351
|
Cohen JB, D'Agostino McGowan L, Jensen ET, Rigdon J, South AM. Evaluating sources of bias in observational studies of angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker use during COVID-19: beyond confounding. J Hypertens 2021; 39:795-805. [PMID: 33186321 PMCID: PMC8164085 DOI: 10.1097/hjh.0000000000002706] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Concerns over ACE inhibitor or ARB use to treat hypertension during COVID-19 remain unresolved. Although studies using more robust methodologies provided some clarity, sources of bias persist and it remains critical to quickly address this question. In this review, we discuss pernicious sources of bias using a causal model framework, including time-varying confounder, collider, information, and time-dependent bias, in the context of recently published studies. We discuss causal inference methodologies that can address these issues, including causal diagrams, time-to-event analyses, sensitivity analyses, and marginal structural modeling. We discuss effect modification and we propose a role for causal mediation analysis to estimate indirect effects via mediating factors, especially components of the renin--angiotensin system. Thorough knowledge of these sources of bias and the appropriate methodologies to address them is crucial when evaluating observational studies to inform patient management decisions regarding whether ACE inhibitors or ARBs are associated with greater risk from COVID-19.
Collapse
Affiliation(s)
- Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Division of Public Health Sciences
| | - Joseph Rigdon
- Department of Biostatistics and Data Science, Division of Public Health Sciences
| | - Andrew M South
- Department of Epidemiology and Prevention, Division of Public Health Sciences
- Section of Nephrology, Department of Pediatrics, Brenner Children's Hospital
- Department of Surgery-Hypertension & Vascular Research
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
352
|
|
353
|
Krumm ZA, Lloyd GM, Francis CP, Nasif LH, Mitchell DA, Golde TE, Giasson BI, Xia Y. Precision therapeutic targets for COVID-19. Virol J 2021; 18:66. [PMID: 33781287 PMCID: PMC8006140 DOI: 10.1186/s12985-021-01526-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 01/18/2023] Open
Abstract
Beginning in late 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged as a novel pathogen that causes coronavirus disease 2019 (COVID-19). SARS-CoV-2 has infected more than 111 million people worldwide and caused over 2.47 million deaths. Individuals infected with SARS-CoV-2 show symptoms of fever, cough, dyspnea, and fatigue with severe cases that can develop into pneumonia, myocarditis, acute respiratory distress syndrome, hypercoagulability, and even multi-organ failure. Current clinical management consists largely of supportive care as commonly administered treatments, including convalescent plasma, remdesivir, and high-dose glucocorticoids. These have demonstrated modest benefits in a small subset of hospitalized patients, with only dexamethasone showing demonstrable efficacy in reducing mortality and length of hospitalization. At this time, no SARS-CoV-2-specific antiviral drugs are available, although several vaccines have been approved for use in recent months. In this review, we will evaluate the efficacy of preclinical and clinical drugs that precisely target three different, essential steps of the SARS-CoV-2 replication cycle: the spike protein during entry, main protease (MPro) during proteolytic activation, and RNA-dependent RNA polymerase (RdRp) during transcription. We will assess the advantages and limitations of drugs that precisely target evolutionarily well-conserved domains, which are less likely to mutate, and therefore less likely to escape the effects of these drugs. We propose that a multi-drug cocktail targeting precise proteins, critical to the viral replication cycle, such as spike protein, MPro, and RdRp, will be the most effective strategy of inhibiting SARS-CoV-2 replication and limiting its spread in the general population.
Collapse
Affiliation(s)
- Zachary A Krumm
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Grace M Lloyd
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Connor P Francis
- College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, 32610, USA
- UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Lith H Nasif
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Duane A Mitchell
- College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, 32610, USA
- UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Todd E Golde
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
354
|
Gili T, Benelli G, Buscarini E, Canetta C, La Piana G, Merli G, Scartabellati A, Viganò G, Sfogliarini R, Melilli G, Assandri R, Cazzato D, Rossi DS, Usai S, Caldarelli G, Tramacere I, Pellegata G, Lauria G. SARS-COV-2 comorbidity network and outcome in hospitalized patients in Crema, Italy. PLoS One 2021; 16:e0248498. [PMID: 33765013 PMCID: PMC7993836 DOI: 10.1371/journal.pone.0248498] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/28/2021] [Indexed: 01/10/2023] Open
Abstract
We report onset, course, correlations with comorbidities, and diagnostic accuracy of nasopharyngeal swab in 539 individuals suspected to carry SARS-COV-2 admitted to the hospital of Crema, Italy. All individuals underwent clinical and laboratory exams, SARS-COV-2 reverse transcriptase-polymerase chain reaction on nasopharyngeal swab, and chest X-ray and/or computed tomography (CT). Data on onset, course, comorbidities, number of drugs including angiotensin converting enzyme (ACE) inhibitors and angiotensin-II-receptor antagonists (sartans), follow-up swab, pharmacological treatments, non-invasive respiratory support, ICU admission, and deaths were recorded. Among 411 SARS-COV-2 patients (67.7% males) median age was 70.8 years (range 5-99). Chest CT was performed in 317 (77.2%) and showed interstitial pneumonia in 304 (96%). Fatality rate was 17.5% (74% males), with 6.6% in 60-69 years old, 21.1% in 70-79 years old, 38.8% in 80-89 years old, and 83.3% above 90 years. No death occurred below 60 years. Non-invasive respiratory support rate was 27.2% and ICU admission 6.8%. Charlson comorbidity index and high C-reactive protein at admission were significantly associated with death. Use of ACE inhibitors or sartans was not associated with outcomes. Among 128 swab negative patients at admission (63.3% males) median age was 67.7 years (range 1-98). Chest CT was performed in 87 (68%) and showed interstitial pneumonia in 76 (87.3%). Follow-up swab turned positive in 13 of 32 patients. Using chest CT at admission as gold standard on the entire study population of 539 patients, nasopharyngeal swab had 80% accuracy. Comorbidity network analysis revealed a more homogenous distribution 60-40 aged SARS-COV-2 patients across diseases and a crucial different interplay of diseases in the networks of deceased and survived patients. SARS-CoV-2 caused high mortality among patients older than 60 years and correlated with pre-existing multiorgan impairment.
Collapse
Affiliation(s)
| | | | | | - Ciro Canetta
- High Care Medical Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Emergency Medicine Unit Ospedale Maggiore di Crema, Crema, Italy
| | | | - Guido Merli
- Intensive Care Unit, Ospedale Maggiore di Crema, Crema, Italy
| | | | | | | | | | - Roberto Assandri
- Clinical Investigation Laboratory, Ospedale Maggiore di Crema, Crema, Italy
| | - Daniele Cazzato
- Department of Diagnostics and Technology, Ospedale Maggiore di Crema, Crema, Italy
| | | | - Susanna Usai
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Guido Caldarelli
- DSMN Ca’Foscari University, Venice Italy and ECLT, Venice, Italy
- ECLT, Venice, Italy
- ISC-CNR Uos "Sapienza", Rome, Italy
| | - Irene Tramacere
- Scientific Directorate, Ospedale Maggiore di Crema, Crema, Italy
| | | | - Giuseppe Lauria
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, University of Milan, Milan, Italy
| |
Collapse
|
355
|
Iheanacho CO, Odili VU, Eze UIH. Risk of SARS-CoV-2 infection and COVID-19 prognosis with the use of renin-angiotensin-aldosterone system (RAAS) inhibitors: a systematic review. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:73. [PMID: 33778087 PMCID: PMC7988641 DOI: 10.1186/s43094-021-00224-4] [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] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/09/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Angiotensin-converting-enzyme-2, being the receptor for SARS-CoV-2, is increased in the use of RAAS inhibitors. Therefore, concerns have been raised over risks of SARS-CoV-2 infection and poor prognosis of COVID-19 in persons with prior exposure to these drugs. This study aimed to systematically review available evidence for associations between exposure to RAAS inhibitors with susceptibility to SARS-CoV-2 infection and clinical outcomes in infected persons. It hopes to address the question on the effects of RAAS inhibitors on the risk of COVID-19 and its prognosis. MAIN BODY Search was conducted in the databases of PubMed, Scopus, Cochrane, Embase and MedRxiv.org from December 2019 to May 31, 2020, using relevant keywords. Additional articles were identified through hand-searching of reference lists. Studies that reported associations between positive tests to COVID-19 and use of RAAS inhibitors, and treatment outcomes of COVID-19 patients who had exposure to RAAS inhibitors were considered eligible. The Newcastle-Ottawa scale was used to assess risk of bias in individual studies. The review was conducted in line with Preferred Regulatory Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines 2009. From the 952 studies screened and 2 studies from reference hand-searching, 18 were reviewed. Four studies evaluated the risks for SARS-CoV-2 infection among RAAS inhibitors users, and 16 (including 2 of the 4 studies) evaluated the clinical outcomes associated with previous exposure to RAAS inhibitors. CONCLUSION Evidence does not suggest higher risks for SARS-CoV-2 infection or poor disease prognosis in the use of RAAS inhibitors. This suggests the continued use of RAAS inhibitors by patients with existing needs, which supports the position statements of American Heart Association and European societies for Cardiology. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1186/s43094-021-00224-4.
Collapse
Affiliation(s)
- Chinonyerem O. Iheanacho
- Department of Clinical Pharmacy and Public Health, Faculty of Pharmacy, University of Calabar, Calabar, Nigeria
| | - Valentine U. Odili
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, niversity of Benin, Benin City, Nigeria
| | - Uchenna I. H. Eze
- Department of Clinical Pharmacy and Biopharmacy, Faculty of Pharmacy, Olabisi Onabanjo University, Sagamu, Nigeria
| |
Collapse
|
356
|
Venketasubramanian N, Anderson C, Ay H, Aybek S, Brinjikji W, de Freitas GR, Del Brutto OH, Fassbender K, Fujimura M, Goldstein LB, Haberl RL, Hankey GJ, Heiss WD, Lestro Henriques I, Kase CS, Kim JS, Koga M, Kokubo Y, Kuroda S, Lee K, Lee TH, Liebeskind DS, Lip GYH, Meairs S, Medvedev R, Mehndiratta MM, Mohr JP, Nagayama M, Pantoni L, Papanagiotou P, Parrilla G, Pastori D, Pendlebury ST, Pettigrew LC, Renjen PN, Rundek T, Schminke U, Shinohara Y, Tang WK, Toyoda K, Wartenberg KE, Wasay M, Hennerici MG. Stroke Care during the COVID-19 Pandemic: International Expert Panel Review. Cerebrovasc Dis 2021; 50:245-261. [PMID: 33756459 PMCID: PMC8089455 DOI: 10.1159/000514155] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/16/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has placed a tremendous strain on healthcare services. This study, prepared by a large international panel of stroke experts, assesses the rapidly growing research and personal experience with COVID-19 stroke and offers recommendations for stroke management in this challenging new setting: modifications needed for prehospital emergency rescue and hyperacute care; inpatient intensive or stroke units; posthospitalization rehabilitation; follow-up including at-risk family and community; and multispecialty departmental developments in the allied professions. SUMMARY The severe acute respiratory syndrome coronavirus 2 uses spike proteins binding to tissue angiotensin-converting enzyme (ACE)-2 receptors, most often through the respiratory system by virus inhalation and thence to other susceptible organ systems, leading to COVID-19. Clinicians facing the many etiologies for stroke have been sobered by the unusual incidence of combined etiologies and presentations, prominent among them are vasculitis, cardiomyopathy, hypercoagulable state, and endothelial dysfunction. International standards of acute stroke management remain in force, but COVID-19 adds the burdens of personal protections for the patient, rescue, and hospital staff and for some even into the postdischarge phase. For pending COVID-19 determination and also for those shown to be COVID-19 affected, strict infection control is needed at all times to reduce spread of infection and to protect healthcare staff, using the wealth of well-described methods. For COVID-19 patients with stroke, thrombolysis and thrombectomy should be continued, and the usual early management of hypertension applies, save that recent work suggests continuing ACE inhibitors and ARBs. Prothrombotic states, some acute and severe, encourage prophylactic LMWH unless bleeding risk is high. COVID-19-related cardiomyopathy adds risk of cardioembolic stroke, where heparin or warfarin may be preferable, with experience accumulating with DOACs. As ever, arteritis can prove a difficult diagnosis, especially if not obvious on the acute angiogram done for clot extraction. This field is under rapid development and may generate management recommendations which are as yet unsettled, even undiscovered. Beyond the acute management phase, COVID-19-related stroke also forces rehabilitation services to use protective precautions. As with all stroke patients, health workers should be aware of symptoms of depression, anxiety, insomnia, and/or distress developing in their patients and caregivers. Postdischarge outpatient care currently includes continued secondary prevention measures. Although hoping a COVID-19 stroke patient can be considered cured of the virus, those concerned for contact safety can take comfort in the increasing use of telemedicine, which is itself a growing source of patient-physician contacts. Many online resources are available to patients and physicians. Like prior challenges, stroke care teams will also overcome this one. Key Messages: Evidence-based stroke management should continue to be provided throughout the patient care journey, while strict infection control measures are enforced.
Collapse
Affiliation(s)
| | - Craig Anderson
- The George Institute for Global Health, Camperdown, Washington, Australia
| | - Hakan Ay
- Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
- Takeda Pharmaceutical Co. Limited, Cambridge, Massachusetts, USA
| | - Selma Aybek
- Department of Neurology, University Hospital Inselspital, Bern University, Bern, Switzerland
| | - Waleed Brinjikji
- Department of Radiology, Vascular Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriel R de Freitas
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil
- Department of Neurology, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Oscar H Del Brutto
- School of Medicine, Universidad Espiritu Santo-Ecuador, Samborondón, Ecuador
| | - Klaus Fassbender
- Department of Neurology, Saarland University Medical Centre, Homburg, Germany
| | - Miki Fujimura
- Department of Neurosurgery, Kohnan Hospital, Sendai, Japan
- Division of Advanced Cerebrovascular Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Larry B Goldstein
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| | - Roman L Haberl
- Department of Neurology and Neurological Intensive Medicine, Munich Clinic gGmbH, Academic Teaching Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Graeme J Hankey
- Medical School, The University of Western Australia, Perth, Washington, Australia
| | | | - Isabel Lestro Henriques
- Department of Neurosciences, Neurology Service, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Carlos S Kase
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jong S Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Masatoshi Koga
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoshihiro Kokubo
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
| | - Kiwon Lee
- Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Tsong-Hai Lee
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - David S Liebeskind
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Stephen Meairs
- Department of Neurology, Universitätsmedizin Mannheim, Mannheim, Germany
| | - Roman Medvedev
- Research Center of Neurology, Moscow, Russian Federation
| | | | - Jay P Mohr
- Tananbaum Stroke Center, New York, New York, USA
| | - Masao Nagayama
- Department of Neurology, International University of Health and Welfare(IUHW), Graduate School of Medicine, Tokyo, Japan
| | - Leonardo Pantoni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Panagiotis Papanagiotou
- Department of Diagnostic and Interventional Neuroradiology, Klinikum Bremen-Mitte, Germany
- Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Guillermo Parrilla
- Department of Neurology, Interventional Neuroradiology, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Sarah T Pendlebury
- Departments of Internal Medicine and Geratology, John Radcliffe Hospital, Oxford, United Kingdom
- Centre for Prevention of Stroke and Dementia, University of Oxford, Oxford, United Kingdom
| | | | | | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ulf Schminke
- Department of Neurology, University Medicine, Greifswald, Germany
| | | | - Wai Kwong Tang
- Department of Psychiatry, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kazunori Toyoda
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Mohammad Wasay
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Michael G Hennerici
- Department of Neurology, Medical Faculty, Mannheim University of Heidelberg, Mannheim, Germany
| |
Collapse
|
357
|
Chang TS, Ding Y, Freund MK, Johnson R, Schwarz T, Yabu JM, Hazlett C, Chiang JN, Wulf DA, Geschwind DH, Butte MJ, Pasaniuc B. Pre-existing conditions in Hispanics/Latinxs that are COVID-19 risk factors. iScience 2021; 24:102188. [PMID: 33615196 PMCID: PMC7879099 DOI: 10.1016/j.isci.2021.102188] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/02/2021] [Accepted: 02/09/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has exposed health care disparities in minority groups including Hispanics/Latinxs (HL). Studies of COVID-19 risk factors for HL have relied on county-level data. We investigated COVID-19 risk factors in HL using individual-level, electronic health records in a Los Angeles health system between March 9, 2020, and August 31, 2020. Of 9,287 HL tested for SARS-CoV-2, 562 were positive. HL constituted an increasing percentage of all COVID-19 positive individuals as disease severity escalated. Multiple risk factors identified in Non-Hispanic/Latinx whites (NHL-W), like renal disease, also conveyed risk in HL. Pre-existing nonrheumatic mitral valve disorder was a risk factor for HL hospitalization but not for NHL-W COVID-19 or HL influenza hospitalization, suggesting it may be a specific HL COVID-19 risk. Admission laboratory values also suggested that HL presented with a greater inflammatory response. COVID-19 risk factors for HL can help guide equitable government policies and identify at-risk populations.
Collapse
Affiliation(s)
- Timothy S. Chang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yi Ding
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Malika K. Freund
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ruth Johnson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julie M. Yabu
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Chad Hazlett
- Department of Political Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Statistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey N. Chiang
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David A. Wulf
- Department of Political Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel H. Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Manish J. Butte
- Divisions of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bogdan Pasaniuc
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
358
|
Issa H, Eid AH, Berry B, Takhviji V, Khosravi A, Mantash S, Nehme R, Hallal R, Karaki H, Dhayni K, Faour WH, Kobeissy F, Nehme A, Zibara K. Combination of Angiotensin (1-7) Agonists and Convalescent Plasma as a New Strategy to Overcome Angiotensin Converting Enzyme 2 (ACE2) Inhibition for the Treatment of COVID-19. Front Med (Lausanne) 2021; 8:620990. [PMID: 33816521 PMCID: PMC8012486 DOI: 10.3389/fmed.2021.620990] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently the most concerning health problem worldwide. SARS-CoV-2 infects cells by binding to angiotensin-converting enzyme 2 (ACE2). It is believed that the differential response to SARS-CoV-2 is correlated with the differential expression of ACE2. Several reports proposed the use of ACE2 pharmacological inhibitors and ACE2 antibodies to block viral entry. However, ACE2 inhibition is associated with lung and cardiovascular pathology and would probably increase the pathogenesis of COVID-19. Therefore, utilizing ACE2 soluble analogs to block viral entry while rescuing ACE2 activity has been proposed. Despite their protective effects, such analogs can form a circulating reservoir of the virus, thus accelerating its spread in the body. Levels of ACE2 are reduced following viral infection, possibly due to increased viral entry and lysis of ACE2 positive cells. Downregulation of ACE2/Ang (1-7) axis is associated with Ang II upregulation. Of note, while Ang (1-7) exerts protective effects on the lung and cardiovasculature, Ang II elicits pro-inflammatory and pro-fibrotic detrimental effects by binding to the angiotensin type 1 receptor (AT1R). Indeed, AT1R blockers (ARBs) can alleviate the harmful effects associated with Ang II upregulation while increasing ACE2 expression and thus the risk of viral infection. Therefore, Ang (1-7) agonists seem to be a better treatment option. Another approach is the transfusion of convalescent plasma from recovered patients with deteriorated symptoms. Indeed, this appears to be promising due to the neutralizing capacity of anti-COVID-19 antibodies. In light of these considerations, we encourage the adoption of Ang (1-7) agonists and convalescent plasma conjugated therapy for the treatment of COVID-19 patients. This therapeutic regimen is expected to be a safer choice since it possesses the proven ability to neutralize the virus while ensuring lung and cardiovascular protection through modulation of the inflammatory response.
Collapse
Affiliation(s)
- Hawraa Issa
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
- College of Public Health, Phoenicia University, Zahrani, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Bassam Berry
- Institut Pasteur, Paris 6 University, Paris, France
| | - Vahideh Takhviji
- Transfusion Research Center, High Institute for Research and Education in Transfusion, Tehran, Iran
| | - Abbas Khosravi
- Transfusion Research Center, High Institute for Research and Education in Transfusion, Tehran, Iran
| | - Sarah Mantash
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Rawan Nehme
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Rawan Hallal
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Hussein Karaki
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Kawthar Dhayni
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
- EA7517, MP3CV, CURS, University of Picardie Jules Verne, Amiens, France
| | - Wissam H. Faour
- School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Nehme
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
359
|
Volpe M, Patrono C. A randomized trial supports the recommendation to continue treatment with ACEi or ARBs during hospitalization for COVID-19. Eur Heart J 2021; 42:1061-1062. [PMID: 33659979 PMCID: PMC7989525 DOI: 10.1093/eurheartj/ehab106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Massimo Volpe
- Cardiology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Carlo Patrono
- Pharmacology, Catholic University School of Medicine, Rome, Italy and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
360
|
De Luca G, Cercek M, Okkels Jensen L, Bushljetikj O, Calmac L, Johnson T, Gracida Blancas M, Ganyukov V, Wojakowski W, von Birgelen C, IJsselmuiden A, Tuccillo B, Versaci F, Ten Berg J, Laine M, Berkout T, Casella G, Kala P, López Ledesma B, Becerra V, Padalino R, Santucci A, Carrillo X, Scoccia A, Amoroso G, Lux A, Kovarnik T, Davlouros P, Gabrielli G, Flores Rios X, Bakraceski N, Levesque S, Guiducci V, Kidawa M, Marinucci L, Zilio F, Galasso G, Fabris E, Menichelli M, Manzo S, Caiazzo G, Moreu J, Sanchis Forés J, Donazzan L, Vignali L, Teles R, Agostoni P, Bosa Ojeda F, Lehtola H, Camacho-Freiere S, Kraaijeveld A, Antti Y, Visconti G, Lozano Martínez-Luengas I, Scheller B, Alexopulos D, Moreno R, Kedhi E, Uccello G, Faurie B, Gutierrez Barrios A, Scotto Di Uccio F, Wilbert B, Cortese G, Dirksen MT, Parodi G, Verdoia M. Impact of renin-angiotensin system inhibitors on mortality during the COVID Pandemic among STEMI patients undergoing mechanical reperfusion: Insight from an international STEMI registry. Biomed Pharmacother 2021; 138:111469. [PMID: 33740523 PMCID: PMC7962982 DOI: 10.1016/j.biopha.2021.111469] [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: 11/16/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
Background Concerns have been raised on a potential interaction between renin-angiotensin system inhibitors (RASI) and the susceptibility to coronavirus disease 2019 (COVID-19). No data have been so far reported on the prognostic impact of RASI in patients suffering from ST-elevation myocardial infarction (STEMI) during COVID-19 pandemic, which was the aim of the present study. Methods STEMI patients treated with primary percutaneous coronary intervention (PPCI) and enrolled in the ISACS-STEMI COVID-19 registry were included in the present sub-analysis and divided according to RASI therapy at admission. Results Our population is represented by 6095 patients, of whom 3654 admitted in 2019 and 2441 in 2020. No difference in the prevalence of SARSCoV2 infection was observed according to RASI therapy at admission (2.5% vs 2.1%, p = 0.5), which was associated with a significantly lower mortality (adjusted OR [95% CI]=0.68 [0.51–0.90], P = 0.006), confirmed in the analysis restricted to 2020 (adjusted OR [95% CI]=0.5[0.33–0.74], P = 0.001). Among the 5388 patients in whom data on in-hospital medication were available, in-hospital RASI therapy was associated with a significantly lower mortality (2.1% vs 16.7%, OR [95% CI]=0.11 [0.084–0.14], p < 0.0001), confirmed after adjustment in both periods. Among the 62 SARSCoV-2 positive patients, RASI therapy, both at admission or in-hospital, showed no prognostic effect. Conclusions This is the first study to investigate the impact of RASI therapy on the prognosis and SARSCoV2 infection of STEMI patients undergoing PPCI during the COVID-19 pandemic. Both pre-admission and in-hospital RASI were associated with lower mortality. Among SARSCoV2-positive patients, both chronic and in-hospital RASI therapy showed no impact on survival.
Collapse
Affiliation(s)
- Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Maggiore della Carità, Università del Piemonte Orientale, Italy.
| | - Miha Cercek
- Centre for Intensive Internal Medicine, University Medical Centre, Ljubljana, Slovenia
| | | | - Oliver Bushljetikj
- University Clinic for Cardiology, Medical Faculty, Ss' Cyril and Methodius University, Skopje, North Macedonia
| | | | - Tom Johnson
- Division of Cardiology, Bristol Heart Institute, University Hospitals Bristol NHSFT & University of Bristol, Bristol
| | | | - Vladimir Ganyukov
- Division of Cardiology, State Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Kemerovo, Russia
| | - Wojtek Wojakowski
- Division of Cardiology, Medical University of Silezia, Katowice, Poland
| | - Clemens von Birgelen
- Department of Cardiology, Medisch Spectrum Twente, Thoraxcentrum Twente, Enschede, The Netherlands
| | | | | | | | - Jurrien Ten Berg
- Division of Cardiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Mika Laine
- Division of Cardiology, Helsinki University Central Hospital, Finland
| | - Tim Berkout
- Division of Cardiology, Northwest Clinics Alkmaar, The Netherlands
| | - Gianni Casella
- Division of Cardiology, Ospedale Maggiore Bologna, Italy
| | - Petr Kala
- University Hospital Brno, Medical Faculty of Masaryk University Brno, Czech Republic
| | | | - Victor Becerra
- Hospital Clínico Universitario Virgen de la Victoria, Málaga, Spain
| | | | | | | | | | | | - Arpad Lux
- Maastricht University Medical Center, The Netherlands
| | | | - Periklis Davlouros
- Invasive Cardiology and Congenital Heart Disease, Patras University Hospital, Patras, Greece
| | - Gabriele Gabrielli
- Interventional Cardiology Unit, Azienda Ospedaliero Universitaria "Ospedali Riuniti", Ancona, Italy
| | | | | | - Sébastien Levesque
- Center Hospitalier Universitaire de Poitiers, Poitiers, University Hospital, Poitiers, France
| | | | - Michał Kidawa
- Central Hospital of Medical University of Lodz, Poland
| | - Lucia Marinucci
- Division of Cardiology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Pesaro, Italy
| | | | - Gennaro Galasso
- Division of Cardiology, Ospedale San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | - Enrico Fabris
- Azienda Ospedaliero - Universitaria Ospedali Riuniti Trieste, Italy
| | | | - Stephane Manzo
- Division of Cardiology, CHU Lariboisière, AP-HP, Paris VII University, INSERM UMRS 942, France
| | | | - Jose Moreu
- Division of Cardiology, Complejo Hospitalario de Toledo, Toledo, Spain
| | - Juan Sanchis Forés
- Division of Cardiology, Hospital Clinico Universitario de Valencia, Spain
| | - Luca Donazzan
- Division of Cardiology, Ospedale "S. Maurizio" Bolzano Ospedale "S. Maurizio" Bolzano (OR), Italy
| | - Luigi Vignali
- Interventional Cardiology Unit, Azienda Ospedaliera Sanitaria, Parma, Italy
| | - Rui Teles
- Division of Cardiology, Hospital de Santa Cruz, CHLO, Carnaxide, Portugal
| | | | - Francisco Bosa Ojeda
- Division of Cardiology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Heidi Lehtola
- Division of Cardiology, Oulu University Hospital, Finland
| | | | | | | | - Gabriella Visconti
- Division of Cardiology, Ospedale Santa Maria delle Grazie, Pozzuoli, Italy
| | | | - Bruno Scheller
- Division of Cardiology, Clinical and Experimental Interventional Cardiology, University of Saarland, Germany
| | | | - Raul Moreno
- Division of Cardiology, Hospital la Paz, Madrid, Spain
| | - Elvin Kedhi
- Division of Cardiology, St-Jan Hospital, Brugge, Belgium
| | | | - Benjamin Faurie
- Division of Cardiology, Groupe Hospitalier Mutualiste de Grenoble, France
| | | | | | - Bor Wilbert
- Division of Cardiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | | | | | - Guido Parodi
- Azienda Ospedaliero-Universitaria Sassari, Italy
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, Italy
| |
Collapse
|
361
|
Haroon S, Subramanian A, Cooper J, Anand A, Gokhale K, Byne N, Dhalla S, Acosta-Mena D, Taverner T, Okoth K, Wang J, Chandan JS, Sainsbury C, Zemedikun DT, Thomas GN, Parekh D, Marshall T, Sapey E, Adderley NJ, Nirantharakumar K. Renin-angiotensin system inhibitors and susceptibility to COVID-19 in patients with hypertension: a propensity score-matched cohort study in primary care. BMC Infect Dis 2021; 21:262. [PMID: 33722197 PMCID: PMC7957446 DOI: 10.1186/s12879-021-05951-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Renin-angiotensin system (RAS) inhibitors have been postulated to influence susceptibility to Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). This study investigated whether there is an association between their prescription and the incidence of COVID-19 and all-cause mortality. METHODS We conducted a propensity-score matched cohort study comparing the incidence of COVID-19 among patients with hypertension prescribed angiotensin-converting enzyme I (ACE) inhibitors or angiotensin II type-1 receptor blockers (ARBs) to those treated with calcium channel blockers (CCBs) in a large UK-based primary care database (The Health Improvement Network). We estimated crude incidence rates for confirmed/suspected COVID-19 in each drug exposure group. We used Cox proportional hazards models to produce adjusted hazard ratios for COVID-19. We assessed all-cause mortality as a secondary outcome. RESULTS The incidence rate of COVID-19 among users of ACE inhibitors and CCBs was 9.3 per 1000 person-years (83 of 18,895 users [0.44%]) and 9.5 per 1000 person-years (85 of 18,895 [0.45%]), respectively. The adjusted hazard ratio was 0.92 (95% CI 0.68 to 1.26). The incidence rate among users of ARBs was 15.8 per 1000 person-years (79 out of 10,623 users [0.74%]). The adjusted hazard ratio was 1.38 (95% CI 0.98 to 1.95). There were no significant associations between use of RAS inhibitors and all-cause mortality. CONCLUSION Use of ACE inhibitors was not associated with the risk of COVID-19 whereas use of ARBs was associated with a statistically non-significant increase compared to the use of CCBs. However, no significant associations were observed between prescription of either ACE inhibitors or ARBs and all-cause mortality.
Collapse
Affiliation(s)
- Shamil Haroon
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | | | - Jennifer Cooper
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Astha Anand
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Krishna Gokhale
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | | | - Samir Dhalla
- The Health Improvement Network (THIN), London, UK
| | | | - Thomas Taverner
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Kelvin Okoth
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jingya Wang
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Joht Singh Chandan
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Christopher Sainsbury
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Department of Diabetes, Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasglow, UK
| | | | - G Neil Thomas
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Department of Acute Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Tom Marshall
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Department of Acute Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, UK
- PIONEER, The Health Data Research UK Hub in Acute Care, Birmingham, UK
| | - Nicola J Adderley
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK.
| | - Krishnarajah Nirantharakumar
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Department of Diabetes and Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Midlands Health Data Research UK, Birmingham, UK
| |
Collapse
|
362
|
Grover A, Oberoi M. A systematic review and meta-analysis to evaluate the clinical outcomes in COVID-19 patients on angiotensin-converting enzyme inhibitors or angiotensin receptor blockers. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 7:148-157. [PMID: 32542337 PMCID: PMC7314072 DOI: 10.1093/ehjcvp/pvaa064] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) share their target receptor site with the SARS-CoV-2 virus, that may cause ACE2 receptor up-regulation which raised concerns regarding ACEI and ARB use in COVID-19 patients. However, many medical professional societies recommended their continued use given the paucity of clinical evidence, but there is a need for an updated systematic review and meta-analysis of the latest clinical studies. METHODS AND RESULTS A search was conducted on PubMed, Google Scholar, EMBASE, and various preprint servers for studies comparing clinical outcomes and mortality in COVID-19 patients on ACEIs and/or ARBs, and a meta-analysis was performed. A total of 16 studies were included for the review and meta-analysis. There were conflicting findings reported in the rates of severity and mortality in several studies. In a pooled analysis of four studies, there was a statistically non-significant association of ACEI/ARB use with lower odds of developing severe disease vs. non-users [odds ratio (OR) = 0.81, 95% confidence interval (CI): 0.41-1.58, I2=50.52, P-value = 0.53). In a pooled analysis of six studies, there was a statistically non-significant association of ACEI/ARB use with lower odds of mortality as compared with non-users (OR = 0.86, 95% CI = 0.53-1.41, I2 = 79.12, P-value = 0.55). CONCLUSION It is concluded that ACEIs and ARBs should be continued in COVID-19 patients, reinforcing the recommendations made by several medical societies. Additionally, the individual patient factors such as ACE2 polymorphisms which might confer higher risk of adverse outcomes need to be evaluated further.
Collapse
Affiliation(s)
- Abhinav Grover
- Internal Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Mansi Oberoi
- Internal Medicine, University of South Dakota, Sanford School of Medicine, Sioux Falls, SD, USA
| |
Collapse
|
363
|
Butler-Laporte G, Nakanishi T, Mooser V, Renieri A, Amitrano S, Zhou S, Chen Y, Forgetta V, Richards JB. The effect of angiotensin-converting enzyme levels on COVID-19 susceptibility and severity: a Mendelian randomization study. Int J Epidemiol 2021; 50:75-86. [PMID: 33349849 PMCID: PMC7799043 DOI: 10.1093/ije/dyaa229] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 11/14/2022] Open
Abstract
Background There has been uncertainty about the safety or benefit of angiotensin-converting enzyme (ACE) inhibitors during the COVID-19 pandemic. We used Mendelian randomization using genetic determinants of serum-ACE levels to test whether decreased ACE levels increase susceptibility to SARS-CoV-2 infection or COVID-19 severity, while reducing potential bias from confounding and reverse causation in observational studies. Methods Genetic variants strongly associated with ACE levels, which were nearby the ACE gene, were identified from the ORIGIN trial and a separate genome-wide association study (GWAS) of ACE levels from the AGES cohort. The ORIGIN trial included 4147 individuals of European and Latino ancestries. Sensitivity analyses were performed using a study of 3200 Icelanders. Cohorts from the COVID-19 Host Genetics Initiative GWAS of up to 960 186 individuals of European ancestry were used for COVID-19 susceptibility, hospitalization and severe-disease outcome. Results Genetic variants were identified that explain between 18% and 37% of variance in ACE levels. Using genetic variants from the ORIGIN trial, a standard-deviation decrease in ACE levels was not associated with an increase in COVID-19 susceptibility [odds ratio (OR): 1.02, 95% confidence interval (CI): 0.90, 1.15], hospitalization (OR: 0.86, 95% CI: 0.68, 1.08) or severe disease (OR: 0.74, 95% CI: 0.51, 1.06). Using genetic variants from the AGES cohort, the result was similar for susceptibility (OR: 0.98, 95% CI: 0.89, 1.09), hospitalization (OR: 0.86, 95% CI: 0.66, 1.11) and severity (OR: 0.75, 95% CI: 0.50, 1.14). Multiple-sensitivity analyses led to similar results. Conclusion Genetically decreased serum ACE levels were not associated with susceptibility to, or severity of, COVID-19 disease. These data suggest that individuals taking ACE inhibitors should not discontinue therapy during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Guillaume Butler-Laporte
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Tomoko Nakanishi
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada.,Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Vincent Mooser
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Canada Excellence Research Chair in Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Sara Amitrano
- Genetica Medica Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Sirui Zhou
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Yiheng Chen
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Vincenzo Forgetta
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - J Brent Richards
- Centre for Clinical Epidemiology Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.,Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Twin Research, King's College London, London, UK
| |
Collapse
|
364
|
Bergman J, Ballin M, Nordström A, Nordström P. Risk factors for COVID-19 diagnosis, hospitalization, and subsequent all-cause mortality in Sweden: a nationwide study. Eur J Epidemiol 2021; 36:287-298. [PMID: 33704634 PMCID: PMC7946619 DOI: 10.1007/s10654-021-00732-w] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/13/2021] [Indexed: 01/08/2023]
Abstract
We conducted a nationwide, registry-based study to investigate the importance of 34 potential risk factors for coronavirus disease 2019 (COVID-19) diagnosis, hospitalization (with or without intensive care unit [ICU] admission), and subsequent all-cause mortality. The study population comprised all COVID-19 cases confirmed in Sweden by mid-September 2020 (68,575 non-hospitalized, 2494 ICU hospitalized, and 13,589 non-ICU hospitalized) and 434,081 randomly sampled general-population controls. Older age was the strongest risk factor for hospitalization, although the odds of ICU hospitalization decreased after 60–69 years and, after controlling for other risk factors, the odds of non-ICU hospitalization showed no trend after 40–49 years. Residence in a long-term care facility was associated with non-ICU hospitalization. Male sex and the presence of at least one investigated comorbidity or prescription medication were associated with both ICU and non-ICU hospitalization. Three comorbidities associated with both ICU and non-ICU hospitalization were asthma, hypertension, and Down syndrome. History of cancer was not associated with COVID-19 hospitalization, but cancer in the past year was associated with non-ICU hospitalization, after controlling for other risk factors. Cardiovascular disease was weakly associated with non-ICU hospitalization for COVID-19, but not with ICU hospitalization, after adjustment for other risk factors. Excess mortality was observed in both hospitalized and non-hospitalized COVID-19 cases. These results confirm that severe COVID-19 is related to age, sex, and comorbidity in general. The study provides new evidence that hypertension, asthma, Down syndrome, and residence in a long-term care facility are associated with severe COVID-19.
Collapse
Affiliation(s)
- Jonathan Bergman
- Unit of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
| | - Marcel Ballin
- Unit of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden.,Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden
| | - Anna Nordström
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden.,School of Sport Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Peter Nordström
- Unit of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden.
| |
Collapse
|
365
|
Coto E, Avanzas P, Gómez J. The Renin-Angiotensin-Aldosterone System and Coronavirus Disease 2019. Eur Cardiol 2021; 16:e07. [PMID: 33737961 PMCID: PMC7967817 DOI: 10.15420/ecr.2020.30] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/10/2020] [Indexed: 12/21/2022] Open
Abstract
The renin-aldosterone-angiotensin system (RAAS) plays an important role in the pathogenesis of coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Angiotensin-converting enzyme 2 (ACE2) is the cellular receptor for SARS-CoV-2 and the host's expression of this membrane-bound protein could affect susceptibility to infection. The RAAS is an important regulator of cardiovascular physiology and ACE2 has an essential role. People with hypertension and other traits have shown to have an imbalance in ACE/ACE2 levels and reduced levels of ACE2 could enhance the risk of adverse outcome in patients with COVID-19. It has been hypothesised that the RAAS may mediate the interplay between cardiovascular disease and COVID-19 severity. Evidence shows that antihypertensive drugs that target the RAAS have no significant effect on the risk of infection and disease outcome. Variations in RAAS genes have been associated with the risk of developing hypertension and cardiovascular disease and could partly explain the heterogenous response to SARS-CoV-2 infection. This article explores the interplay between the RAAS and COVID-19, with emphasis on the possible relationship between genetic variations and disease severity.
Collapse
Affiliation(s)
- Eliecer Coto
- Genética Molecular, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain.,Universidad de Oviedo Oviedo, Spain.,Red de Investigación Renal (REDINREN) Madrid, Spain
| | - Pablo Avanzas
- Cardiología, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain.,Universidad de Oviedo Oviedo, Spain
| | - Juan Gómez
- Genética Molecular, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain
| |
Collapse
|
366
|
Zhang Y, Sha T, Wu F, Hu H, Chen Z, Li H, Han J, Song W, Huang Q, Zeng Z. Hypertension in Patients Hospitalized with COVID-19 in Wuhan, China. Int Heart J 2021; 62:337-343. [PMID: 33678794 DOI: 10.1536/ihj.20-323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
It is unclear whether patients with hypertension are more likely to be infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) than the general population and whether there is a difference in the severity of coronavirus disease (COVID-19) pneumonia in patients who have taken ACEI/ARB drugs compared with those who have not.This observational study included data from all patients with clinically confirmed COVID-19 admitted to Hankou Hospital, Wuhan, China, between January 5 and March 8, 2020. Data were extracted from clinical and laboratory records. Follow-up was cut off on March 8, 2020.A total of 274 patients, 75 with hypertension and 199 without hypertension, were included in the analysis. Compared with patients without hypertension, patients with hypertension were older and were more likely to have preexisting comorbidities, including chronic renal insufficiency, cardiovascular disease, diabetes mellitus, and cerebrovascular disease. Moreover, patients with hypertension tended to have higher positive rate for SARS-CoV-2 PCR detection. Multivariate logistic regression analysis showed that age (P = 0.005) and gender (P = 0.019) were independent risk factors associated with the severity of pneumonia in patients on admission, whereas ACEI/ARB treatment (P = 0.184) was not.Patients with COVID-19 with hypertension were significantly older and were more likely to have underlying comorbidities, including chronic renal insufficiency, cardiovascular disease, diabetes mellitus, and cerebrovascular disease. ACEI/ARB drugs did not influence the severity of pneumonia in patients with SARS-CoV-2. In future studies, a larger sample size and multi-center clinical data would be needed to support these conclusions.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University
| | - Tong Sha
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University
| | - Feng Wu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University
| | - Hongbin Hu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University
| | - Haijun Li
- Department of Radiology, Hankou Hospital of Wuhan
| | - Jiafa Han
- Department of Radiology, Hankou Hospital of Wuhan
| | - Wenhong Song
- Department of Radiology, Hankou Hospital of Wuhan
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University
| |
Collapse
|
367
|
COVID-19 and Vulnerable Populations in Sub-Saharan Africa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33656721 DOI: 10.1007/978-3-030-59261-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The novel corona virus 2019 (COVID-19) outbreak which started in Hubei province in China has now spread to every corner of the earth. While the pandemic started later in Africa, it is now found in all African countries to varying degrees. It is thought that the prevalence and severity of disease is influenced by a number of non-communicable diseases (NCDs) which are all becoming increasingly prevalent in sub-Saharan Africa (SSA). In addition, SSA bears the major burden of human immunodeficiency virus (HIV) and tuberculosis (TB) infections. While data from Europe and the United States show that children are spared severe disease, it is uncertain if the same holds true in SSA where children suffer from sickle cell disease and malnutrition in addition to other infectious diseases. There is limited data from Africa on the effects of these conditions on COVID-19. In this review, we discuss the epidemiology of some of these conditions in Africa and the possible pathogenesis for the interactions of these with COVID-19.
Collapse
|
368
|
Vervoort D, Ma X, Luc JGY. COVID-19 pandemic: a time for collaboration and a unified global health front. Int J Qual Health Care 2021; 33:mzaa065. [PMID: 32592480 PMCID: PMC7337739 DOI: 10.1093/intqhc/mzaa065] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/27/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Coronavirus disease 2019 has, in the span of weeks, immobilized entire countries and mobilized leading institutions worldwide in a race towards treatments and preventions. Although several solutions such as telemedicine and online education platforms have been implemented to reduce human contact and further transmission, countries need to favour collectivism both within and beyond their borders. Inspired by experiences of previous outbreaks such as SARS in 2003 and Ebola in 2014-2015, global solidarity is a must in order to prevent further morbidity and mortality. Examples in leadership and collaborations ranging from research funds from the Bill and Melinda Gates Foundation to mask donations by the Jack Ma Foundation should be celebrated as examples to follow. Open communication and transparency will be crucial in monitoring the evolution of the disease in the global effort of flattening the curve. This crisis will challenge the integrity and fuel innovation of health systems worldwide, whilst posing a new quality chasm that warrants increased recognition.
Collapse
Affiliation(s)
- Dominique Vervoort
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, 21205 Baltimore, Maryland, USA
| | - Xiya Ma
- Faculty of Medicine, Universite de Montreal, 2900 Edouard Montpetit Boulevard, Montreal, Quebec H3T 1J4, Canada
| | - Jessica G Y Luc
- Division of Cardiovascular Surgery, University of British Columbia, 2329 West Mall, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
369
|
Martínez-Botía P, Bernardo Á, Acebes-Huerta A, Caro A, Leoz B, Martínez-Carballeira D, Palomo-Antequera C, Soto I, Gutiérrez L. Clinical Management of Hypertension, Inflammation and Thrombosis in Hospitalized COVID-19 Patients: Impact on Survival and Concerns. J Clin Med 2021; 10:jcm10051073. [PMID: 33806709 PMCID: PMC7961450 DOI: 10.3390/jcm10051073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
The most severe clinical manifestations of the Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are due to an unbalanced immune response and a pro-thrombotic hemostatic disturbance, with arterial hypertension or diabetes as acknowledged risk factors. While waiting for a specific treatment, the clinical management of hospitalized patients is still a matter of debate, and the effectiveness of treatments to manage clinical manifestations and comorbidities has been questioned. In this study, we aim to assess the impact of the clinical management of arterial hypertension, inflammation and thrombosis on the survival of COVID-19 patients. The Spanish cohorts included in this observational retrospective study are from HM Hospitales (2035 patients) and from Hospital Universitario Central de Asturias (72 patients). Kaplan Meier survival curves, Cox regression and propensity score matching analyses were employed, considering demographic variables, comorbidities and treatment arms (when opportune) as covariates. The management of arterial hypertension with angiotensin-converting enzyme 2 (ACE2) inhibitors or angiotensin receptor blockers is not detrimental, as was initially reported, and neither was the use of non-steroidal anti-inflammatory drugs (NSAIDs). On the contrary, our analysis shows that the use on itself of corticosteroids is not beneficial. Importantly, the management of COVID-19 patients with low molecular weight heparin (LMWH) as an anticoagulant significantly improves the survival of hospitalized patients. These results delineate the current treatment options under debate, supporting the effectiveness of thrombosis prophylaxis on COVID-19 patients as a first-line treatment without the need for compromising the treatment of comorbidities, while suggesting cautiousness when administering corticosteroids.
Collapse
Affiliation(s)
- Patricia Martínez-Botía
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
| | - Ángel Bernardo
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Clinical Diagnosis Laboratory, Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Andrea Acebes-Huerta
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
| | - Alberto Caro
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Blanca Leoz
- Department of Intensive Medicine, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain;
- Translational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Daniel Martínez-Carballeira
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Carmen Palomo-Antequera
- Department of Internal Medicine, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain;
- Bone Metabolism, Vascular Metabolism and Chronic Inflammatory Diseases Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Inmaculada Soto
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
- Correspondence: (I.S.); (L.G.)
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (I.S.); (L.G.)
| |
Collapse
|
370
|
Biswas M, Kali MSK. Association of angiotensin-converting enzyme inhibitors and angiotensin-receptor blockers with risk of mortality, severity or SARS-CoV-2 test positivity in COVID-19 patients: meta-analysis. Sci Rep 2021; 11:5012. [PMID: 33658619 PMCID: PMC7930241 DOI: 10.1038/s41598-021-84678-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/18/2021] [Indexed: 01/10/2023] Open
Abstract
The effects of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) in the treatment of COVID-19 are highly debated. This study was aimed to assess aggregated risk by investigating the association of ACEIs/ARBs users against non-users of ACEIs/ARBs with the risk of mortality or severe clinical manifestations or magnitude of SARS-CoV-2 test positivity in COVID-19 patients. Systematic literature search was carried out in different databases for eligible studies. The pooled relative risks (RRs) were measured using RevMan software where P<0.05 was set as statistical significance. In total, 10 studies were included in this analysis. After pooled estimation, it was demonstrated that SARS-CoV-2 positive patients taking ACEIs/ARBs were not associated with an increased risk of mortality compared to those not taking ACEIs/ARBs (RR 0.89; 95% CI 0.64-1.23; P=0.48). Furthermore, the risk of composite severe clinical manifestations was not significantly different between the positive patients with or without ACEIs/ARBs users (RR 1.29; 95% CI 0.81-2.04; P=0.28). There was no risk difference for SARS-CoV-2 test positivity in patients with or without ACEIs/ARBs users (RR 1.00; 95% CI 0.95-1.05; P=0.91). These findings may augment current professional society guidelines for not discontinuing ACEIs/ARBs in treating COVID-19 patients where it is clinically indicated.
Collapse
Affiliation(s)
- Mohitosh Biswas
- Department of Pharmacy, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | | |
Collapse
|
371
|
Armstrong K, Soltoff A, Rieu-Werden M, Metlay J, Haas J. Use of angiotensin converting enzyme inhibitors and angiotensin receptor blockers associated with lower risk of COVID-19 in household contacts. PLoS One 2021; 16:e0247548. [PMID: 33651840 PMCID: PMC7924745 DOI: 10.1371/journal.pone.0247548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/09/2021] [Indexed: 01/08/2023] Open
Abstract
Background Use of angiotensin converting enzyme inhibitors (ACEI) and angiotensin receptor blockers (ARB) has been hypothesized to affect COVID-19 risk. Objective To examine the association between use of ACEI/ARB and household transmission of COVID-19. Methods We conducted a modified cohort study of household contacts of patients who tested positive for COVID-19 between March 4 and May 17, 2020 in a large Northeast US health system. Household members were identified by geocoding and full address matching with exclusion of addresses with >10 matched residents or known congregate living functions. Medication use, clinical conditions and sociodemographic characteristics were obtained from electronic medical record (EMR) data on cohort entry. Cohort members were followed for at least one month after exposure to determine who tested positive for SARS-CoV-2. Mixed effects logistic regression and propensity score analyses were used to assess adjusted associations between medication use and testing positive. Results 1,499 of the 9,101 household contacts were taking an ACEI or an ARB. Probability of COVID-19 diagnosis during the study period was slightly higher among ACEI/ARB users in unadjusted analyses. However, ACEI/ARB users were older and more likely to have clinical comorbidities so that use of ACEI/ARB was associated with a decreased risk of being diagnosed with COVID-19 in mixed effect models (OR 0.60, 95% CI 0.44–0.81) or propensity score analyses (predicted probability 18.6% in ACEI/ARB users vs. 24.5% in non-users, p = 0.03). These associations were similar within age and comorbidity subgroups, including patients with documented hypertension, diabetes or cardiovascular disease, as well as when including other medications in the models. Conclusions In this observational study of household transmission, use of ACEIs or ARBs was associated with a decreased risk of being diagnosed with COVID-19. While causality cannot be inferred from these observational data, our results support current recommendations to continue ACEI/ARB in individuals at risk of COVID-19 exposure.
Collapse
Affiliation(s)
- Katrina Armstrong
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| | - Alex Soltoff
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Meghan Rieu-Werden
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Joshua Metlay
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Jennifer Haas
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
372
|
Lee J, Jo SJ, Cho Y, Lee JH, Oh IY, Park JJ, Cho YS, Choi DJ. Effects of renin-angiotensin system blockers on the risk and outcomes of severe acute respiratory syndrome coronavirus 2 infection in patients with hypertension. Korean J Intern Med 2021; 36:S123-S131. [PMID: 32872731 PMCID: PMC8009159 DOI: 10.3904/kjim.2020.390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND/AIMS There are concerns that the use of renin-angiotensin system (RAS) blockers may increase the risk of being infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or progressing to a severe clinical course after infection. This this study aimed to investigate the influence of RAS blockers on the risk and severity of SARS-CoV-2 infection. METHODS We conducted a retrospective cohort study analyzing nationwide claims data of 215,184 adults who underwent SARS-CoV-2 tests in South Korea. The SARS-CoV-2 positive rates and clinical outcomes were evaluated according to the use of RAS blockers in patients with hypertension (n = 64,243). RESULTS In total, 38,919 patients with hypertension were on RAS blockers. The SARS-CoV-2 positive rates were significantly higher in the RAS blocker group than in the control group after adjustments (adjusted odds ratio [OR], 1.22; 95% confidence interval [CI], 1.10 to 1.36; p < 0.001), and matching by propensity score (adjusted OR, 1.16; 95% CI, 1.03 to 1.32; p = 0.017). Among the 1,609 SARS-CoV-2-positive patients with hypertension, the use of RAS blockers was not associated with poor outcomes, such as mortality (adjusted OR, 0.81; 95% CI, 0.56 to 1.17; p = 0.265), and a composite of admission to the intensive care unit and mortality (adjusted OR, 0.95; 95% CI, 0.73 to 1.22; p = 0.669). Analysis in the propensity scorematched population showed consistent results. CONCLUSION In this Korean nationwide claims dataset, the use of RAS blockers was associated with a higher risk to SARS-CoV-2 infection but not with higher mortality or other severe clinical courses.
Collapse
Affiliation(s)
- Jinwoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Jin Jo
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | - Youngjin Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Hyun Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Il-Young Oh
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Joo Park
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young-Seok Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong-Ju Choi
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
373
|
Williams B. Renin-angiotensin system inhibitors in hospitalised patients with COVID-19. THE LANCET. RESPIRATORY MEDICINE 2021; 9:221-222. [PMID: 33422264 PMCID: PMC7836517 DOI: 10.1016/s2213-2600(21)00003-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Bryan Williams
- Institute of Cardiovascular Sciences, University College London, London W1T 7DN, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
374
|
Tajbakhsh A, Gheibi Hayat SM, Taghizadeh H, Akbari A, Inabadi M, Savardashtaki A, Johnston TP, Sahebkar A. COVID-19 and cardiac injury: clinical manifestations, biomarkers, mechanisms, diagnosis, treatment, and follow up. Expert Rev Anti Infect Ther 2021; 19:345-357. [PMID: 32921216 DOI: 10.1080/14787210.2020.1822737] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/09/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) has the characteristics of high transmission, diverse clinical manifestations, and a long incubation period. In addition to infecting the respiratory system, COVID-19 also has adverse effects on the cardiovascular system. COVID-19 causes acute myocardial injuries, as well as chronic damage to the cardiovascular system. AREAS COVERED The present review is aimed at providing current information on COVID-19 and the cardiovascular system. PubMed, Scopus, Science direct, and Google Scholar were searched. EXPERT OPINION It is suggested that heart injury caused by COVID-19 infection might be an important cause of severe clinical phenotypes or adverse events in affected patients. Myocardial damage is closely related to the severity of the disease and even the prognosis in patients with COVID-19. In addition to disorders that are caused by COVID-19 on the cardiovascular system, more protection should be employed for patients with preexisting cardiovascular disease (CVD). Hence, it is very important that once relevant symptoms appear, patients with COVID-19 be rapidly treated to reduce mortality. Thus, early measurements of cardiac damage via biomarkers following hospitalization for COVID-19 infections in a patient with preexisting CVD are recommended, together with careful monitoring of any myocardial injury that might be caused by the infection.Abbreviations: ICU: An intensive care unit; 2019-nCoV: 2019 novel coronavirus; ACEI: ACE inhibitor; ACS: Acute coronary syndrome; ARDS: Acute respiratory distress syndrome; AT1R: Ang II type 1 receptor; ATP: Adenosine triphosphate; ACC: American College of Cardiology; ACE: Angiotensin converting enzyme; Ang II: Angiotensin II; ARB: Angiotensin II receptor blocker; AV block: Atrioventricular block; CAD: Coronary artery disease; CVD: Cardiovascular disease; CT: Computerized tomography; CHF: Congestive heart failure; CHD: Coronary heart disease; CK-MB: Creatine kinase isoenzyme-MB; CRP: C-reactive protein; cTnI: Cardiac troponin I; EAT: Epicardial adipose tissue; ECMO: Extracorporeal membrane oxygenation; FDA: Food and Drug Administration; G-CSF: Granulocyte colony-stimulating factor; HFrEF: HF with a reduced ejection fraction; synhACE2: Human isoform of ACE2; IL: Interleukin; IABP: Intra-aortic balloon counterpulsation; IP10: Interferon γ-induced protein 10 kDa; LPC: Lysophosphatidylcholine; Mas: Mitochondrial assembly receptor; MCP1: Monocyte chemoattractant protein-1; MERS: Middle East respiratory syndrome; MIP1a: macrophage inflammatory protein 1a: MOF: Multiple organ failure; MI: Myocardial infarction; MRI: Magnetic resonance imaging; MYO: Myohe-moglobin; NT-proBNP: N-terminal pro-brain natriuretic peptide; PCPS: Percutaneous cardiopulmonary assistance; rhACE2: Recombinant human ACE2; SARS: Severe acute respiratory syndrome; Th: T helper; RAS: Renin-angiotensin system; TNF-α: Tumor necrosis factor-α; WHO: World Health Organization.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibi Hayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hajar Taghizadeh
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Akbari
- Department of Anesthesiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Inabadi
- Department of Biology, Islamic Azad University, Jahrom Branch, Jahrom, Iran
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies' Shiraz University of Medical Sciences, Shiraz, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| |
Collapse
|
375
|
Seo J, Son M. Update on association between exposure to renin-angiotensin-aldosterone system inhibitors and coronavirus disease 2019 in South Korea. Korean J Intern Med 2021; 36:S114-S122. [PMID: 32872736 PMCID: PMC8009148 DOI: 10.3904/kjim.2020.380] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/18/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/AIMS Since the onset of the coronavirus disease 2019 (COVID-19) pandemic, there have been concerns about the association between exposure to renin-angiotensin-aldosterone system (RAAS) inhibitors and the risk and severity of COVID-19. METHODS We performed a case-control study that utilized up-to-date data on the South Korean population provided by the Korean National Health Insurance System. Of the 62,909 patients with hypertension or heart failure tested for COVID-19, there were 1,644 (2.6%) confirmed cases. After case-control matching, multivariable-adjusted conditional logistic regression analysis was performed. RESULTS Comparison between patients exposed to RAAS inhibitors and those not exposed to RAAS inhibitors revealed that the adjusted odds ratio (OR) and 95% confidence interval (CI) for COVID-19 infection and death were 0.981 (95% CI, 0.849 to 1.135) and 0.875 (95% CI, 0.548 to 1.396), respectively. Subgroup analysis for the major confounders, age and region of diagnosis, resulted in OR of 0.912 (95% CI, 0.751 to 1.108) and 0.942 (95% CI, 0.791 to 1.121), respectively. CONCLUSION The present study demonstrated no evidence of association between RAAS inhibitor exposure and risk and severity of COVID-19.
Collapse
Affiliation(s)
- Jeongkuk Seo
- Department of Internal Medicine, Armed Forces Goyang Hospital, Goyang, Korea
- Correspondence to Jeongkuk Seo, M.D. Department of Internal Medicine, Armed Forces Goyang Hospital, 111-8 Byeokje-dong, Deokyang-gu, Goyang 10267, Korea Tel: +82-31-963-6657 Fax: +82-31-964-9905 E-mail:
| | - Minkook Son
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
376
|
Ng TSB, Leblanc K, Yeung DF, Tsang TSM. Medication use during COVID-19: Review of recent evidence. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2021; 67:171-179. [PMID: 33727376 PMCID: PMC7963004 DOI: 10.46747/cfp.6703171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To keep health care providers, in response to the ongoing coronavirus disease 2019 (COVID-19) pandemic, informed about the medications that have been proposed to treat the disease and the evidence supporting their use. QUALITY OF EVIDENCE A narrative review of medications most widely used to treat COVID-19 was conducted, outlining the best available evidence for each pharmacologic treatment to date. Searches were performed in PubMed, EMBASE, and MEDLINE using key words COVID-19 and treatment, as well as related terms. Relevant research studies conducted in human populations and cases specific to patients with COVID-19 were included, as were relevant hand-searched papers and reviews. Only articles in English and Chinese were included. MAIN MESSAGE While current management of patients with COVID-19 largely involves supportive care, without a widely available vaccine, practitioners have also resorted to repurposing medications used for other indications. This has caused considerable controversy, as many of these treatments have limited clinical evidence supporting their use and therefore pose implications for patient safety, drug access, and public health. For instance, medications such as hydroxychloroquine and chloroquine, lopinavir-ritonavir, nonsteroidal anti-inflammatory drugs, angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers gained widespread media attention owing to hype, misinformation, or misinterpretation of research evidence. CONCLUSION Given the severity of the pandemic and the potential broad effects of implementing safe and effective treatment, this article provides a narrative review of the current evidence behind the most widely used medications to treat COVID-19 in order to enable health care practitioners to make informed decisions in the care of patients with this life-threatening disease.
Collapse
Affiliation(s)
- T S Brandon Ng
- Medical student in the Faculty of Medicine at the University of British Columbia in Vancouver
| | - Kori Leblanc
- Assistant Professor in the Leslie Dan Faculty of Pharmacy at the University of Toronto in Ontario
| | - Darwin F Yeung
- Clinical Assistant Professor in the Faculty of Medicine at the University of British Columbia
| | - Teresa S M Tsang
- Director of Echocardiography at Vancouver General Hospital and the UBC Hospital, and Professor of Medicine in the Division of Cardiology and Associate Head of Research for the Department of Medicine at the University of British Columbia.
| |
Collapse
|
377
|
Pathangey G, Fadadu PP, Hospodar AR, Abbas AE. Angiotensin-converting enzyme 2 and COVID-19: patients, comorbidities, and therapies. Am J Physiol Lung Cell Mol Physiol 2021; 320:L301-L330. [PMID: 33237815 PMCID: PMC7938645 DOI: 10.1152/ajplung.00259.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
On March 11, 2020, the World Health Organization declared coronavirus disease 2019 (COVID-19) a pandemic, and the reality of the situation has finally caught up to the widespread reach of the disease. The presentation of the disease is highly variable, ranging from asymptomatic carriers to critical COVID-19. The availability of angiotensin-converting enzyme 2 (ACE2) receptors may reportedly increase the susceptibility and/or disease progression of COVID-19. Comorbidities and risk factors have also been noted to increase COVID-19 susceptibility. In this paper, we hereby review the evidence pertaining to ACE2's relationship to common comorbidities, risk factors, and therapies associated with the susceptibility and severity of COVID-19. We also highlight gaps of knowledge that require further investigation. The primary comorbidities of respiratory disease, cardiovascular disease, renal disease, diabetes, obesity, and hypertension had strong evidence. The secondary risk factors of age, sex, and race/genetics had limited-to-moderate evidence. The tertiary factors of ACE inhibitors and angiotensin II receptor blockers had limited-to-moderate evidence. Ibuprofen and thiazolidinediones had limited evidence.
Collapse
Affiliation(s)
- Girish Pathangey
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
| | | | | | - Amr E Abbas
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
- Department of Cardiovascular Medicine, Beaumont Hospital Royal Oak, Royal Oak, Michigan
| |
Collapse
|
378
|
Li D, Ding X, Xie M, Tian D, Xia L. COVID-19-associated liver injury: from bedside to bench. J Gastroenterol 2021; 56:218-230. [PMID: 33527211 PMCID: PMC7849620 DOI: 10.1007/s00535-021-01760-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been a global challenge since December 2019. Although most patients with COVID-19 exhibit mild clinical manifestations, in approximately 5% of these patients, the disease eventually progresses to severe lung injury or even multiorgan dysfunction. This situation represents various challenges to hepatology. In the context of liver injury in patients with COVID-19, several key problems need to be solved. For instance, it is important to determine whether SARS-CoV-2 can directly invade liver, especially when ACE2 appears to be negligibly expressed on hepatocytes. In addition, the mechanisms underlying liver dysfunction in COVID-19 patients are not fully understood, which are likely multifactorial and related to hyperinflammation, dysregulated immune responses, abnormal coagulation and drugs. Here, we systematically describe the potential pathogenesis of COVID-19-associated liver injury and propose several hypotheses about its etiopathogenesis.
Collapse
Affiliation(s)
- Dongxiao Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiangming Ding
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan Province, China
| | - Meng Xie
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dean Tian
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
379
|
Chong WH, Saha BK. Relationship Between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Etiology of Acute Kidney Injury (AKI). Am J Med Sci 2021; 361:287-296. [PMID: 33358501 PMCID: PMC7590839 DOI: 10.1016/j.amjms.2020.10.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/23/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since it was first recognized in December 2019, it has resulted in the ongoing worldwide pandemic. Although acute hypoxic respiratory failure (AHRF) and acute respiratory distress syndrome (ARDS) are the main features of the disease, the involvement of other organs needs to be explored. There has been a growing concern regarding the association between acute kidney injury (AKI) and poor outcomes in SARS-CoV-2 patients. Based on current observational data, AKI is the 2nd most common cause of morbidity and mortality behind ARDS in SARS-CoV-2 patients. Angiotensin-converting enzyme 2 (ACE2) receptor has been shown to be the cornerstone of SARS-CoV-2 infection and possibly plays a significant role in the occurrence of renal injury. The pathogenesis of AKI is likely multifactorial that involves not only direct viral invasion but also dysregulated immune response in the form of cytokine storm, ischemia to kidneys, hypercoagulable state, and rhabdomyolysis, among others. We performed a literature search of the Pubmed and Google Scholar database from 1996 to 2020 using the following keywords: severe acute respiratory syndrome coronavirus 2, coronavirus disease 2019, angiotensin-converting enzyme 2 receptor, and acute kidney injury to find the most pertinent and highest-quality of evidence. Any cited references were reviewed to identify relevant literature. The purpose of this review is to discuss, explore, and summarize the relationship between AKI in SARS-CoV-2 patients, with a focus on its epidemiology, association with ACE2 receptors, and pathophysiology of AKI.
Collapse
Affiliation(s)
- Woon H Chong
- Department of Pulmonary and Critical Care Medicine; Albany Medical Center, Albany, New York, USA.
| | - Biplab K Saha
- Department of Pulmonary and Critical Care; Ozarks Medical Center, West Plains, Missouri, USA
| |
Collapse
|
380
|
Darwesh AM, Bassiouni W, Sosnowski DK, Seubert JM. Can N-3 polyunsaturated fatty acids be considered a potential adjuvant therapy for COVID-19-associated cardiovascular complications? Pharmacol Ther 2021; 219:107703. [PMID: 33031856 PMCID: PMC7534795 DOI: 10.1016/j.pharmthera.2020.107703] [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: 08/13/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has currently led to a global pandemic with millions of confirmed and increasing cases around the world. The novel SARS-CoV-2 not only affects the lungs causing severe acute respiratory dysfunction but also leads to significant dysfunction in multiple organs and physiological systems including the cardiovascular system. A plethora of studies have shown the viral infection triggers an exaggerated immune response, hypercoagulation and oxidative stress, which contribute significantly to poor cardiovascular outcomes observed in COVID-19 patients. To date, there are no approved vaccines or therapies for COVID-19. Accordingly, cardiovascular protective and supportive therapies are urgent and necessary to the overall prognosis of COVID-19 patients. Accumulating literature has demonstrated the beneficial effects of n-3 polyunsaturated fatty acids (n-3 PUFA) toward the cardiovascular system, which include ameliorating uncontrolled inflammatory reactions, reduced oxidative stress and mitigating coagulopathy. Moreover, it has been demonstrated the n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are precursors to a group of potent bioactive lipid mediators, generated endogenously, which mediate many of the beneficial effects attributed to their parent compounds. Considering the favorable safety profile for n-3 PUFAs and their metabolites, it is reasonable to consider n-3 PUFAs as potential adjuvant therapies for the clinical management of COVID-19 patients. In this article, we provide an overview of the pathogenesis of cardiovascular complications secondary to COVID-19 and focus on the mechanisms that may contribute to the likely benefits of n-3 PUFAs and their metabolites.
Collapse
Affiliation(s)
- Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
381
|
Richez C, Flipo RM, Berenbaum F, Cantagrel A, Claudepierre P, Debiais F, Dieudé P, Goupille P, Roux C, Schaeverbeke T, Wendling D, Pham T, Thomas T. Prise en charge des patients atteints de maladies rhumatismales pendant la pandémie de COVID-19 : la Société française de rhumatologie répond aux questions fréquentes posées jusqu’en mai 2020. REVUE DU RHUMATISME 2021; 88:93-100. [PMID: 33488063 PMCID: PMC7813495 DOI: 10.1016/j.rhum.2021.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 05/19/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Christophe Richez
- FHU ACRONIM, CHU de Bordeaux, place Amélie-Raba-Léon, 33076 Bordeaux, France
| | - René-Marc Flipo
- Service de rhumatologie, centre hospitalier régional universitaire de Lille, hôpital Roger-Salengro, rue du Professeur Émile-Laine, 59037 Lille cedex, France
| | - Francis Berenbaum
- Service de rhumatologie, CHU de Saint-Antoine, 184, rue du Faubourg Saint-Antoine, 755571 Paris cedex 12, France
| | - Alain Cantagrel
- Service de rhumatologie, CHU de Toulouse, Hôpital Purpan, place du Docteur Baylac, TSA 40031, 31059 Toulouse cedex 9, France
| | - Pascal Claudepierre
- Service de rhumatologie, CHU de Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France
| | - Françoise Debiais
- Service de rhumatologie, CHU, 2, rue de la Milétrie, BP 577, 86021 Poitiers cedex, France
| | - Philippe Dieudé
- Service de rhumatologie, groupe hospitalier universitaire Bichat-Claude Bernard, 46, rue Henri-Huchard, 75018 Paris, France
| | - Philippe Goupille
- Service de rhumatologie, centre hospitalier régional universitaire de Tours, hôpital Trousseau, 37044 Tours cedex 9, France
| | - Christian Roux
- Service de rhumatologie, CHU de Cochin, 27, rue du Faubourg Saint-Jacques, 75679 Paris cedex 14, France
| | | | - Daniel Wendling
- Service de rhumatologie, CHU de Jean-Minjoz, 1, boulevard Fleming, 25030 Besançon cedex, France
| | - Thao Pham
- Service de rhumatologie, CHU de Sainte-Marguerite, 270, boulevard de Sainte-Marguerite, 13274 Marseille cedex 9, France
| | - Thierry Thomas
- Department of Rheumatology, CHU de Saint-Étienne, Hôpital Nord, Saint-Étienne, France
- Inserm U1059, université de Lyon, université Jean-Monnet, Saint-Étienne, France
| |
Collapse
|
382
|
Gilbert RE, Caldwell L, Misra PS, Chan K, Burns KD, Wrana JL, Yuen DA. Overexpression of the Severe Acute Respiratory Syndrome Coronavirus-2 Receptor, Angiotensin-Converting Enzyme 2, in Diabetic Kidney Disease: Implications for Kidney Injury in Novel Coronavirus Disease 2019. Can J Diabetes 2021; 45:162-166.e1. [PMID: 32917504 PMCID: PMC7368650 DOI: 10.1016/j.jcjd.2020.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Diabetes is associated with adverse outcomes, including death, after coronavirus disease 19 (COVID-19) infection. Beyond the lungs, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), the etiologic agent of the COVID-19 pandemic, can infect a range of other tissues, including the kidney, potentially contributing to acute kidney injury in those with severe disease. We hypothesized that the renal abundance of angiotensin-converting enzyme (ACE) 2, the cell surface receptor for SARS-CoV-2, may be modulated by diabetes and agents that block the renin-angiotensin-aldosterone system (RAAS). METHODS The expression of ACE 2 was examined in 49 archival kidney biopsies from patients with diabetic kidney disease and from 12 healthy, potential living allograft donors using next-generation sequencing technology (RNA Seq). RESULTS Mean ACE 2 messenger RNA was increased approximately 2-fold in diabetes when compared with healthy control subjects (mean ± SD, 13.2±7.9 vs 7.7±3.6 reads per million reads, respectively; p=0.001). No difference in transcript abundance was noted between recipients and nonrecipients of agents that block the RAAS (12.2±6.7 vs 16.2±10.7 reads per million reads, respectively; p=0.25). CONCLUSIONS Increased ACE 2 messenger RNA in the diabetic kidney may increase the risk and/or severity of kidney infection with SARS-CoV-2 in the setting of COVID-19 disease. Further studies are needed to ascertain whether this diabetes-related overexpression is generalizable to other tissues, most notably the lungs.
Collapse
Affiliation(s)
- Richard E Gilbert
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Lauren Caldwell
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Paraish S Misra
- Division of Nephrology, Department of Medicine, University of Toronto, Ontario, Ontario, Canada
| | - Kin Chan
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Darren A Yuen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
383
|
Khawaja SA, Mohan P, Jabbour R, Bampouri T, Bowsher G, Hassan AMM, Huq F, Baghdasaryan L, Wang B, Sethi A, Sen S, Petraco R, Ruparelia N, Nijjer S, Malik I, Foale R, Bellamy M, Kooner J, Rana B, Cole G, Sutaria N, Kanaganayagam G, Nihoyannopoulos P, Fox K, Plymen C, Pabari P, Howard L, Davies R, Haji G, Lo Giudice F, Kanagaratnam P, Anderson J, Chukwuemeka A, Khamis R, Varnava A, Baker CSR, Francis DP, Asaria P, Al-Lamee R, Mikhail GW. COVID-19 and its impact on the cardiovascular system. Open Heart 2021; 8:e001472. [PMID: 33723014 PMCID: PMC7969760 DOI: 10.1136/openhrt-2020-001472] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/23/2020] [Accepted: 02/22/2021] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES The clinical impact of SARS-CoV-2 has varied across countries with varying cardiovascular manifestations. We review the cardiac presentations, in-hospital outcomes and development of cardiovascular complications in the initial cohort of SARS-CoV-2 positive patients at Imperial College Healthcare National Health Service Trust, UK. METHODS We retrospectively analysed 498 COVID-19 positive adult admissions to our institute from 7 March to 7 April 2020. Patient data were collected for baseline demographics, comorbidities and in-hospital outcomes, especially relating to cardiovascular intervention. RESULTS Mean age was 67.4±16.1 years and 62.2% (n=310) were male. 64.1% (n=319) of our cohort had underlying cardiovascular disease (CVD) with 53.4% (n=266) having hypertension. 43.2%(n=215) developed acute myocardial injury. Mortality was significantly increased in those patients with myocardial injury (47.4% vs 18.4%, p<0.001). Only four COVID-19 patients had invasive coronary angiography, two underwent percutaneous coronary intervention and one required a permanent pacemaker implantation. 7.0% (n=35) of patients had an inpatient echocardiogram. Acute myocardial injury (OR 2.39, 95% CI 1.31 to 4.40, p=0.005) and history of hypertension (OR 1.88, 95% CI 1.01 to 3.55, p=0.049) approximately doubled the odds of in-hospital mortality in patients admitted with COVID-19 after other variables had been controlled for. CONCLUSION Hypertension, pre-existing CVD and acute myocardial injury were associated with increased in-hospital mortality in our cohort of COVID-19 patients. However, only a low number of patients required invasive cardiac intervention.
Collapse
Affiliation(s)
| | - Poornima Mohan
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Richard Jabbour
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | - Gemma Bowsher
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | - Farhan Huq
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | | | - Amarjit Sethi
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Sayan Sen
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Ricardo Petraco
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Neil Ruparelia
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | - Iqbal Malik
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Rodney Foale
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Michael Bellamy
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Jaspal Kooner
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Bushra Rana
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Graham Cole
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Nilesh Sutaria
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | | | - Kevin Fox
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Carla Plymen
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Punam Pabari
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Luke Howard
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Rachel Davies
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Gulammehdi Haji
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | | | - Jon Anderson
- Cardiothoracic Surgery, Imperial College Healthcare NHS Trust, London, UK
| | - Andrew Chukwuemeka
- Cardiothoracic Surgery, Imperial College Healthcare NHS Trust, London, UK
| | - Ramzi Khamis
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Amanda Varnava
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | | | | | - Perviz Asaria
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Rasha Al-Lamee
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - Ghada W Mikhail
- Cardiology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
384
|
Verhoef PA, Kannan S, Sturgill JL, Tucker EW, Morris PE, Miller AC, Sexton TR, Koyner JL, Hejal R, Brakenridge SC, Moldawer LL, Hotchkiss RS, Blood TM, Mazer MB, Bolesta S, Alexander SA, Armaignac DL, Shein SL, Jones C, Hoemann CD, Doctor A, Friess SH, Parker RI, Rotta AT, Remy KE. Severe Acute Respiratory Syndrome-Associated Coronavirus 2 Infection and Organ Dysfunction in the ICU: Opportunities for Translational Research. Crit Care Explor 2021; 3:e0374. [PMID: 33786450 PMCID: PMC7994036 DOI: 10.1097/cce.0000000000000374] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Since the beginning of the coronavirus disease 2019 pandemic, hundreds of thousands of patients have been treated in ICUs across the globe. The severe acute respiratory syndrome-associated coronavirus 2 virus enters cells via the angiotensin-converting enzyme 2 receptor and activates several distinct inflammatory pathways, resulting in hematologic abnormalities and dysfunction in respiratory, cardiac, gastrointestinal renal, endocrine, dermatologic, and neurologic systems. This review summarizes the current state of research in coronavirus disease 2019 pathophysiology within the context of potential organ-based disease mechanisms and opportunities for translational research. DATA SOURCES Investigators from the Research Section of the Society of Critical Care Medicine were selected based on expertise in specific organ systems and research focus. Data were obtained from searches conducted in Medline via the PubMed portal, Directory of Open Access Journals, Excerpta Medica database, Latin American and Caribbean Health Sciences Literature, and Web of Science from an initial search from December 2019 to October 15, 2020, with a revised search to February 3, 2021. The medRxiv, Research Square, and clinical trial registries preprint servers also were searched to limit publication bias. STUDY SELECTION Content experts selected studies that included mechanism-based relevance to the severe acute respiratory syndrome-associated coronavirus 2 virus or coronavirus disease 2019 disease. DATA EXTRACTION Not applicable. DATA SYNTHESIS Not applicable. CONCLUSIONS Efforts to improve the care of critically ill coronavirus disease 2019 patients should be centered on understanding how severe acute respiratory syndrome-associated coronavirus 2 infection affects organ function. This review articulates specific targets for further research.
Collapse
Affiliation(s)
- Philip A Verhoef
- Department of Medicine, University of Hawaii-Manoa, Honolulu, HI
- Kaiser Permanente Hawaii, Honolulu, HI
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jamie L Sturgill
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky, Lexington, KY
| | - Elizabeth W Tucker
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Peter E Morris
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky, Lexington, KY
| | - Andrew C Miller
- Department of Emergency Medicine, Nazareth Hospital, Philadelphia, PA
| | - Travis R Sexton
- Department of Internal Medicine, The University of Kentucky-Lexington School of Medicine, The Gill Heart and Vascular Institute, Lexington, KY
| | - Jay L Koyner
- Section of Nephrology, University of Chicago, Chicago, IL
| | - Rana Hejal
- Department of Internal Medicine, Division of Pulmonary Critical Care, Case Western School of Medicine, Cleveland, OH
| | - Scott C Brakenridge
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, FL
| | - Lyle L Moldawer
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, FL
| | - Richard S Hotchkiss
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Surgery, St. Louis, Washington University School of Medicine, MO
| | - Teresa M Blood
- Department of Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
| | - Monty B Mazer
- Department of Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
| | - Scott Bolesta
- Department of Pharmacy Practice, Nesbitt School of Pharmacy, Wilkes University, Wilkes-Barre, PA
| | | | | | - Steven L Shein
- Department of Pediatrics, Division of Critical Care, Rainbow Babies and Children's Hospital, Cleveland, OH
| | - Christopher Jones
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Allan Doctor
- Department of Pediatrics, Division of Critical Care Medicine, The University of Maryland School of Medicine, Baltimore, MD
| | - Stuart H Friess
- Department of Pediatrics, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
| | - Robert I Parker
- Department of Pediatrics, Hematology Hematology/Oncology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY
| | - Alexandre T Rotta
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Duke University Medical Center, Durham, NC
| | - Kenneth E Remy
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pediatrics, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
385
|
Spijkerman R, Bongers SH, Bindels BJJ, Tinnevelt GH, Giustarini G, Jorritsma NKN, Buitenwerf W, van Spengler DEJ, Delemarre EM, Nierkens S, van Goor HMR, Jansen JJ, Vrisekoop N, Hietbrink F, Leenen LPH, Kaasjager KAH, Koenderman L. Flow cytometric evaluation of the neutrophil compartment in COVID-19 at hospital presentation: A normal response to an abnormal situation. J Leukoc Biol 2021; 109:99-114. [PMID: 33617030 DOI: 10.1002/jlb.5cova0820-520rrr] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly emerging pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Critical COVID-19 is thought to be associated with a hyper-inflammatory process that can develop into acute respiratory distress syndrome, a critical disease normally mediated by dysfunctional neutrophils. This study tested the hypothesis whether the neutrophil compartment displays characteristics of hyperinflammation in COVID-19 patients. Therefore, a prospective study was performed on all patients with suspected COVID-19 presenting at the emergency room of a large academic hospital. Blood drawn within 2 d after hospital presentation was analyzed by point-of-care automated flow cytometry and compared with blood samples collected at later time points. COVID-19 patients did not exhibit neutrophilia or eosinopenia. Unexpectedly neutrophil activation markers (CD11b, CD16, CD10, and CD62L) did not differ between COVID-19-positive patients and COVID-19-negative patients diagnosed with other bacterial/viral infections, or between COVID-19 severity groups. In all patients, a decrease was found in the neutrophil maturation markers indicating an inflammation-induced left shift of the neutrophil compartment. In COVID-19 this was associated with disease severity.
Collapse
Affiliation(s)
- Roy Spijkerman
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Department of Trauma Surgery, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Suzanne H Bongers
- Department of Trauma Surgery, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Bas J J Bindels
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Gerjen H Tinnevelt
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg, Nijmegen, The Netherlands
| | - Giulio Giustarini
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Nikita K N Jorritsma
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Wiebe Buitenwerf
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Daan E J van Spengler
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Eveline M Delemarre
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Harriët M R van Goor
- Department of Internal Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Jeroen J Jansen
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg, Nijmegen, The Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Falco Hietbrink
- Department of Trauma Surgery, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Luke P H Leenen
- Department of Trauma Surgery, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Karin A H Kaasjager
- Department of Internal Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | -
- Department of Respiratory Medicine, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| |
Collapse
|
386
|
Flam B, Wintzell V, Ludvigsson JF, Mårtensson J, Pasternak B. Direct oral anticoagulant use and risk of severe COVID-19. J Intern Med 2021; 289:411-419. [PMID: 33258156 PMCID: PMC7753564 DOI: 10.1111/joim.13205] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hypercoagulability and thromboembolism are prominent features of severe COVID-19, and ongoing anticoagulant use might be protective. METHODS We conducted a nationwide register-based cohort study in Sweden, February through May, 2020, to assess whether ongoing direct oral anticoagulant (DOAC) use was associated with reduced risk of hospital admission for laboratory-confirmed COVID-19, or a composite of intensive care unit (ICU) admission or death due to laboratory-confirmed COVID-19. RESULTS DOAC use (n = 103 703) was not associated with reduced risk of hospital admission for COVID-19 (adjusted hazard ratio [aHR] [95% confidence interval] 1.00 [0.75-1.33] vs. nonuse atrial fibrillation comparator [n = 36 875]; and aHR 0.94 [0.80-1.10] vs. nonuse cardiovascular disease comparator [n = 355 699]), or ICU admission or death due to COVID-19 (aHRs 0.76 [0.51-1.12], and 0.90 [0.71-1.15], respectively). CONCLUSION Ongoing DOAC use was not associated with reduced risk of severe COVID-19, indicating that prognosis would not be modified by early outpatient DOAC initiation.
Collapse
Affiliation(s)
- B Flam
- From the, Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.,Section of Anaesthesiology and Intensive Care Medicine, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - V Wintzell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - J F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Paediatrics, Örebro University Hospital, Örebro, Sweden.,Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - J Mårtensson
- From the, Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.,Section of Anaesthesiology and Intensive Care Medicine, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - B Pasternak
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
387
|
Kintscher U, Wenzel U. ACE2, SARS-CoV-2 und RAAS-Blocker. CARDIOVASC 2021. [PMCID: PMC7966906 DOI: 10.1007/s15027-021-3471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
388
|
Tomasoni D, Petrie MC, Adamo M, Metra M. Renin-angiotensin-aldosterone inhibitors and COVID-19: nearing the end of a media-fuelled controversy. Eur J Heart Fail 2021; 23:486-488. [PMID: 33421242 PMCID: PMC8013611 DOI: 10.1002/ejhf.2098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 02/05/2023] Open
Affiliation(s)
- Daniela Tomasoni
- Cardiology and Cardiac Catheterization Laboratory, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Mark C. Petrie
- British Heart Foundation Institute of Cardiovascular and Medical SciencesUniversity of GlasgowGlasgowUK
| | - Marianna Adamo
- Cardiology and Cardiac Catheterization Laboratory, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Marco Metra
- Cardiology and Cardiac Catheterization Laboratory, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| |
Collapse
|
389
|
Arterielle Hypertonie – Was war 2020 wichtig? DER KARDIOLOGE 2021. [PMCID: PMC7943936 DOI: 10.1007/s12181-021-00470-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Die arterielle Hypertonie bleibt weltweit der prävalenteste Risikofaktor für kardiovaskuläre Erkrankungen und damit einhergehende Behinderungen. Auch im Bereich der arteriellen Hypertonie bestimmte die COVID-19-Pandemie einen Teil der wissenschaftlichen Debatte. Die arterielle Hypertonie ist mit einem schwereren Krankheitsverlauf von COVID-19 assoziiert, wohingegen das SARS-CoV-2-Infektionsrisiko bei Hypertonikern nicht erhöht zu sein scheint. Nach aktueller Datenlage ist die Therapie mit ACE(Angiotensin-Converting-Enzym)-Hemmer und Angiotensin-Typ-1-Rezeptorblocker weder mit einem erhöhten SARS-CoV-2-Infektionsrisiko noch mit einem schwereren Krankheitsverlauf von COVID-19 verbunden. Eine Studie zur antihypertensiven Chronotherapie bestimmte den wissenschaftlichen Diskurs zur medikamentösen Therapie der Hypertonie. Die HYGIA-Studie kam zu der Schlussfolgerung, dass eine abendliche Medikamenteneinnahme das kardiovaskuläre Risiko von Patienten*innen mit arterieller Hypertonie reduziert. Aufgrund einiger Limitationen ist die Datenlage aktuell unzureichend, um eine routinemäßige nächtliche Gabe von antihypertensiven Medikamenten zu empfehlen. Wir werden einige der Aspekte diskutieren. Ein weiterer wissenschaftlicher Schwerpunkt lag auf den neuen Studien zur renalen Denervation. Hier kann man zusammenfassen, dass nach neuer Studienlage die katheterbasierte renale Denervation ein effektives und sicheres Verfahren zur Behandlung der arteriellen Hypertonie darstellt, das sich als alternatives Verfahren zur medikamentösen Blutdrucksenkung weiter etablieren könnte.
Collapse
|
390
|
Savarese G, Benson L, Sundström J, Lund LH. Association between renin-angiotensin-aldosterone system inhibitor use and COVID-19 hospitalization and death: a 1.4 million patient nationwide registry analysis. Eur J Heart Fail 2021; 23:476-485. [PMID: 33222412 PMCID: PMC7753665 DOI: 10.1002/ejhf.2060] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/29/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
AIMS Renin-angiotensin-aldosterone system inhibitors (RAASi) improve outcomes in cardiorenal disease but concerns have been raised over increased risk of incident hospitalization and death from coronavirus disease 2019 (COVID-19). We investigated the association between use of angiotensin-converting enzyme inhibitors (ACEi), angiotensin receptor blockers (ARBs) or mineralocorticoid receptor antagonists (MRAs) and COVID-19 hospitalization/death in a large nationwide population. METHODS AND RESULTS Patients with hypertension, heart failure, diabetes, kidney disease, or ischaemic heart disease registered in the Swedish National Patient Registry until 1 February 2020 were included and followed until 31 May 2020. COVID-19 cases were defined based on hospitalization/death for COVID-19. Multivariable logistic and Cox regressions were fitted to investigate the association between ACEi/ARB and MRA and risk of hospitalization/death for COVID-19 in the overall population, and of all-cause mortality in COVID-19 cases. We performed consistency analysis to quantify the impact of potential unmeasured confounding. Of 1 387 746 patients (60% receiving ACEi/ARB and 5.8% MRA), 7146 (0.51%) had incident hospitalization/death from COVID-19. After adjustment for 45 variables, ACEi/ARB use was associated with a reduced risk of hospitalization/death for COVID-19 (odds ratio 0.86, 95% confidence interval 0.81-0.91) in the overall population, and with reduced mortality in COVID-19 cases (hazard ratio 0.89, 95% confidence interval 0.82-0.96). MRA use was not associated with risk of any outcome. Consistency analysis showed that unmeasured confounding would need to be large for there to be harmful signals associated with RAASi use. CONCLUSIONS In a 1.4 million nationwide cohort, use of RAASi was not associated with increased risk of hospitalization for or death from COVID-19.
Collapse
Affiliation(s)
- Gianluigi Savarese
- Division of Cardiology, Department of MedicineKarolinska InstitutetStockholmSweden
- Heart and Vascular ThemeKarolinska University HospitalStockholmSweden
| | - Lina Benson
- Division of Cardiology, Department of MedicineKarolinska InstitutetStockholmSweden
| | - Johan Sundström
- Department of Medical SciencesClinical Epidemiology Uppsala UniversitetUppsalaSweden
| | - Lars H. Lund
- Division of Cardiology, Department of MedicineKarolinska InstitutetStockholmSweden
- Heart and Vascular ThemeKarolinska University HospitalStockholmSweden
| |
Collapse
|
391
|
Baral R, Tsampasian V, Debski M, Moran B, Garg P, Clark A, Vassiliou VS. Association Between Renin-Angiotensin-Aldosterone System Inhibitors and Clinical Outcomes in Patients With COVID-19: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e213594. [PMID: 33787911 PMCID: PMC8013817 DOI: 10.1001/jamanetworkopen.2021.3594] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE The chronic receipt of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) has been assumed to exacerbate complications associated with COVID-19 and produce worse clinical outcomes. OBJECTIVE To conduct an updated and comprehensive systematic review and meta-analysis comparing mortality and severe adverse events (AEs) associated with receipt vs nonreceipt of ACEIs or ARBs among patients with COVID-19. DATA SOURCES PubMed and Embase databases were systematically searched from December 31, 2019, until September 1, 2020. STUDY SELECTION The meta-analysis included any study design, with the exception of narrative reviews or opinion-based articles, in which COVID-19 was diagnosed through laboratory or radiological test results and in which clinical outcomes (unadjusted or adjusted) associated with COVID-19 were assessed among adult patients (≥18 years) receiving ACEIs or ARBs. DATA EXTRACTION AND SYNTHESIS Three authors independently extracted data on mortality and severe AEs associated with COVID-19. Severe AEs were defined as intensive care unit admission or the need for assisted ventilation. For each outcome, a random-effects model was used to compare the odds ratio (OR) between patients receiving ACEIs or ARBs vs those not receiving ACEIs or ARBs. MAIN OUTCOMES AND MEASURES Unadjusted and adjusted ORs for mortality and severe AEs associated with COVID-19. RESULTS A total of 1788 records from the PubMed and Embase databases were identified; after removal of duplicates, 1664 records were screened, and 71 articles underwent full-text evaluation. Clinical data were pooled from 52 eligible studies (40 cohort studies, 6 case series, 4 case-control studies, 1 randomized clinical trial, and 1 cross-sectional study) enrolling 101 949 total patients, of whom 26 545 (26.0%) were receiving ACEIs or ARBs. When adjusted for covariates, significant reductions in the risk of death (adjusted OR [aOR], 0.57; 95% CI, 0.43-0.76; P < .001) and severe AEs (aOR, 0.68; 95% CI, 0.53-0.88; P < .001) were found. Unadjusted and adjusted analyses of a subgroup of patients with hypertension indicated decreases in the risk of death (unadjusted OR, 0.66 [95% CI, 0.49-0.91]; P = .01; aOR, 0.51 [95% CI, 0.32-0.84]; P = .008) and severe AEs (unadjusted OR, 0.70 [95% CI, 0.54-0.91]; P = .007; aOR, 0.55 [95% CI, 0.36-0.85]; P = .007). CONCLUSIONS AND RELEVANCE In this systematic review and meta-analysis, receipt of ACEIs or ARBs was not associated with a higher risk of multivariable-adjusted mortality and severe AEs among patients with COVID-19 who had either hypertension or multiple comorbidities, supporting the recommendations of medical societies. On the contrary, ACEIs and ARBs may be associated with protective benefits, particularly among patients with hypertension. Future randomized clinical trials are warranted to establish causality.
Collapse
Affiliation(s)
- Ranu Baral
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Department of Cardiology, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Vasiliki Tsampasian
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Maciej Debski
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Brendan Moran
- National Health Service 111 COVID-19 Clinical Assessment Service, Bicester, United Kingdom
- Neasden Medical Centre, London, United Kingdom
- Healix International, Esher, United Kingdom
| | - Pankaj Garg
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Department of Cardiology, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Allan Clark
- Department of Medical Statistics, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Vassilios S. Vassiliou
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Department of Cardiology, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
392
|
Ng TSB, Leblanc K, Yeung DF, Tsang TSM. Médicaments utilisés durant la COVID-19. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2021; 67:e69-e78. [PMID: 33727386 PMCID: PMC7963008 DOI: 10.46747/cfp.6703e69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Objectif En réponse à la pandémie actuelle de maladie à coronavirus 2019 (COVID-19), garder les médecins au fait des médicaments qui ont été proposés pour combattre la maladie, et des données probantes à l’appui de leur utilisation. Sources d’information Une revue narrative des médicaments les plus fréquemment utilisés pour combattre la COVID-19 a été réalisée, afin de souligner les meilleures données probantes disponibles concernant chaque traitement pharmacologique jusqu’ici. Des recherches ont été effectuées sur PubMed, EMBASE et MEDLINE à l’aide des mots-clés anglais COVID-19 et treatment , ainsi que d’autres mots-clés connexes. Ont été inclus les études pertinentes menées auprès de populations humaines et des cas de patients atteints de la COVID-19, ainsi que les articles et revues relevés à la main. Seuls les articles rédigés en anglais et en chinois ont été retenus. Message principal Alors que la prise en charge actuelle des patients atteints de la COVID-19 consiste principalement en soins de soutien, sans accès aux vaccins, les praticiens se sont tournés vers des médicaments utilisés dans d’autres indications. Cela a causé une grande controverse, puisque des données cliniques limitées étayaient l’utilisation de beaucoup de ces traitements, et cela pouvait se répercuter sur la sécurité du patient, l’accès aux médicaments et la santé publique. Par exemple, les médicaments tels que l’hydroxychloroquine et la chloroquine, le lopinavir-ritonavir, les anti-inflammatoires non stéroïdiens, les inhibiteurs de l’enzyme de conversion de l’angiotensine et les antagonistes des récepteurs de l’angiotensine ont capté l’attention des médias en raison de la médiatisation, de la mésinformation ou de la mauvaise interprétation des données de recherche. Conclusion Vu la gravité de la pandémie et les vastes effets éventuels de l’adoption de traitements sûrs et efficaces, cet article se veut être une revue narrative des données probantes actuelles étayant les médicaments les plus utilisés pour le traitement de la COVID-19 afin de permettre aux professionnels de la santé de prendre des décisions éclairées en matière de soins pour les patients qui sont atteints de cette maladie potentiellement mortelle.
Collapse
Affiliation(s)
- T S Brandon Ng
- Étudiant en médecine à la faculté de médecine de l'Université de la Colombie-Britannique à Vancouver
| | - Kori Leblanc
- Professeure adjointe à la faculté de pharmacie Leslie Dan de l'Université de Toronto en Ontario
| | - Darwin F Yeung
- Professeur clinique adjoint à la faculté de médecine de l'Université de la Colombie-Britannique
| | - Teresa S M Tsang
- Directrice de l'échocardiographie à l'Hôpital Vancouver General Hospital et UBC Hospital, et elle est professeure de médecine à la division de cardiologie et directrice associée de recherche au département de médecine de l'Université de la Colombie-Britannique.
| |
Collapse
|
393
|
Price S, Targoński R, Sadowski J, Targoński R. To Curb the Progression of Fatal COVID-19 Course-Dream or Reality. Curr Hypertens Rep 2021; 23:12. [PMID: 33638064 PMCID: PMC7910199 DOI: 10.1007/s11906-021-01130-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To analyze the impact of sodium retention states on the course of COVID-19 and propose possible interventions to curb disease progression. RECENT FINDINGS Numerous data confirm a positive association of non-communicable diseases, aging, and other sodium-retaining states, including iatrogenic ones, with more severe sometimes fatal clinical course of COVID-19. Reasons for this effect could include increased angiotensin signaling via the AT1R receptor. The endothelial glycocalyx also plays an important role in infection, leading to a vicious cycle of inflammation and tissue sodium retention when damaged. RAS inhibitors may help restore glycocalyx function and prevent severe organ damage. Anticoagulants, especially heparin, may also have therapeutic applications due to antithrombotic, anti-inflammatory, glycocalyx-repairing, and antialdosteronic properties. The ambiguous influence of some diuretics on sodium balance was also discussed. Abnormal sodium storage and increased angiotensin-converting enzyme activity are related to the severity of COVID-19. Inducing sodium removal and reducing intake might improve outcomes.
Collapse
Affiliation(s)
- Szymon Price
- Miejski Szpital Zespolony w Olsztynie, Klinika Kardiologii i Chorób Wewnętrznych, Clinic of Cardiology and Internal Medicine, Metropolitan Hospital of Nicolaus Copernicus, University of Warmia and Mazury, Niepodległości 44, 10-045 Olsztyn, Poland
| | - Radosław Targoński
- Department of Cardiac & Vascular Surgery, University Hospital of Gdańsk, M. Skłodowskiej-Curie 3a street, 80-210 Gdańsk, Poland
| | - Janusz Sadowski
- Miejski Szpital Zespolony w Olsztynie, Klinika Kardiologii i Chorób Wewnętrznych, Clinic of Cardiology and Internal Medicine, Metropolitan Hospital of Nicolaus Copernicus, University of Warmia and Mazury, Niepodległości 44, 10-045 Olsztyn, Poland
| | - Ryszard Targoński
- Department of Cardiac & Vascular Surgery, University Hospital of Gdańsk, M. Skłodowskiej-Curie 3a street, 80-210 Gdańsk, Poland
| |
Collapse
|
394
|
Khera R, Clark C, Lu Y, Guo Y, Ren S, Truax B, Spatz ES, Murugiah K, Lin Z, Omer SB, Vojta D, Krumholz HM. Association of Angiotensin-Converting Enzyme Inhibitors and Angiotensin Receptor Blockers With the Risk of Hospitalization and Death in Hypertensive Patients With COVID-19. J Am Heart Assoc 2021; 10:e018086. [PMID: 33624516 PMCID: PMC8403305 DOI: 10.1161/jaha.120.018086] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Despite its clinical significance, the risk of severe infection requiring hospitalization among outpatients with severe acute respiratory syndrome coronavirus 2 infection who receive angiotensin‐converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) remains uncertain. Methods and Results In a propensity score–matched outpatient cohort (January–May 2020) of 2263 Medicare Advantage and commercially insured individuals with hypertension and a positive outpatient SARS‐CoV‐2, we determined the association of ACE inhibitors and ARBs with COVID‐19 hospitalization. In a concurrent inpatient cohort of 7933 hospitalized with COVID‐19, we tested their association with in‐hospital mortality. The robustness of the observations was assessed in a contemporary cohort (May–August). In the outpatient study, neither ACE inhibitors (hazard ratio [HR], 0.77; 0.53–1.13, P=0.18) nor ARBs (HR, 0.88; 0.61–1.26, P=0.48) were associated with hospitalization risk. ACE inhibitors were associated with lower hospitalization risk in the older Medicare group (HR, 0.61; 0.41–0.93, P=0.02), but not the younger commercially insured group (HR, 2.14; 0.82–5.60, P=0.12; P‐interaction 0.09). Neither ACE inhibitors nor ARBs were associated with lower hospitalization risk in either population in the validation cohort. In the primary inpatient study cohort, neither ACE inhibitors (HR, 0.97; 0.81–1.16; P=0.74) nor ARBs (HR, 1.15; 0.95–1.38, P=0.15) were associated with in‐hospital mortality. These observations were consistent in the validation cohort. Conclusions ACE inhibitors and ARBs were not associated with COVID‐19 hospitalization or mortality. Despite early evidence for a potential association between ACE inhibitors and severe COVID‐19 prevention in older individuals, the inconsistency of this observation in recent data argues against a role for prophylaxis.
Collapse
Affiliation(s)
- Rohan Khera
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT
| | - Callahan Clark
- Research & Development at UnitedHealth Group Minnetonka MN UAS
| | - Yuan Lu
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT
| | - Yinglong Guo
- Research & Development at UnitedHealth Group Minnetonka MN UAS
| | - Sheng Ren
- Research & Development at UnitedHealth Group Minnetonka MN UAS
| | - Brandon Truax
- Research & Development at UnitedHealth Group Minnetonka MN UAS
| | - Erica S Spatz
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT
| | - Karthik Murugiah
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT
| | - Zhenqiu Lin
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT
| | - Saad B Omer
- Section of Infectious Diseases Department of Internal Medicine Yale School of Medicine New Haven CT.,Yale Institute for Global Health New Haven CT.,Department of Epidemiology of Microbial Diseases Yale School of Public Health New Haven CT
| | - Deneen Vojta
- Research & Development at UnitedHealth Group Minnetonka MN UAS
| | - Harlan M Krumholz
- Section of Cardiovascular Medicine Department of Internal Medicine Yale School of Medicine New Haven CT.,Center for Outcomes Research and Evaluation Yale-New Haven Hospital New Haven CT.,Department of Health Policy and Management Yale School of Public Health New Haven CT
| |
Collapse
|
395
|
Collard D, Nurmohamed NS, Kaiser Y, Reeskamp LF, Dormans T, Moeniralam H, Simsek S, Douma R, Eerens A, Reidinga AC, Elbers PWG, Beudel M, Vogt L, Stroes ESG, van den Born BJH. Cardiovascular risk factors and COVID-19 outcomes in hospitalised patients: a prospective cohort study. BMJ Open 2021; 11:e045482. [PMID: 33619201 PMCID: PMC7902321 DOI: 10.1136/bmjopen-2020-045482] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Recent reports suggest a high prevalence of hypertension and diabetes in COVID-19 patients, but the role of cardiovascular disease (CVD) risk factors in the clinical course of COVID-19 is unknown. We evaluated the time-to-event relationship between hypertension, dyslipidaemia, diabetes and COVID-19 outcomes. DESIGN We analysed data from the prospective Dutch CovidPredict cohort, an ongoing prospective study of patients admitted for COVID-19 infection. SETTING Patients from eight participating hospitals, including two university hospitals from the CovidPredict cohort were included. PARTICIPANTS Admitted, adult patients with a positive COVID-19 PCR or high suspicion based on CT-imaging of the thorax. Patients were followed for major outcomes during the hospitalisation. CVD risk factors were established via home medication lists and divided in antihypertensives, lipid-lowering therapy and antidiabetics. PRIMARY AND SECONDARY OUTCOMES MEASURES The primary outcome was mortality during the first 21 days following admission, secondary outcomes consisted of intensive care unit (ICU) admission and ICU mortality. Kaplan-Meier and Cox regression analyses were used to determine the association with CVD risk factors. RESULTS We included 1604 patients with a mean age of 66±15 of whom 60.5% were men. Antihypertensives, lipid-lowering therapy and antidiabetics were used by 45%, 34.7% and 22.1% of patients. After 21-days of follow-up; 19.2% of the patients had died or were discharged for palliative care. Cox regression analysis after adjustment for age and sex showed that the presence of ≥2 risk factors was associated with increased mortality risk (HR 1.52, 95% CI 1.15 to 2.02), but not with ICU admission. Moreover, the use of ≥2 antidiabetics and ≥2 antihypertensives was associated with mortality independent of age and sex with HRs of, respectively, 2.09 (95% CI 1.55 to 2.80) and 1.46 (95% CI 1.11 to 1.91). CONCLUSIONS The accumulation of hypertension, dyslipidaemia and diabetes leads to a stepwise increased risk for short-term mortality in hospitalised COVID-19 patients independent of age and sex. Further studies investigating how these risk factors disproportionately affect COVID-19 patients are warranted.
Collapse
Affiliation(s)
- Didier Collard
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Laurens F Reeskamp
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom Dormans
- Department of Intensive Care, Zuyderland Medical Centre Sittard-Geleen, Sittard-Geleen, The Netherlands
| | - Hazra Moeniralam
- Internal Medicine, Sint Antonius Hospital, Nieuwegein, The Netherlands
| | - Suat Simsek
- Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Renee Douma
- Internal Medicine, Flevoziekenhuis, Almere, Flevoland, The Netherlands
| | - Annet Eerens
- Oncology, Treant Healthcare Group, Amsterdam, The Netherlands
| | - Auke C Reidinga
- Intensive Care, Martini Ziekenhuis, Groningen, Groningen, The Netherlands
| | - Paul W G Elbers
- Department of Intensive Care, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martijn Beudel
- Department of Neurology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Nephrology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
396
|
Ajmera V, Thompson WK, Smith DM, Malhotra A, Mehta RL, Tolia V, Yin J, Sriram K, Insel PA, Collier S, Richards L, Loomba R. RAMIC: Design of a randomized, double-blind, placebo-controlled trial to evaluate the efficacy of ramipril in patients with COVID-19. Contemp Clin Trials 2021; 103:106330. [PMID: 33631357 PMCID: PMC7899027 DOI: 10.1016/j.cct.2021.106330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIMS Retrospective studies have shown that angiotensin-converting-enzyme (ACE) inhibitors are associated with a reduced risk of complications and mortality in persons with novel coronavirus disease 2019 (COVID-19). Thus, we aimed to examine the efficacy of ramipril, an ACE-inhibitor, in preventing ICU admission, mechanical ventilation and/or mortality while also minimizing the risk of transmission and use of personal protective equipment (PPE). METHODS RAMIC is a multicenter, randomized, double-blind, allocation-concealed, placebo-controlled trial comparing the efficacy of treatment with ramipril 2.5 mg orally daily compared to placebo for 14 days. The study population includes adult patients with COVID-19 who were admitted to a hospital or assessed in an emergency department or ambulatory clinic. Key exclusion criteria include ICU admission or need for mechanical ventilation at screening, use of an ACE inhibitor or angiotensin-receptor-II blocker within 7 days, glomerular filtration rate < 40 mL/min or a systolic blood pressure (BP) < 100 mmHg or diastolic BP < 65 mmHg. Patients are randomized 2:1 to receive ramipril (2.5 mg) or placebo daily. Informed consent and study visits occur virtually to minimize the risk of SARS-CoV-2 transmission and preserve PPE. The primary composite endpoint of ICU admission, invasive mechanical ventilation and death are adjudicated virtually. CONCLUSIONS RAMIC is designed to assess the efficacy of treatment with ramipril for 14 days to decrease ICU admission, mechanical ventilator use and mortality in patients with COVID-19 and leverages virtual study visits and endpoint adjudication to mitigate risk of infection and to preserve PPE (ClinicalTrials.gov, NCT04366050).
Collapse
Affiliation(s)
- Veeral Ajmera
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, United States of America; NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Wesley K Thompson
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, United States of America
| | - Davey M Smith
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Atul Malhotra
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ravindra L Mehta
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Vaishal Tolia
- Department of Emergency Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jeffrey Yin
- Department of Pharmacy, University of California San Diego, La Jolla, California, United States of America
| | - Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
| | - Paul A Insel
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America; Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, California, United States of America
| | - Summer Collier
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, United States of America; NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, California, United States of America.
| |
Collapse
|
397
|
Gressens SB, Leftheriotis G, Dussaule JC, Flamant M, Levy BI, Vidal-Petiot E. Controversial Roles of the Renin Angiotensin System and Its Modulators During the COVID-19 Pandemic. Front Physiol 2021; 12:624052. [PMID: 33692701 PMCID: PMC7937723 DOI: 10.3389/fphys.2021.624052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
Since December 2019, the coronavirus 2019 (COVID-19) pandemic has rapidly spread and overwhelmed healthcare systems worldwide, urging physicians to understand how to manage this novel infection. Early in the pandemic, more severe forms of COVID-19 have been observed in patients with cardiovascular comorbidities, who are often treated with renin-angiotensin aldosterone system (RAAS)-blockers, such as angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), but whether these are indeed independent risk factors is unknown. The cellular receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the membrane-bound angiotensin converting enzyme 2 (ACE2), as for SARS-CoV(-1). Experimental data suggest that expression of ACE2 may be increased by RAAS-blockers, raising concerns that these drugs may facilitate viral cell entry. On the other hand, ACE2 is a key counter-regulator of the RAAS, by degrading angiotensin II into angiotensin (1-7), and may thereby mediate beneficial effects in COVID-19. These considerations have raised concerns about the management of these drugs, and early comments shed vivid controversy among physicians. This review will describe the homeostatic balance between ACE-angiotensin II and ACE2-angiotensin (1-7) and summarize the pathophysiological rationale underlying the debated role of the RAAS and its modulators in the context of the pandemic. In addition, we will review available evidence investigating the impact of RAAS blockers on the course and prognosis of COVID-19 and discuss why retrospective observational studies should be interpreted with caution. These considerations highlight the importance of solid evidence-based data in order to guide physicians in the management of RAAS-interfering drugs in the general population as well as in patients with more or less severe forms of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Simon B Gressens
- Department of Infectious and Tropical Diseases, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France
| | - Georges Leftheriotis
- Laboratory of Molecular Physiology and Medicine, Université Cote d'Azur, Nice, France
| | - Jean-Claude Dussaule
- Sorbonne Université, INSERM, Unité des Maladies Rénales Fréquentes et Rares: des Mécanismes Moléculaires à la Médecine Personnalisée, AP-HP, Hôpital Tenon, Paris, France.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Martin Flamant
- Department of Physiology, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France.,Inserm U1149, Centre for Research on Inflammation, Université de Paris, Paris, France
| | | | - Emmanuelle Vidal-Petiot
- Department of Physiology, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France.,Inserm U1149, Centre for Research on Inflammation, Université de Paris, Paris, France
| |
Collapse
|
398
|
Experimental data using candesartan and captopril indicate no double-edged sword effect in COVID-19. Clin Sci (Lond) 2021; 135:465-481. [PMID: 33479758 PMCID: PMC7851407 DOI: 10.1042/cs20201511] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
The key link between renin–angiotensin system (RAS) and COVID-19 is ACE2 (angiotensin-converting enzyme 2), which acts as a double-edged sword, because ACE2 increases the tissue anti-inflammatory response but it is also the entry receptor for the virus. There is an important controversy on several drugs that regulate RAS activity and possibly ACE2, and are widely used, particularly by patients most vulnerable to severe COVID-19. In the lung of healthy rats, we observed that candesartan (an angiotensin type-1, AT1, receptor blocker; ARB) and captopril (an ACE inhibitor; ACEI) up-regulated expression of tissue ACE2 and RAS anti-inflammatory axis receptors (AT2 and Mas receptors). This effect was particularly pronounced in rats with metabolic syndrome (obesity, increased blood pressure and hyperglycemia) and aged rats. Treatment of cultures of human type-II pneumocytes with candesartan or captopril induced up-regulation of ACE2 expression in cells. Treatment with viral spike protein induced a decrease in full-length (i.e. transmembrane) ACE2, an increase in levels of a short intracellular ACE2 polypeptide and an increase in ADAM17 activity in cells, together with an increase in levels of soluble ACE2 and major proinflammatory cytokines in the culture medium. Spike protein-induced changes and levels of spike protein internalization in cells were inhibited by pretreatment with the above-mentioned drugs. The results suggest that these drugs increase ACE2 levels and promote the anti-inflammatory RAS axis in the lung. Furthermore, possible up-regulation of viral entry by the drug-induced increase in expression of transmembrane ACE2 is counteracted by additional mechanisms, particularly by drug-induced inhibition of ADAM17 activity.
Collapse
|
399
|
McKeigue PM, Kennedy S, Weir A, Bishop J, McGurnaghan SJ, McAllister D, Robertson C, Wood R, Lone N, Murray J, Caparrotta TM, Smith-Palmer A, Goldberg D, McMenamin J, Guthrie B, Hutchinson S, Colhoun HM. Relation of severe COVID-19 to polypharmacy and prescribing of psychotropic drugs: the REACT-SCOT case-control study. BMC Med 2021; 19:51. [PMID: 33612113 PMCID: PMC7897516 DOI: 10.1186/s12916-021-01907-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The objective of this study was to investigate the relation of severe COVID-19 to prior drug prescribing. METHODS Severe cases were defined by entry to critical care or fatal outcome. For this matched case-control study (REACT-SCOT), all 4251 cases of severe COVID-19 in Scotland since the start of the epidemic were matched for age, sex and primary care practice to 36,738 controls from the population register. Records were linked to hospital discharges since June 2015 and dispensed prescriptions issued in primary care during the last 240 days. RESULTS Severe COVID-19 was strongly associated with the number of non-cardiovascular drug classes dispensed. This association was strongest in those not resident in a care home, in whom the rate ratio (95% CI) associated with dispensing of 12 or more drug classes versus none was 10.8 (8.8, 13.3), and in those without any of the conditions designated as conferring increased risk of COVID-19. Of 17 drug classes postulated at the start of the epidemic to be "medications compromising COVID", all were associated with increased risk of severe COVID-19 and these associations were present in those without any of the designated risk conditions. The fraction of cases in the population attributable to exposure to these drug classes was 38%. The largest effect was for antipsychotic agents: rate ratio 4.18 (3.42, 5.11). Other drug classes with large effects included proton pump inhibitors (rate ratio 2.20 (1.72, 2.83) for = 2 defined daily doses/day), opioids (3.66 (2.68, 5.01) for = 50 mg morphine equivalent/day) and gabapentinoids. These associations persisted after adjusting for covariates and were stronger with recent than with non-recent exposure. CONCLUSIONS Severe COVID-19 is associated with polypharmacy and with drugs that cause sedation, respiratory depression, or dyskinesia; have anticholinergic effects; or affect the gastrointestinal system. These associations are not easily explained by co-morbidity. Measures to reduce the burden of mortality and morbidity from COVID-19 should include reinforcing existing guidance on reducing overprescribing of these drug classes and limiting inappropriate polypharmacy. REGISTRATION ENCEPP number EUPAS35558.
Collapse
Affiliation(s)
- Paul M McKeigue
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland. .,Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland.
| | - Sharon Kennedy
- NHS Information Services Division (Public Health Scotland), Gyle Square, 1 South Gyle Crescent, Edinburgh, EH12 9EB, Scotland
| | - Amanda Weir
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - Jen Bishop
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - Stuart J McGurnaghan
- Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, EH4 2XUC, Scotland
| | - David McAllister
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland.,Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow, G12 8RZ, Scotland
| | - Chris Robertson
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland.,Department of Mathematics and Statistics, University of Strathclyde, 16 Richmond Street, Glasgow, G1 1XQ, Scotland
| | - Rachael Wood
- NHS Information Services Division (Public Health Scotland), Gyle Square, 1 South Gyle Crescent, Edinburgh, EH12 9EB, Scotland
| | - Nazir Lone
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Janet Murray
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - Thomas M Caparrotta
- Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, EH4 2XUC, Scotland
| | - Alison Smith-Palmer
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - David Goldberg
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - Jim McMenamin
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland
| | - Bruce Guthrie
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Sharon Hutchinson
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland.,School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Helen M Colhoun
- Public Health Scotland, Meridian Court, 5 Cadogan Street, Glasgow, G2 6QE, Scotland.,Institute of Genetics and Molecular Medicine, College of Medicine and Veterinary Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, EH4 2XUC, Scotland
| | | |
Collapse
|
400
|
Hendricks CL, Herd C, Nel M, Tintinger G, Pepper MS. The COVID-19 Treatment Landscape: A South African Perspective on a Race Against Time. Front Med (Lausanne) 2021; 8:604087. [PMID: 33681243 PMCID: PMC7933453 DOI: 10.3389/fmed.2021.604087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
The pandemic caused by SARS-CoV-2 has infected more than 94 million people worldwide (as of 17 January 2020). Severe disease is believed to be secondary to the cytokine release syndrome (CRS or "cytokine storm") which causes local tissue damage as well as multi-organ dysfunction and thrombotic complications. Due to the high mortality rates in patients receiving invasive ventilation, practice has changed from "early-intubation" for acute respiratory distress syndrome (ARDS) to a trial of non-invasive ventilation (NIV) or high flow nasal cannula (HFNC) oxygen. Reports indicating the benefit of NIV and HFNC have been encouraging and have led to more than 20,000 such devices being manufactured and ready for roll-out in South Africa (SA) as of July 2020. The need to identify drugs with clear clinical benefits has led to an array of clinical trials, most of which are repurposing drugs for COVID-19. The treatment landscape reflects the need to target both the virus and its effects such as the CRS and thrombotic complications. Conflicting results have the potential to confuse the implementation of coordinated treatment strategies and guidelines. The purpose of this review is to address pertinent areas in the current literature on the available medical treatment options for COVID-19. Remdesivir, tocilizumab, and dexamethasone are some of the treatment options that have shown the most promise, but further randomized trials are required to particularly address timing and dosages to confidently create standardized protocols. For the SA population, two healthcare sectors exist. In the private sector, patients with medical insurance may have greater access to a wider range of treatment options than those in the public sector. The latter serves >80% of the population, and resource constraints require the identification of drugs with the most cost-effective use for the greatest number of affected patients.
Collapse
Affiliation(s)
- Candice Laverne Hendricks
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Candice Herd
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Marcel Nel
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Gregory Tintinger
- Department of Internal Medicine, University of Pretoria, Pretoria, South Africa
| | - Michael Sean Pepper
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|