intensive research in most scientific areas and in a short period of time, several vaccines have been developed. But, while the race to find vaccines for COVID-19 has dominated the headlines, other types of therapeutic agents are being developed. In this mini-review, we report several databases and online tools that could assist the discovery of anti-SARS-CoV-2 small chemical compounds and peptides. We then give examples of studies that combined in silico and in vitro screening, either for drug repositioning purposes or to search for novel bioactive compounds. Finally, we question the overall lack of discussion and plan observed in academic research in many countries during this crisis and suggest that there is room for improvement.
Collapse
Affiliation(s)
- Natesh Singh
- Université de Paris, Inserm UMR 1141 NeuroDiderot, Robert-Debré Hospital, 75019 Paris, France
| | - Bruno O. Villoutreix
- Université de Paris, Inserm UMR 1141 NeuroDiderot, Robert-Debré Hospital, 75019 Paris, France
| |
Collapse
|
148
|
Ingelman-Sundberg M, Lauschke VM. 3D human liver spheroids for translational pharmacology and toxicology. Basic Clin Pharmacol Toxicol 2021; 130 Suppl 1:5-15. [PMID: 33872466 DOI: 10.1111/bcpt.13587] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Drug development is a failure-prone endeavour, and more than 85% of drugs fail during clinical development, showcasing that current preclinical systems for compound selection are clearly inadequate. Liver toxicity remains a major reason for safety failures. Furthermore, all efforts to develop pharmacological therapies for a variety of chronic liver diseases, such as non-alcoholic steatohepatitis (NASH) and fibrosis, remain unsuccessful. Considering the time and expense of clinical trials, as well as the substantial burden on patients, new strategies are thus of paramount importance to increase clinical success rates. To this end, human liver spheroids are becoming increasingly utilized as they allow to preserve patient-specific phenotypes and functions for multiple weeks in culture. We here review the recent application of such systems for i) predictive and mechanistic analyses of drug hepatotoxicity, ii) the evaluation of hepatic disposition and metabolite formation of low clearance drugs and iii) the development of drugs for metabolic and infectious liver diseases, including NASH, fibrosis, malaria and viral hepatitis. We envision that with increasing dissemination, liver spheroids might become the new gold standard for such applications in translational pharmacology and toxicology.
Collapse
Affiliation(s)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
149
|
Aldea M, Michot JM, Danlos FX, Ribas A, Soria JC. Repurposing of Anticancer Drugs Expands Possibilities for Antiviral and Anti-Inflammatory Discovery in COVID-19. Cancer Discov 2021; 11:1336-1344. [PMID: 33846172 DOI: 10.1158/2159-8290.cd-21-0144] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
In 2020, the COVID-19 pandemic led to an unprecedented destabilization of the world's health and economic systems. The rapid spread and life-threatening consequences of COVID-19 have imposed testing of repurposed drugs, by investigating interventions already used in other indications, including anticancer drugs. The contours of anticancer drug repurposing have been shaped by similarities between the pathogenesis of COVID-19 and malignancies, including abnormal inflammatory and immunologic responses. In this review, we discuss the salient positive and negative points of repurposing anticancer drugs to advance treatments for COVID-19. SIGNIFICANCE: Targeting anti-inflammatory pathways with JAK/STAT inhibitors or anticytokine therapies aiming to curb COVID-19-related cytokine storm, using antiangiogenic drugs to reduce vascular abnormalities or immune-checkpoint inhibitors to improve antiviral defenses, could be of value in COVID-19. However, conflicting data on drug efficacy point to the need for better patient selection and biomarker studies.
Collapse
Affiliation(s)
- Mihaela Aldea
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | | | - Francois-Xavier Danlos
- INSERM U1015, Gustave Roussy, Villejuif, France.,Paris Saclay University, Saint-Aubin, France
| | - Antoni Ribas
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angles, Los Angeles, California
| | - Jean-Charles Soria
- Drug Development Department, Gustave Roussy, Villejuif, France. .,Paris Saclay University, Saint-Aubin, France
| |
Collapse
|
150
|
Buszko M, Nita-Lazar A, Park JH, Schwartzberg PL, Verthelyi D, Young HA, Rosenberg AS. Lessons learned: new insights on the role of cytokines in COVID-19. Nat Immunol 2021; 22:404-411. [PMID: 33723418 PMCID: PMC9317654 DOI: 10.1038/s41590-021-00901-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the midst of resurging COVID-19 cases, the second NIH/FDA virtual COVID-19 and Cytokines symposium was held on 1 December 2020, focusing on longitudinal studies of COVID-19 immunity, including long-term consequences, potential associations with autoimmunity and the multisystem inflammatory syndrome in children (MIS-C).
Collapse
Affiliation(s)
- Maja Buszko
- Cellular Immunology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Pamela L Schwartzberg
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Daniela Verthelyi
- Laboratory of Innate Immunity, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Howard A Young
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, USA
| | - Amy S Rosenberg
- Laboratory of Immunology, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|