1
|
Tawfiq E, Chen R, Honeyman DA, Dawson R, Kunasekaran M, Notaras A, Gurdasani D, Skouteris H, Ayton D, MacIntyre CR. Long Covid Symptom Clusters, Correlates and Predictors in a Highly Vaccinated Australian Population in 2023. Health Expect 2025; 28:e70273. [PMID: 40342248 PMCID: PMC12059467 DOI: 10.1111/hex.70273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Limited data exists regarding long Covid burden following Omicron infection in highly vaccinated populations. OBJECTIVE To (1) characterise long Covid prevalence and predictors and (2) identify key symptom clusters and their correlates among long Covid patients, during an Omicron-predominant period in a highly vaccinated population. DESIGN Anonymous, online, cross-sectional survey. SETTING January 2023, Australia. PARTICIPANTS Residents aged ≥ 18 years with self-reported history of test-positive Covid-19. The main variables studied were socio-demographic characteristics, Covid-19 risk factors, vaccination, infection history and experiences with long Covid. MAIN OUTCOME MEASURES Long Covid symptoms. Symptom-based clustering was used to identify long Covid symptom clusters and their functional correlates. Predictors of long Covid occurrence and severity were assessed using multivariable logistic regression. RESULTS Overall, 240/1205 participants (19.9%) reported long Covid. Long Covid risk was significantly higher for women OR 1.71 (95% CI: 1.17-2.51), people with comorbidities 2.19 (95% CI: 1.56-3.08) and those using steroid inhalers for Covid-19 treatment (2.34 [95% CI: 1.29-4.24]). Long-Covid risk increased with increasing Covid-19 infection severity (moderately severe symptoms: 2.23 [95% CI: 1.50-3.30], extremely severe symptoms: 5.80 [95% CI: 3.48-9.66], presented to ED/hospitalised: 7.22 [95% CI: 3.06-17.03]). We found no significant difference in the likelihood of long Covid between the Omicron and pre-Omicron periods, vaccination status and participant age. We identified two long Covid clusters (pauci-symptomatic, n = 170, vs. polysymptomatic, n = 66). Polysymptomatic cluster membership was associated with worse functioning (impacts on work, moderate activity, emotions and energy). Severity acute infection was strongly predictive of polysymptomatic cluster membership (5.72 [2.04-17.58]). Monoclonal antibody treatment was strongly associated with pauci-symptomatic cluster membership (0.02 [0.00-0.13]). DISCUSSION Our study shows that long Covid is an important health burden in Australia, including during the Omicron era, and identifies several risk factors. We found a subgroup of patients characterised by more symptoms and worse functional outcomes. Our findings can inform policies for protecting vulnerable populations and frameworks for long Covid risk assessment and management. CONCLUSIONS One-in-five people may suffer long Covid after acute Covid-19 infection, with similar risk across age groups. Omicron variants appear not to have a lower risk compared to earlier variants in our study. A cumulative number of symptoms can help triage long Covid patients. PATIENT OR PUBLIC CONTRIBUTION We did not involve patients or the public in the design of the questionnaire. However, after a soft launch, we revised some survey questions by reviewing early responses from patients and the public.
Collapse
Affiliation(s)
- Essa Tawfiq
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Rosalie Chen
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Damian Alexander Honeyman
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Rebecca Dawson
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Mohana Kunasekaran
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Adriana Notaras
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
| | - Deepti Gurdasani
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
- University of Western Australia Medical SchoolUniversity of Western AustraliaPerthAustralia
- William Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Helen Skouteris
- Health and Social Care UnitSchool of Public Health and Preventive Medicine, Monash University in collaboration with Monash HealthMelbourneAustralia
| | - Darshini Ayton
- Health and Social Care UnitSchool of Public Health and Preventive Medicine, Monash University in collaboration with Monash HealthMelbourneAustralia
| | - Chandini Raina MacIntyre
- Biosecurity ProgramThe Kirby Institute, Faculty of Medicine and HealthThe University of New South WalesSydneyAustralia
- College of Public Service & Community Solutions, and College of Health SolutionsArizona State UniversityTempeArizonaUSA
| |
Collapse
|
2
|
Quintana JM, Larrea N, Menéndez L, Legarreta MJ, Gascon M, Garcia-Asensio J, España PP. Effectiveness of drugs employed in the treatment of COVID-19: real-world evidence. Expert Rev Respir Med 2025; 19:493-498. [PMID: 40186558 DOI: 10.1080/17476348.2025.2488966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Drugs used to treat patients with SARS-CoV-2 infection have been tested in different clinical trials but less in real-life studies. Our goal was to assess the effectiveness of several specific COVID-19 drugs. RESEARCH DESIGN AND METHODS Retrospective study of all patients testing positive for SARS-CoV-2 infection between 1 March 2020 and 9 January 2022. We extracted sociodemographic, basal comorbidities, specific medication for their COVID-19, COVID-19 vaccination data, and outcomes such as death and admission to hospital and intensive care unit (ICU) during the different periods of the pandemic. RESULTS The prescription of corticosteroids to out-of-hospital patients was related to a higher likelihood of hospital admission. Among the hospitalized patients, all of the drugs studied (dexamethasone, prednisone, methylprednisolone, remdesivir, ritonavir/lopinavir, and tocilizumab) were related to a higher likelihood of dying at 90 days or being admitted to an ICU. In patients admitted to an ICU, corticosteroids prevented intrahospital mortality (Odds ratio: 0.73; 95% confident intervals: 0.554-0.969). All drugs were related to longer length of hospital admission and ICU stays. CONCLUSIONS Most drugs used for COVID-19 patients had no clear benefit except in the case of corticosteroids, associated with a reduced risk of intrahospital mortality among ICU patients. TRIAL REGISTRATION The study is registered at ClinicalTrials.gov (CT.gov identifier: NCT04463706).
Collapse
Affiliation(s)
- Jose M Quintana
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Research Unit, Galdakao, Spain
- Biosistemak Institute for Health System Research, Barakaldo, Spain
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Bilbao, Spain
| | - Nere Larrea
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Research Unit, Galdakao, Spain
- Biosistemak Institute for Health System Research, Barakaldo, Spain
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Bilbao, Spain
| | - Lara Menéndez
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Pharmacy Service, Galdakao, Spain
| | - Maria J Legarreta
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Research Unit, Galdakao, Spain
- Biosistemak Institute for Health System Research, Barakaldo, Spain
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Bilbao, Spain
| | - Maria Gascon
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Bilbao, Spain
- Bioaraba Health Research Institute, Araba University Hospital, Research Unit, Vitoria-Gasteiz, Spain
| | - Julia Garcia-Asensio
- Basque Government Department of Health, Office of Healthcare Planning, Organization and Evaluation, Basque Country, Spain
| | - Pedro-Pablo España
- Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Respiratory Service, Galdakao, Spain
- BioBizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
3
|
Sheikhian F, Golparvar MM, Ahmadi I, Anvari E, Majdolashrafi F, Ghazanfari Jajin M, Sakhaee F, Sheikhpour M, Fateh A. CTLA-4 rs5742909 but not ADAM33 rs2280091 is a predictor factor for COVID-19 mortality. J Infect Public Health 2025; 18:102618. [PMID: 39673939 DOI: 10.1016/j.jiph.2024.102618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Research has demonstrated the association between susceptibility to coronavirus disease 2019 (COVID-19) and single nucleotide polymorphisms (SNPs). On the other hand, the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) serves as a pivotal inhibitory receptor with a substantial impact on the advancement of viral infections. Besides, the disintegrin and metalloproteinase33 (ADAM33) gene is associated with both asthma and heightened airway responsiveness. Hence, this investigation sought to elucidate the potential association between the CTLA-4 rs5742909 and ADAM33 rs2280091 SNPs and the fatality rate of COVID-19 across various variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS Both SNPs were genotyped with the PCR-RFLP assay in 1734 improved and 1450 deceased individuals. RESULTS Our obtained results revealed a significant association between the CTLA-4 rs5742909 C/T-T/T genotypes and increased mortality risk of COVID-19 in three different SARS-CoV-2 variants. The ADAM33 rs2280091 polymorphism demonstrated no significant association with COVID-19 mortality under various inheritance models. Nevertheless, subsequent adjustments for SARS-CoV-2 variants revealed a notable association between the GA genotype of ADAM33 rs2280091 and mortality rates specifically among individuals infected with the Delta variant. CONCLUSIONS In summary, the prediction of COVID-19 severity could be facilitated through the utilization of the CTLA-4 rs5742909 marker. Conversely, in the case of ADAM33 rs2280091, such prognostication appears to be contingent upon the specific variants of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Farzaneh Sheikhian
- Department of Biology, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | | | - Iraj Ahmadi
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Enayat Anvari
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Fatemeh Majdolashrafi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Sakhaee
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Mohamed Elmenshawy SS, Ahmed Abdelsalam MF, El Nagdy TR, Salam Elgohary MA, Sabri NA, El-kholy A. Evaluation of the efficacy of triazavirin versus oseltamivir in management of COVID-19. Future Sci OA 2024; 10:2418798. [PMID: 39539144 PMCID: PMC11572151 DOI: 10.1080/20565623.2024.2418798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: To compare the safety and efficacy of triazavirin (TZV) 250 mg capsules versus oseltamivir 75 mg capsules in the treatment of individuals diagnosed with COVID-19.Materials & methods: A double-blind, randomized, interventional pilot study was conducted in a parallel design with a 7-day treatment period.Results: The results showed that TZV is superior to oseltamivir regarding length of hospital stay, days until clinical symptoms improvement, and cure of chest pneumonia.Discussion: Different literature data indicate the superiority of TZV in treating COVID-19 when compared with other therapeutic agents. While there is controversy about the efficacy of oseltamivir.Conclusion: It was concluded that TZV is more effective than oseltamivir in the management of COVID-19 disease.Trial registration number: NCT04973462.
Collapse
Affiliation(s)
| | | | | | | | - Nagwa Ali Sabri
- Clinical Pharmacy-Clinical Pharmacy Department-Faculty of Pharmacy-Ain Shams University, Cairo, Egypt
| | - Amal El-kholy
- Clinical Pharmacy-Clinical Pharmacy Department-Faculty of Pharmacy-Ain Shams University, Cairo, Egypt
| |
Collapse
|
5
|
Abe S, Wannigama DL, Suzuki Y, Akaneya D, Igarashi J, Suto M, Moriya K, Ishizawa D, Okuma Y, Hongsing P, Hurst C, Saethang T, Higgins PG, Stick SM, Kicic A. Real world effectiveness of early ensitrelvir treatment in patients with SARS-CoV-2, a retrospective case series. New Microbes New Infect 2024; 62:101522. [PMID: 39552926 PMCID: PMC11567130 DOI: 10.1016/j.nmni.2024.101522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Background Ensitrelvir, a 3C-like protease inhibitor, received emergency approval in Japan in November 2022 for treating non-hospitalized patients with mild-to-moderate COVID-19. However, confirmation of its real-world clinical effectiveness is limited. Methods This retrospective study evaluated 18 vaccinated outpatients (15 men; median age, 39.5 years; range, 26-56), treated with a 5-day oral ensitrelvir regimen (375 mg loading dose, followed by 125 mg daily) between December 1, 2022, and January 31, 2023. Nasal swabs were collected on days 0, 3, 6, and 9 for RT-qPCR to assess viral load. Variants were identified by Sanger sequencing, and outcomes were compared to historical controls. Patients were followed for 60 days to monitor for post-acute sequelae of COVID-19 (PASC). Results Symptoms such as mild fever and sore throat improved rapidly after one day of ensitrelvir treatment, with 66 % of patients recovering within six days. All individuals were infected with the BA.5 Omicron variant. Viral loads, as measured by Ct values, increased significantly from 21.82 at symptom onset to 37.65 b y day 6, with SARS-CoV-2 RNA undetectable in most patients by day 9. Those treated within 48 h of symptom onset showed the viral load reduction. Compared to historical controls, where symptom resolution took 8.5 days, ensitrelvir shortened recovery time to as little as 1.4 days for over 66 % of patients. Conclusion Ensitrelvir treatment resulted in rapid symptom relief and significant viral load reduction, with no adverse events, viral rebound, or PASC symptoms, demonstrating its potential efficacy and safety. Larger studies are needed for further confirmation.
Collapse
Affiliation(s)
- Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Department of Infectious Diseases, Faculty of Medicine Yamagata University, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom
| | - Yu Suzuki
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Daisuke Akaneya
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Junko Igarashi
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Mayu Suto
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Kazunori Moriya
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Daisuke Ishizawa
- Department of Pharmacy, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Yoshikazu Okuma
- Department of Pharmacy, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Cameron Hurst
- Molly Wardaguga Research Centre, Charles Darwin University, Queensland, Australia
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Rangsit, Thailand, 10120
- Center of Excellence in Applied Epidemiology, Thammasat University, Rangsit, Thailand, 10120
| | - Thammakorn Saethang
- Department of Computer Science, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Paul G. Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Stephen M. Stick
- Wal-yan Respiratory Centre, Telethon Kids Institute, University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Centre, Telethon Kids Institute, University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
- School of Population Health, Curtin University, Bentley, 6102, Western Australia, Australia
| |
Collapse
|
6
|
Wang N, Lin Q, Fan H, Wang Y, Shu C, Wang N, Zhang D. BIC/FTC/TAF Benefits People Living With HIV After Omicron Breakthrough Infection in Shortening Duration of Symptoms, Enhancing Specific Immune Response and Increasing Total CD4 Cells. J Med Virol 2024; 96:e70036. [PMID: 39529466 PMCID: PMC11600477 DOI: 10.1002/jmv.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/03/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
The clinical manifestations of variability in PLWH with SARS-CoV-2 acquired may be attributed to the use of ART drugs. Omicron breakthrough infection and reinfection in 27 PLWH treated with BIC/FTC/TAF and 51 PLWH treated with TDF/3TC/EFV were investigated, and humoral and cellular immunity after Omicron breakthrough infection were also compared. The results showed that Omicron breakthrough infection symptoms in BIC/FTC/TAF-treated had a shorter duration, and BIC/FTC/TAF could further reduce the duration of symptoms when re-infected. It was also found that the BA.5 specific IgG titers of BIC/FTC/TAF-treated were significantly increased and increased with the extension of the duration of BIC/FTC/TAF. In cellular immunity, the use of BIC/FTC/TAF was also found to increase not only BA.5-specific B lymphocytes and BA.5-specific CD4+T lymphocytes, but also total CD4+T cells. In conclusion, compared to TDF/3TC/EFV, BIC/FTC/TAF can help PLWH shorten the duration of COVID-19 symptoms, enhance SARS-CoV-2 specific humoral and cellular immune responses, help to expand the total CD4+T cells increase in PLWH after Omicron infection and may be more beneficial for PLWH with high risk of infection in the context of COVID-19 normalization.
Collapse
Affiliation(s)
- Ni Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Qing Lin
- Department of Infectious DiseasesThe People's Hospital of Jiulongpo DistrictChongqingChina
| | - Huimin Fan
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Yixuan Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Chang Shu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Na Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
- Department of Infectious Diseases, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Dazhi Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
- Department of Infectious Diseases, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| |
Collapse
|
7
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
8
|
Zheng Q, Li Y, Sheng G, Li L. The Value of Ursodeoxycholic Acid and Mesenchymal Stem Cells in the Treatment of Severe COVID-19. Microorganisms 2024; 12:1269. [PMID: 39065038 PMCID: PMC11279161 DOI: 10.3390/microorganisms12071269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Objective: The objective of this study was to evaluate the therapeutic efficacy of ursodeoxycholic acid (UDCA) and mesenchymal stem cells (MSCs) in patients with severe COVID-19. Methods: We included severe COVID-19 patients hospitalized at Shulan (Hangzhou) Hospital between December 2022 and June 2023. We used a logistic regression model to compare the use of UDCA and MSCs in the two distinct groups of improved and poor outcomes. It is noteworthy that the deterioration group encompassed instances of both death and abandonment of treatment. The receiver operating characteristic (ROC) curve was plotted to assess the performance of the model. The aim was to assess the therapeutic effect of UDCA and MSCs on the outcome of severe COVID-19 patients. Results: A total of 167 patients with severe COVID-19 were included in this study. The analysis revealed that out of 42 patients (25.1%), 17 patients (10.2%) had taken UDCA, and 17 patients (10.2%) had used MSCs. Following a multivariable logistic regression, the results indicated a negative association between UDCA treatment (OR = 0.38 (0.16-0.91), p = 0.029), MSCs treatment (OR = 0.21 (0.07-0.65), p = 0.007), and the risk of severe COVID-19 mortality. Additionally, age showed a positive association with the risk of mortality (OR = 1.03 (1.01-1.07), p = 0.025). Conclusions: UDCA and MSCs have shown potential in improving the prognosis of severe COVID-19 patients and could be considered as additional treatments for COVID-19 in the future.
Collapse
Affiliation(s)
- Qi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| | - Yuetong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| | - Guoping Sheng
- Department of Infectious Disease, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou 310022, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| |
Collapse
|
9
|
Aktaş E, Özdemir Özgentürk N. A comprehensive examination of ACE2 receptor and prediction of spike glycoprotein and ACE2 interaction based on in silico analysis of ACE2 receptor. J Biomol Struct Dyn 2024; 42:4412-4428. [PMID: 37349943 DOI: 10.1080/07391102.2023.2220814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/28/2023] [Indexed: 06/24/2023]
Abstract
The ACE2 receptor plays a vital role not only in the SARS-CoV-induced epidemic but also in various other diseases, including cardiovascular diseases and ARDS. While studies have explored the interactions between ACE2 and SARS-CoV proteins, comprehensive research utilizing bioinformatic tools on the ACE2 protein has been lacking. The one aim of present study was to extensively analyze the regions of the ACE2 protein. After utilizing all bioinformatics tools especially G104 and L108 regions on ACE2 were come forward. The results of our analysis revealed that possible mutations or deletions in the G104 and L108 regions play a critical role in both the biological functioning and the determination of the chemical-physical properties of ACE2. Additionally, these regions were found to be more susceptible to mutations or deletions compared to other regions of the ACE2 protein. Notably, the randomly selected peptide, LQQNGSSVLS (100-109), which includes G104 and L108, exhibited a crucial role in binding the RBD of the spike protein, as supported by docking scores. Furthermore, both MDs and iMODs results provided evidence that G104 and L108 influence the dynamics of ACE2-spike complexes. This study is expected to offer a new perspective on the ACE2-SARS-CoV interaction and other research areas where ACE2 plays a significant role, such as biotechnology (protein engineering, enzyme optimization), medicine (RAS, pulmonary and cardiac diseases), and basic research (structural motifs, stabilizing protein folds, or facilitating important inter molecular contacts, protein's proper structure and function).Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Emre Aktaş
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
10
|
Zhao Z, Bashiri S, Ziora ZM, Toth I, Skwarczynski M. COVID-19 Variants and Vaccine Development. Viruses 2024; 16:757. [PMID: 38793638 PMCID: PMC11125726 DOI: 10.3390/v16050757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), the global pandemic caused by severe acute respiratory syndrome 2 virus (SARS-CoV-2) infection, has caused millions of infections and fatalities worldwide. Extensive SARS-CoV-2 research has been conducted to develop therapeutic drugs and prophylactic vaccines, and even though some drugs have been approved to treat SARS-CoV-2 infection, treatment efficacy remains limited. Therefore, preventive vaccination has been implemented on a global scale and represents the primary approach to combat the COVID-19 pandemic. Approved vaccines vary in composition, although vaccine design has been based on either the key viral structural (spike) protein or viral components carrying this protein. Therefore, mutations of the virus, particularly mutations in the S protein, severely compromise the effectiveness of current vaccines and the ability to control COVID-19 infection. This review begins by describing the SARS-CoV-2 viral composition, the mechanism of infection, the role of angiotensin-converting enzyme 2, the host defence responses against infection and the most common vaccine designs. Next, this review summarizes the common mutations of SARS-CoV-2 and how these mutations change viral properties, confer immune escape and influence vaccine efficacy. Finally, this review discusses global strategies that have been employed to mitigate the decreases in vaccine efficacy encountered against new variants.
Collapse
Affiliation(s)
- Ziyao Zhao
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (Z.Z.); (S.B.); (I.T.)
| | - Sahra Bashiri
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (Z.Z.); (S.B.); (I.T.)
| | - Zyta M. Ziora
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (Z.Z.); (S.B.); (I.T.)
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia;
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (Z.Z.); (S.B.); (I.T.)
| |
Collapse
|
11
|
Yousef M, Rob M, Varghese S, Rao S, Zamir F, Paul P, Chaari A. The effect of microbiome therapy on COVID-19-induced gut dysbiosis: A narrative and systematic review. Life Sci 2024; 342:122535. [PMID: 38408636 DOI: 10.1016/j.lfs.2024.122535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
AIMS Emerging evidence highlights the role of COVID-19 in instigating gut dysbiosis, with repercussions on disease severity and bidirectional gut-organ communication involving the lung, heart, brain, and liver. This study aims to evaluate the efficacy of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) in addressing gut dysbiosis associated with COVID-19, as well as their impact on related disease severity and clinical outcomes. MATERIALS AND METHODS We systematically review 27 studies exploring the efficacy of different microbiome-modulating therapies: probiotics, prebiotics, synbiotics, and fecal microbiota transplantation as potential interventions for COVID-19. KEY FINDINGS The probiotics and synbiotics investigated encompassed a spectrum of eight bacterial and fungal genera, namely Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, Pediococcus, Bacillus, Saccharomyces, and Kluyveromyces. Noteworthy prebiotics employed in these studies included chestnut tannin, galactooligosaccharides, fructooligosaccharides, xylooligosaccharide, and resistant dextrin. The majority of the investigated biotics exhibited positive effects on COVID-19 patients, manifesting in symptom alleviation, inflammation reduction, and notable decreases in mortality rates. Five studies reported death rates, showing an average mortality ranging from 0 % to 11 % in the intervention groups, as compared to 3 % to 30 % in the control groups. Specifically, probiotics, prebiotics, and synbiotics demonstrated efficacy in diminishing the duration and severity of symptoms while significantly accelerating viral and symptomatic remission. FMT emerged as a particularly effective strategy, successfully restoring gut microbiota and ameliorating gastrointestinal disorders. SIGNIFICANCE The insights gleaned from this review significantly contribute to our broader comprehension of the therapeutic potential of biotics in addressing COVID-19-related gut dysbiosis and mitigating secondary multi-organ complications.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Mlaak Rob
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Sanish Varghese
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Shrinidhi Rao
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Fahad Zamir
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
12
|
Wannigama DL, Hurst C, Phattharapornjaroen P, Hongsing P, Sirichumroonwit N, Chanpiwat K, Rad S.M. AH, Storer RJ, Ounjai P, Kanthawee P, Ngamwongsatit N, Kupwiwat R, Kupwiwat C, Brimson JM, Devanga Ragupathi NK, Charuluxananan S, Leelahavanichkul A, Kanjanabuch T, Higgins PG, Badavath VN, Amarasiri M, Verhasselt V, Kicic A, Chatsuwan T, Pirzada K, Jalali F, Reiersen AM, Abe S, Ishikawa H. Early treatment with fluvoxamine, bromhexine, cyproheptadine, and niclosamide to prevent clinical deterioration in patients with symptomatic COVID-19: a randomized clinical trial. EClinicalMedicine 2024; 70:102517. [PMID: 38516100 PMCID: PMC10955208 DOI: 10.1016/j.eclinm.2024.102517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
Background Repurposed drugs with host-directed antiviral and immunomodulatory properties have shown promise in the treatment of COVID-19, but few trials have studied combinations of these agents. The aim of this trial was to assess the effectiveness of affordable, widely available, repurposed drugs used in combination for treatment of COVID-19, which may be particularly relevant to low-resource countries. Methods We conducted an open-label, randomized, outpatient, controlled trial in Thailand from October 1, 2021, to June 21, 2022, to assess whether early treatment within 48-h of symptoms onset with combinations of fluvoxamine, bromhexine, cyproheptadine, and niclosamide, given to adults with confirmed mild SARS-CoV-2 infection, can prevent 28-day clinical deterioration compared to standard care. Participants were randomly assigned to receive treatment with fluvoxamine alone, fluvoxamine + bromhexine, fluvoxamine + cyproheptadine, niclosamide + bromhexine, or standard care. The primary outcome measured was clinical deterioration within 9, 14, or 28 days using a 6-point ordinal scale. This trial is registered with ClinicalTrials.gov (NCT05087381). Findings Among 1900 recruited, a total of 995 participants completed the trial. No participants had clinical deterioration by day 9, 14, or 28 days among those treated with fluvoxamine plus bromhexine (0%), fluvoxamine plus cyproheptadine (0%), or niclosamide plus bromhexine (0%). Nine participants (5.6%) in the fluvoxamine arm had clinical deterioration by day 28, requiring low-flow oxygen. In contrast, most standard care arm participants had clinical deterioration by 9, 14, and 28 days. By day 9, 32.7% (110) of patients in the standard care arm had been hospitalized without requiring supplemental oxygen but needing ongoing medical care. By day 28, this percentage increased to 37.5% (21). Additionally, 20.8% (70) of patients in the standard care arm required low-flow oxygen by day 9, and 12.5% (16) needed non-invasive or mechanical ventilation by day 28. All treated groups significantly differed from the standard care group by days 9, 14, and 28 (p < 0.0001). Also, by day 28, the three 2-drug treatments were significantly better than the fluvoxamine arm (p < 0.0001). No deaths occurred in any study group. Compared to standard care, participants treated with the combination agents had significantly decreased viral loads as early as day 3 of treatment (p < 0.0001), decreased levels of serum cytokines interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β) as early as day 5 of treatment, and interleukin-8 (IL-8) by day 7 of treatment (p < 0.0001) and lower incidence of post-acute sequelae of COVID-19 (PASC) symptoms (p < 0.0001). 23 serious adverse events occurred in the standard care arm, while only 1 serious adverse event was reported in the fluvoxamine arm, and zero serious adverse events occurred in the other arms. Interpretation Early treatment with these combinations among outpatients diagnosed with COVID-19 was associated with lower likelihood of clinical deterioration, and with significant and rapid reduction in the viral load and serum cytokines, and with lower burden of PASC symptoms. When started very soon after symptom onset, these repurposed drugs have high potential to prevent clinical deterioration and death in vaccinated and unvaccinated COVID-19 patients. Funding Ped Thai Su Phai (Thai Ducks Fighting Danger) social giver group.
Collapse
Affiliation(s)
- Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
| | - Cameron Hurst
- Molly Wardaguga Research Centre, Charles Darwin University, Queensland, Australia
| | - Phatthranit Phattharapornjaroen
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, Gothenburg University, 40530, Gothenburg, Sweden
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Natchalaikorn Sirichumroonwit
- Institute of Medical Research and Technology Assessment, Department of Medical Services, Ministry of Public Health, Thailand
| | | | - Ali Hosseini Rad S.M.
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9010, Otago, New Zealand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Robin James Storer
- Office of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Puey Ounjai
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Phitsanuruk Kanthawee
- Public Health Major, School of Health Science, Mae Fah Luang University, Chiang Rai, Thailand
| | - Natharin Ngamwongsatit
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Rosalyn Kupwiwat
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chaisit Kupwiwat
- Department of Critical Care Medicine, Vibhavadi Hospital, Bangkok, Thailand
| | - James Michael Brimson
- Department of Innovation and International Affair, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Naveen Kumar Devanga Ragupathi
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom
- Department of Chemical and Biological Engineering, The University of Sheffield, Sheffield, United Kingdom
- Division of Microbial Interactions, Department of Research and Development, Bioberrys Healthcare and Research Centre, Vellore, 632009, India
| | - Somrat Charuluxananan
- Department of Anesthesiology, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Talerngsak Kanjanabuch
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Kidney Metabolic Disorders, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Dialysis Policy and Practice Program (DiP3), School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Peritoneal Dialysis Excellence Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Paul G. Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50935, Cologne, Germany
| | - Vishnu Nayak Badavath
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Hyderabad, 509301, India
| | - Mohan Amarasiri
- Laboratory of Environmental Hygiene, Department of Health Science, School of Allied Health Sciences, Graduate School of Medical Sciences, Kitasato University, Kitasato, Sagamihara-Minami, Kanagawa, 252-0373, Japan
| | - Valerie Verhasselt
- Centre of Research for Immunology and Breastfeeding (CIBF), Medical School and School of Biomedical Science, University of Western Australia, Perth, Western Australia, 6009, Australia
- Immunology and Breastfeeding Group, Neonatal and Life Course Health Program, Telethon Kids Institute, Perth, Western Australia, 6009, Australia
| | - Anthony Kicic
- Telethon Kids Institute, University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
- School of Public Health, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kashif Pirzada
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Family and Community Medicine, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Farid Jalali
- Department of Gastroenterology, Saddleback Medical Group, Laguna Hills, CA, United States
| | - Angela M. Reiersen
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Hitoshi Ishikawa
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
| |
Collapse
|
13
|
Garcia-Vidal C, Teijón-Lumbreras C, Aiello TF, Chumbita M, Menendez R, Mateu-Subirà A, Peyrony O, Monzó P, Lopera C, Gallardo-Pizarro A, Méndez R, Calbo E, Xercavins M, Cuesta-Chasco G, Martínez JA, Marcos MA, Mensa J, Soriano A. K-Means Clustering Identifies Diverse Clinical Phenotypes in COVID-19 Patients: Implications for Mortality Risks and Remdesivir Impact. Infect Dis Ther 2024; 13:715-726. [PMID: 38489118 PMCID: PMC11058153 DOI: 10.1007/s40121-024-00938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/05/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION The impact of remdesivir on mortality in patients with COVID-19 is still controversial. We aimed to identify clinical phenotype clusters of COVID-19 hospitalized patients with highest benefit from remdesivir use and validate these findings in an external cohort. METHODS We included consecutive patients hospitalized between February 2020 and February 2021 for COVID-19. The derivation cohort comprised subjects admitted to Hospital Clinic of Barcelona. The validation cohort included patients from Hospital Universitari Mutua de Terrassa (Terrassa) and Hospital Universitari La Fe (Valencia), all tertiary centers in Spain. We employed K-means clustering to group patients according to reverse transcription polymerase chain reaction (rRT-PCR) cycle threshold (Ct) values and lymphocyte counts at diagnosis, and pre-test symptom duration. The impact of remdesivir on 60-day mortality in each cluster was assessed. RESULTS A total of 1160 patients (median age 66, interquartile range (IQR) 55-78) were included. We identified five clusters, with mortality rates ranging from 0 to 36.7%. Highest mortality rate was observed in the cluster including patients with shorter pre-test symptom duration, lower lymphocyte counts, and lower Ct values at diagnosis. The absence of remdesivir administration was associated with worse outcome in the high-mortality cluster (10.5% vs. 36.7%; p < 0.001), comprising subjects with higher viral loads. These results were validated in an external multicenter cohort of 981 patients. CONCLUSIONS Patients with COVID-19 exhibit varying mortality rates across different clinical phenotypes. K-means clustering aids in identifying patients who derive the greatest mortality benefit from remdesivir use.
Collapse
Affiliation(s)
- Carolina Garcia-Vidal
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain.
- CIBERINF, Barcelona, Spain.
| | - Christian Teijón-Lumbreras
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Tommaso Francesco Aiello
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain.
| | - Mariana Chumbita
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Rosario Menendez
- Respiratory Department, Hospital Universitari La Fe, Valencia, Spain
| | - Aina Mateu-Subirà
- Infectious Disease Department, Hospital Universitari Mutua de Terrassa, Terrassa, Barcelona, Spain
| | - Olivier Peyrony
- Emergency Department, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patricia Monzó
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Carlos Lopera
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Antonio Gallardo-Pizarro
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Raúl Méndez
- Respiratory Department, Hospital Universitari La Fe, Valencia, Spain
| | - Esther Calbo
- Infectious Disease Department, Hospital Universitari Mutua de Terrassa, Terrassa, Barcelona, Spain
- Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mariona Xercavins
- CATLAB. Hospital Universitari Mútua de Terrassa, Terrassa, Barcelona, Spain
| | - Genoveva Cuesta-Chasco
- Microbiology Department, Hospital Clinic, University of Barcelona, ISGLOBAL, Barcelona, Spain
| | - José A Martínez
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
- CIBERINF, Barcelona, Spain
| | - Ma Angeles Marcos
- CIBERINF, Barcelona, Spain
- Microbiology Department, Hospital Clinic, University of Barcelona, ISGLOBAL, Barcelona, Spain
| | - Josep Mensa
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
| | - Alex Soriano
- Infectious Disease Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, C/ Villarroel 170, 08036, Barcelona, Spain
- CIBERINF, Barcelona, Spain
| |
Collapse
|
14
|
Ono R, Iwahana T, Aoki K, Kato H, Okada S, Kobayashi Y. Fulminant Myocarditis with SARS-CoV-2 Infection: A Narrative Review from the Case Studies. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:9000598. [PMID: 38469104 PMCID: PMC10927348 DOI: 10.1155/2024/9000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
One of the severe complications of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is myocarditis. However, the characteristics of fulminant myocarditis with SARS-CoV-2 infection are still unclear. We systematically reviewed the previously reported cases of fulminant myocarditis associated with SARS-CoV-2 infection from January 2020 to December 2022, identifying 108 cases. Of those, 67 were male and 41 female. The average age was 34.8 years; 30 patients (27.8%) were ≤20 years old, whereas 10 (9.3%) were ≥60. Major comorbidities included hypertension, obesity, diabetes mellitus, asthma, heart disease, gynecologic disease, hyperlipidemia, and connective tissue disorders. Regarding left ventricular ejection fraction (LVEF) at admission, 93% of the patients with fulminant myocarditis were classified as having heart failure with reduced ejection fraction (LVEF ≤ 40%). Most of the cases were administered catecholamines (97.8%), and mechanical circulatory support (MCS) was required in 67 cases (62.0%). The type of MCS was extracorporeal membrane oxygenation (n = 56, 83.6%), percutaneous ventricular assist device (Impella®) (n = 19, 28.4%), intra-aortic balloon pumping (n = 12, 12.9%), or right ventricular assist device (n = 2, 3.0%); combination of these devices occurred in 20 cases (29.9%). The average duration of MCS was 7.7 ± 3.8 days. Of the 76 surviving patients whose cardiac function was available for follow-up, 65 (85.5%) recovered normally. The overall mortality rate was 22.4%, and the recovery rate was 77.6% (alive: 83 patients, dead: 24 patients; outcome not described: 1 patient).
Collapse
Affiliation(s)
- Ryohei Ono
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Togo Iwahana
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kaoruko Aoki
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hirotoshi Kato
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
15
|
Brogna C, Montano L, Zanolin ME, Bisaccia DR, Ciammetti G, Viduto V, Fabrowski M, Baig AM, Gerlach J, Gennaro I, Bignardi E, Brogna B, Frongillo A, Cristoni S, Piscopo M. A retrospective cohort study on early antibiotic use in vaccinated and unvaccinated COVID-19 patients. J Med Virol 2024; 96:e29507. [PMID: 38504586 DOI: 10.1002/jmv.29507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/14/2024] [Accepted: 02/23/2024] [Indexed: 03/21/2024]
Abstract
The bacteriophage behavior of SARS-CoV-2 during the acute and post-COVID-19 phases appears to be an important factor in the development of the disease. The early use of antibiotics seems to be crucial to inhibit disease progression-to prevent viral replication in the gut microbiome, and control toxicological production from the human microbiome. To study the impact of specific antibiotics on recovery from COVID-19 and long COVID (LC) taking into account: vaccination status, comorbidities, SARS-CoV-2 wave, time of initiation of antibiotic therapy and concomitant use of corticosteroids and nonsteroidal anti-inflammatory drugs (NSAIDs). A total of 211 COVID-19 patients were included in the study: of which 59 were vaccinated with mRNA vaccines against SARS-CoV-2 while 152 were unvaccinated. Patients were enrolled in three waves: from September 2020 to October 2022, corresponding to the emergence of the pre-Delta, Delta, and Omicron variants of the SARS-CoV-2 virus. The three criteria for enrolling patients were: oropharyngeal swab positivity or fecal findings; moderate symptoms with antibiotic intake; and measurement of blood oxygen saturation during the period of illness. The use of antibiotic combinations, such as amoxicillin with clavulanic acid (875 + 125 mg tablets, every 12 h) plus rifaximin (400 mg tablets every 12 h), as first choice, as suggested from the previous data, or azithromycin (500 mg tablets every 24 h), plus rifaximin as above, allows healthcare professionals to focus on the gut microbiome and its implications in COVID-19 disease during patient care. The primary outcome measured in this study was the estimated average treatment effect, which quantified the difference in mean recovery between patients receiving antibiotics and those not receiving antibiotics at 3 and 9 days after the start of treatment. In the analysis, both vaccinated and unvaccinated groups had a median illness duration of 7 days (interquartile range [IQR] 6-9 days for each; recovery crude hazard ratio [HR] = 0.94, p = 0.700). The median illness duration for the pre-Delta and Delta waves was 8 days (IQR 7-10 days), while it was shorter, 6.5 days, for Omicron (IQR 6-8 days; recovery crude HR = 1.71, p < 0.001). These results were confirmed by multivariate analysis. Patients with comorbidities had a significantly longer disease duration: median 8 days (IQR 7-10 days) compared to 7 days (IQR 6-8 days) for those without comorbidities (crude HR = 0.75, p = 0.038), but this result was not confirmed in multivariate analysis as statistical significance was lost. Early initiation of antibiotic therapy resulted in a significantly shorter recovery time (crude HR = 4.74, p < 0.001). Concomitant use of NSAIDs did not reduce disease duration and in multivariate analysis prolonged the disease (p = 0.041). A subgroup of 42 patients receiving corticosteroids for a median of 3 days (IQR 3-6 days) had a longer recovery time (median 9 days, IQR 8-10 days) compared to others (median 7 days, IQR 6-8 days; crude HR = 0.542, p < 0.001), as confirmed also by the adjusted HR. In this study, a statistically significant reduction in recovery time was observed among patients who received early antibiotic treatment. Early initiation of antibiotics played a crucial role in maintaining higher levels of blood oxygen saturation. In addition, it is worth noting that a significant number of patients who received antibiotics in the first 3 days and for a duration of 7 days, during the acute phase did not develop LC.
Collapse
Affiliation(s)
- Carlo Brogna
- Craniomed Group Srl. Research Facility, Bresso, Italy
| | - Luigi Montano
- Andrology Unit and Service of Lifestyle Medicine in Uro-Andrology, Local Health Authority (ASL), Salerno, Italy
| | | | | | - Gianluca Ciammetti
- Otorhinolaryngology Unit, Hospital Ferdinando Veneziale Isernia, Regional Health Authority of Molise, Italy
| | | | - Mark Fabrowski
- Department of Emergency Medicine, Royal Sussex County Hospital, University Hospitals Sussex, Brighton, UK
| | - Abdul M Baig
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Iapicca Gennaro
- Pineta Grande Hospital Group, Department of Urology, Santa Rita Clinic, Atripalda, Italy
| | | | - Barbara Brogna
- Department of Radiology, Moscati Hospital, Avellino, Italy
| | | | | | - Marina Piscopo
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
16
|
Khalil AM, Nogales A, Martínez-Sobrido L, Mostafa A. Antiviral responses versus virus-induced cellular shutoff: a game of thrones between influenza A virus NS1 and SARS-CoV-2 Nsp1. Front Cell Infect Microbiol 2024; 14:1357866. [PMID: 38375361 PMCID: PMC10875036 DOI: 10.3389/fcimb.2024.1357866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Following virus recognition of host cell receptors and viral particle/genome internalization, viruses replicate in the host via hijacking essential host cell machinery components to evade the provoked antiviral innate immunity against the invading pathogen. Respiratory viral infections are usually acute with the ability to activate pattern recognition receptors (PRRs) in/on host cells, resulting in the production and release of interferons (IFNs), proinflammatory cytokines, chemokines, and IFN-stimulated genes (ISGs) to reduce virus fitness and mitigate infection. Nevertheless, the game between viruses and the host is a complicated and dynamic process, in which they restrict each other via specific factors to maintain their own advantages and win this game. The primary role of the non-structural protein 1 (NS1 and Nsp1) of influenza A viruses (IAV) and the pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respectively, is to control antiviral host-induced innate immune responses. This review provides a comprehensive overview of the genesis, spatial structure, viral and cellular interactors, and the mechanisms underlying the unique biological functions of IAV NS1 and SARS-CoV-2 Nsp1 in infected host cells. We also highlight the role of both non-structural proteins in modulating viral replication and pathogenicity. Eventually, and because of their important role during viral infection, we also describe their promising potential as targets for antiviral therapy and the development of live attenuated vaccines (LAV). Conclusively, both IAV NS1 and SARS-CoV-2 Nsp1 play an important role in virus-host interactions, viral replication, and pathogenesis, and pave the way to develop novel prophylactic and/or therapeutic interventions for the treatment of these important human respiratory viral pathogens.
Collapse
Affiliation(s)
- Ahmed Magdy Khalil
- Disease Intervention & Prevention and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, TX, United States
- Department of Zoonotic Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Madrid, Spain
| | - Luis Martínez-Sobrido
- Disease Intervention & Prevention and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Ahmed Mostafa
- Disease Intervention & Prevention and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, TX, United States
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| |
Collapse
|
17
|
Arman BY, Brun J, Hill ML, Zitzmann N, von Delft A. An Update on SARS-CoV-2 Clinical Trial Results-What We Can Learn for the Next Pandemic. Int J Mol Sci 2023; 25:354. [PMID: 38203525 PMCID: PMC10779148 DOI: 10.3390/ijms25010354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 7 million lives worldwide, providing a stark reminder of the importance of pandemic preparedness. Due to the lack of approved antiviral drugs effective against coronaviruses at the start of the pandemic, the world largely relied on repurposed efforts. Here, we summarise results from randomised controlled trials to date, as well as selected in vitro data of directly acting antivirals, host-targeting antivirals, and immunomodulatory drugs. Overall, repurposing efforts evaluating directly acting antivirals targeting other viral families were largely unsuccessful, whereas several immunomodulatory drugs led to clinical improvement in hospitalised patients with severe disease. In addition, accelerated drug discovery efforts during the pandemic progressed to multiple novel directly acting antivirals with clinical efficacy, including small molecule inhibitors and monoclonal antibodies. We argue that large-scale investment is required to prepare for future pandemics; both to develop an arsenal of broad-spectrum antivirals beyond coronaviruses and build worldwide clinical trial networks that can be rapidly utilised.
Collapse
Affiliation(s)
- Benediktus Yohan Arman
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Juliane Brun
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Michelle L. Hill
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Nicole Zitzmann
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Annette von Delft
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- Centre for Medicine Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
18
|
Seah VXF, Ong RYL, Kam KQ, Thoon KC, Tan NWH, Li J, Nadua KD, Chong CY, Yung CF. Remdesivir therapy for severe pediatric COVID-19 in Singapore: A single-center retrospective observational cohort study. Health Sci Rep 2023; 6:e1698. [PMID: 38098972 PMCID: PMC10719654 DOI: 10.1002/hsr2.1698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/14/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Background and Aims There is a paucity of information on remdesivir (RDV) use in severe pediatric coronavirus disease 2019 (COVID-19). We aimed to explore the effectiveness of RDV as the cumulative proportion of pediatric COVID-19 patients deescalated from Day 5 of high dependency or intensive care unit (HD/ICU). Methods All children ≤18 years admitted to Singapore's largest pediatric hospital from January 1, 2020 to March 18, 2022 were reviewed retrospectively. Patients were included if they were positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on reverse transcriptase polymerase chain reaction, required oxygen, and HD/ICU care. The characteristics and outcomes of those who received RDV or not (no-RDV) were compared. Results We reviewed 15 children with a median age of 2.5 years (interquartile range [IQR]: 0.8-11.0), of which 7 (46.7%) received RDV. There was no difference in cumulative proportion of children deescalated from Day 5 of HD/ICU care in the RDV versus the no-RDV group (5/7, 70% vs. 7/8, 87.5%, p = 0.57). The RDV versus no-RDV group had higher disease severity, that is, WHO Ordinal Scale scores (median 6, IQR: 5-7 vs. 5, IQR: 4-5, p = 0.03), higher procalcitonin levels (ug/L) (median 4.31, IQR: 0.8-24.2 vs. 0.12, IQR: 0.09-0.26, p = 0.02), and longer HD/ICU care days (median 5, IQR: 4-9, vs. 1, IQR: 1-4, p = 0.01). There was no significant difference in hospitalization days. There were no adverse events directly attributable to RDV. None died from COVID-19 infection. Conclusion Our observational analysis was unable to detect any clear benefit of RDV in terms of reducing duration in HD/ICU. RDV was well-tolerated in children with severe COVID-19.
Collapse
Affiliation(s)
| | - Rina Yue Ling Ong
- Department of PharmacyKK Women's and Children's HospitalSingaporeSingapore
| | - Kai Qian Kam
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Koh Cheng Thoon
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Natalie Woon Hui Tan
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Jiahui Li
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Karen Donceras Nadua
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Chia Yin Chong
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Chee Fu Yung
- Infectious Disease Service, Department of PaediatricsKK Women's and Children's HospitalSingaporeSingapore
- Duke‐National University of Singapore Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
19
|
Lushington GH, Linde A, Melgarejo T. Bacterial Proteases as Potentially Exploitable Modulators of SARS-CoV-2 Infection: Logic from the Literature, Informatics, and Inspiration from the Dog. BIOTECH 2023; 12:61. [PMID: 37987478 PMCID: PMC10660736 DOI: 10.3390/biotech12040061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/19/2023] [Accepted: 10/18/2023] [Indexed: 11/22/2023] Open
Abstract
(1) Background: The COVID-19 pandemic left many intriguing mysteries. Retrospective vulnerability trends tie as strongly to odd demographics as to exposure profiles, genetics, health, or prior medical history. This article documents the importance of nasal microbiome profiles in distinguishing infection rate trends among differentially affected subgroups. (2) Hypothesis: From a detailed literature survey, microbiome profiling experiments, bioinformatics, and molecular simulations, we propose that specific commensal bacterial species in the Pseudomonadales genus confer protection against SARS-CoV-2 infections by expressing proteases that may interfere with the proteolytic priming of the Spike protein. (3) Evidence: Various reports have found elevated Moraxella fractions in the nasal microbiomes of subpopulations with higher resistance to COVID-19 (e.g., adolescents, COVID-19-resistant children, people with strong dietary diversity, and omnivorous canines) and less abundant ones in vulnerable subsets (the elderly, people with narrower diets, carnivorous cats and foxes), along with bioinformatic evidence that Moraxella bacteria express proteases with notable homology to human TMPRSS2. Simulations suggest that these proteases may proteolyze the SARS-CoV-2 spike protein in a manner that interferes with TMPRSS2 priming.
Collapse
Affiliation(s)
| | - Annika Linde
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Tonatiuh Melgarejo
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
20
|
Rasmussen HB, Hansen PR. Molnupiravir Revisited-Critical Assessment of Studies in Animal Models of COVID-19. Viruses 2023; 15:2151. [PMID: 38005828 PMCID: PMC10675540 DOI: 10.3390/v15112151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 11/26/2023] Open
Abstract
Molnupiravir, a prodrug known for its broad antiviral activity, has demonstrated efficacy in animal models of COVID-19, prompting clinical trials, in which initial results indicated a significant effect against the disease. However, subsequent clinical studies did not confirm these findings, leading to the refusal of molnupiravir for permanent market authorization in many countries. This report critically assessed 22 studies published in 18 reports that investigated the efficacy of molnupiravir in animal models of COVID-19, with the purpose of determining how well the design of these models informed human studies. We found that the administered doses of molnupiravir in most studies involving animal COVID-19 models were disproportionately higher than the dose recommended for human use. Specifically, when adjusted for body surface area, over half of the doses of molnupiravir used in the animal studies exceeded twice the human dose. Direct comparison of reported drug exposure across species after oral administration of molnupiravir indicated that the antiviral efficacy of the dose recommended for human use was underestimated in some animal models and overestimated in others. Frequently, molnupiravir was given prophylactically or shortly after SARS-CoV-2 inoculation in these models, in contrast to clinical trials where such timing is not consistently achieved. Furthermore, the recommended five-day treatment duration for humans was exceeded in several animal studies. Collectively, we suggest that design elements in the animal studies under examination contributed to a preference favoring molnupiravir, and thus inflated expectations for its efficacy against COVID-19. Addressing these elements may offer strategies to enhance the clinical efficacy of molnupiravir for the treatment of COVID-19. Such strategies include dose increment, early treatment initiation, administration by inhalation, and use of the drug in antiviral combination therapy.
Collapse
Affiliation(s)
- Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, 4000 Roskilde, Denmark
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Peter Riis Hansen
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2900 Hellerup, Denmark;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
21
|
Guo BC, Wu KH, Chen CY, Lin WY, Chang YJ, Lee TA, Lin MJ, Wu HP. Mesenchymal Stem Cells in the Treatment of COVID-19. Int J Mol Sci 2023; 24:14800. [PMID: 37834246 PMCID: PMC10573267 DOI: 10.3390/ijms241914800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Since the emergence of the coronavirus disease 2019 (COVID-19) pandemic, many lives have been tragically lost to severe infections. The COVID-19 impact extends beyond the respiratory system, affecting various organs and functions. In severe cases, it can progress to acute respiratory distress syndrome (ARDS) and multi-organ failure, often fueled by an excessive immune response known as a cytokine storm. Mesenchymal stem cells (MSCs) have considerable potential because they can mitigate inflammation, modulate immune responses, and promote tissue regeneration. Accumulating evidence underscores the efficacy and safety of MSCs in treating severe COVID-19 and ARDS. Nonetheless, critical aspects, such as optimal routes of MSC administration, appropriate dosage, treatment intervals, management of extrapulmonary complications, and potential pediatric applications, warrant further exploration. These research avenues hold promise for enriching our understanding and refining the application of MSCs in confronting the multifaceted challenges posed by COVID-19.
Collapse
Affiliation(s)
- Bei-Cyuan Guo
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chun-Yu Chen
- Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 43503, Taiwan;
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Wen-Ya Lin
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung 43503, Taiwan
| | - Yu-Jun Chang
- Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Tai-An Lee
- Department of Emergency Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan;
| | - Mao-Jen Lin
- Division of Cardiology, Department of Medicine, Taichung Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97002, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
22
|
Elkousy RH, Said ZNA, Ali MA, Kutkat O, Abu El Wafa SA. Anti-SARS-CoV-2 in vitro potential of castor oil plant ( Ricinus communis) leaf extract: in-silico virtual evidence. Z NATURFORSCH C 2023; 78:365-376. [PMID: 37401758 DOI: 10.1515/znc-2023-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023]
Abstract
Ricinus communis L. is a medicinal plant that displays valuable pharmacological properties, including antioxidant, antimicrobial, analgesic, antibacterial, antiviral and anti-inflammatory properties. This study targeted to isolate and identify some constituents of R. communis leaves using ultra-performance liquid chromatography coupled with mass spectroscopy (UPLC-MS/MS) and different chromatographic techniques. In vitro anti-MERS and anti-SARS-CoV-2 activity for different fractions and for two pure isolated compounds, lupeol (RS) and ricinine (RS1) were evaluated using a plaque reduction assay with three different mechanisms and IC50 based on their cytotoxic concentration (CC50) from an MTT assay using Vero E6 cell line. Isolated phytoconstituents and remdesivir are assessed for in-silico anti-COVID-19 activity using molecular docking tools. The methylene chloride extract showed pronounced virucidal activity against SARS-CoV-2 (IC50 = 1.76 μg/ml). It was also shown that ricinine had superior potential activity against SARS-CoV-2, (IC50 = 2.5 μg/ml). Lupeol displayed the most potency against MERS, (IC50 = 5.28 μg/ml). Ricinine appeared to be the most biologically active compound. The study showed that R. communis and its isolated compounds have potential natural virucidal activity against SARS-COV-2; however, additional exploration is necessary and study for their in vivo activity.
Collapse
Affiliation(s)
- Rawah H Elkousy
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (for Girls), Al-Azhar University, P.O. Box 11651, Nasr City, Cairo, Egypt
| | - Zeinab N A Said
- Department of Medical Microbiology & Immunology, Faculty of Medicine (for Girls), Al-Azhar University, P.O. Box 11754, Nasr City, Cairo, Egypt
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, P.O. Box 12622, Giza, Egypt
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, P.O. Box 12622, Giza, Egypt
| | - Salwa A Abu El Wafa
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (for Girls), Al-Azhar University, P.O. Box 11651, Nasr City, Cairo, Egypt
| |
Collapse
|
23
|
Tucci AR, da Rosa RM, Rosa AS, Augusto Chaves O, Ferreira VNS, Oliveira TKF, Coutinho Souza DD, Borba NRR, Dornelles L, Rocha NS, Mayer JCP, da Rocha JBT, Rodrigues OED, Miranda MD. Antiviral Effect of 5'-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023; 28:6696. [PMID: 37764472 PMCID: PMC10537738 DOI: 10.3390/molecules28186696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The understanding that zidovudine (ZDV or azidothymidine, AZT) inhibits the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 and that chalcogen atoms can increase the bioactivity and reduce the toxicity of AZT has directed our search for the discovery of novel potential anti-coronavirus compounds. Here, the antiviral activity of selenium and tellurium containing AZT derivatives in human type II pneumocytes cell model (Calu-3) and monkey kidney cells (Vero E6) infected with SARS-CoV-2, and their toxic effects on these cells, was evaluated. Cell viability analysis revealed that organoselenium (R3a-R3e) showed lower cytotoxicity than organotellurium (R3f, R3n-R3q), with CC50 ≥ 100 µM. The R3b and R3e were particularly noteworthy for inhibiting viral replication in both cell models and showed better selectivity index. In Vero E6, the EC50 values for R3b and R3e were 2.97 ± 0.62 µM and 1.99 ± 0.42 µM, respectively, while in Calu-3, concentrations of 3.82 ± 1.42 µM and 1.92 ± 0.43 µM (24 h treatment) and 1.33 ± 0.35 µM and 2.31 ± 0.54 µM (48 h) were observed, respectively. The molecular docking calculations were carried out to main protease (Mpro), papain-like protease (PLpro), and RdRp following non-competitive, competitive, and allosteric inhibitory approaches. The in silico results suggested that the organoselenium is a potential non-competitive inhibitor of RdRp, interacting in the allosteric cavity located in the palm region. Overall, the cell-based results indicated that the chalcogen-zidovudine derivatives were more potent than AZT in inhibiting SARS-CoV-2 replication and that the compounds R3b and R3e play an important inhibitory role, expanding the knowledge about the promising therapeutic capacity of organoselenium against COVID-19.
Collapse
Affiliation(s)
- Amanda Resende Tucci
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Raquel Mello da Rosa
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Otávio Augusto Chaves
- CQC-IMS, Departamento de Química, Universidade de Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
- Laboratório de Imunofarmacologia, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Daniel Dias Coutinho Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Nathalia Roberto Resende Borba
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Luciano Dornelles
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Nayra Salazar Rocha
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João Candido Pilar Mayer
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João B. Teixeira da Rocha
- Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Oscar Endrigo D. Rodrigues
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| |
Collapse
|
24
|
Cristea AM, Zaharia DC, Jipa-Duna D, Dumitrache-Rujinski S, Parliteanu OA, Bogdan AM, Toma CL. Predictors of negative outcomes in hospitalized patients with SARS‑CoV‑2 pneumonia: A retrospective study. Exp Ther Med 2023; 26:437. [PMID: 37614431 PMCID: PMC10443062 DOI: 10.3892/etm.2023.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic posed a serious threat to human health worldwide after the first case was identified in December 2019. Specific therapeutic options for COVID-19 are lacking; thus, the treatment of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is complex in clinical practice. Despite the development of treatment options and methods to limit the spread of SARS-CoV-2, certain patients experience critical illness and numerous deaths have occurred. Notably, treatment of this disease is complex due to the evolution of viral mutations and variants with different rates of infection. Moreover, specific patient characteristics may be associated with rapid disease progression and poor outcomes. Thus, the present study aimed to identify the specific characteristics of patients who developed poor outcomes, including clinical manifestations, blood samples (blood cell count and coagulation tests) at hospital admission and comorbidities. The present study included a total of 1,813 patients hospitalized with pneumonia and SARS-CoV-2 infection, and mortality rates associated with each patient characteristic were calculated. The characteristics associated with the highest risk of mortality were as follows: Age >90 years (OR, 105; 95% CI, 17.70-2,023.00); oxygen saturation at the time of hospital admission <89% in room air (OR, 14.3; 95% CI, 7.54-30.7), admission to the Intensive Care Unit (OR, 39.4; 95% CI, 27.7-57.0); and a neutrophil/lymphocyte ratio of 8.76-54.2 (OR, 14; 95% CI, 7.62-29.0). Treatment of patients with SARS-CoV-2 pneumonia represents a challenge for the healthcare system, but there are a number of predictors for poor patient outcomes that could be identified at the time of hospital admission.
Collapse
Affiliation(s)
- Alexandra-Maria Cristea
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
- Department of Pneumology VII, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| | - Dragos-Cosmin Zaharia
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
- Department of Pneumology VII, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| | - Daniela Jipa-Duna
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
- Department of Pneumology II, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| | - Stefan Dumitrache-Rujinski
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
- Department of Pneumology IV, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| | - Oana Andreea Parliteanu
- Department of Ambulatory Diabetes, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| | - Alexandru Miron Bogdan
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
| | - Claudia Lucia Toma
- Department of Pneumology I, Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania
- Department of Pneumology IV, Marius Nasta Institute of Pneumology, Bucharest 050159, Romania
| |
Collapse
|
25
|
Starkey T, Ionescu MC, Tilby M, Little M, Burke E, Fittall MW, Khan S, Liu JKH, Platt JR, Mew R, Tripathy AR, Watts I, Williams ST, Appanna N, Al-Hajji Y, Barnard M, Benny L, Burnett A, Bytyci J, Cattell EL, Cheng V, Clark JJ, Eastlake L, Gerrand K, Ghafoor Q, Grumett S, Harper-Wynne C, Kahn R, Lee AJX, Lomas O, Lydon A, Mckenzie H, Panneerselvam H, Pascoe JS, Patel G, Patel V, Potter VA, Randle A, Rigg AS, Robinson TM, Roylance R, Roques TW, Rozmanowski S, Roux RL, Shah K, Sheehan R, Sintler M, Swarup S, Taylor H, Tillett T, Tuthill M, Williams S, Ying Y, Beggs A, Iveson T, Lee SM, Middleton G, Middleton M, Protheroe A, Fowler T, Johnson P, Lee LYW. A population-scale temporal case-control evaluation of COVID-19 disease phenotype and related outcome rates in patients with cancer in England (UKCCP). Sci Rep 2023; 13:11327. [PMID: 37491478 PMCID: PMC10368624 DOI: 10.1038/s41598-023-36990-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/14/2023] [Indexed: 07/27/2023] Open
Abstract
Patients with cancer are at increased risk of hospitalisation and mortality following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, the SARS-CoV-2 phenotype evolution in patients with cancer since 2020 has not previously been described. We therefore evaluated SARS-CoV-2 on a UK populationscale from 01/11/2020-31/08/2022, assessing case-outcome rates of hospital assessment(s), intensive care admission and mortality. We observed that the SARS-CoV-2 disease phenotype has become less severe in patients with cancer and the non-cancer population. Case-hospitalisation rates for patients with cancer dropped from 30.58% in early 2021 to 7.45% in 2022 while case-mortality rates decreased from 20.53% to 3.25%. However, the risk of hospitalisation and mortality remains 2.10x and 2.54x higher in patients with cancer, respectively. Overall, the SARS-CoV-2 disease phenotype is less severe in 2022 compared to 2020 but patients with cancer remain at higher risk than the non-cancer population. Patients with cancer must therefore be empowered to live more normal lives, to see loved ones and families, while also being safeguarded with expanded measures to reduce the risk of transmission.
Collapse
Affiliation(s)
- Thomas Starkey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | | | - Michael Tilby
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Emma Burke
- Oxford University Hospitals NHS Trust, Oxford, UK
| | | | - Sam Khan
- University of Leicester, Leicester, UK
| | | | - James R Platt
- Leeds Institute of Medical Research at St James's, Leeds, UK
| | - Rosie Mew
- Torbay and South Devon NHS Foundation Trust, Torquay, UK
| | | | | | | | | | - Youssra Al-Hajji
- Birmingham Medical School, University of Birmingham, Birmingham, UK
| | | | | | | | - Jola Bytyci
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Qamar Ghafoor
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Simon Grumett
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | | | - Oliver Lomas
- Oxford University Hospitals NHS Trust, Oxford, UK
| | - Anna Lydon
- Torbay and South Devon NHS Foundation Trust, Torquay, UK
| | - Hayley Mckenzie
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Jennifer S Pascoe
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | - Vanessa A Potter
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | | | - Anne S Rigg
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Rebecca Roylance
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Tom W Roques
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | | | - René L Roux
- Oxford University Hospitals NHS Trust, Oxford, UK
| | - Ketan Shah
- Oxford University Hospitals NHS Trust, Oxford, UK
| | - Remarez Sheehan
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Martin Sintler
- Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | | | | | | | - Mark Tuthill
- Oxford University Hospitals NHS Trust, Oxford, UK
| | - Sarah Williams
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Yuxin Ying
- Department of Oncology, University of Oxford, Oxford, UK
| | - Andrew Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Tim Iveson
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Siow Ming Lee
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mark Middleton
- Department of Oncology, University of Oxford, Oxford, UK
| | - Andrew Protheroe
- Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Tom Fowler
- UK Health Security Agency, London, UK
- William Harvey Research Institute, London, UK
| | | | - Lennard Y W Lee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
- Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
26
|
von Delft A, Hall MD, Kwong AD, Purcell LA, Saikatendu KS, Schmitz U, Tallarico JA, Lee AA. Accelerating antiviral drug discovery: lessons from COVID-19. Nat Rev Drug Discov 2023; 22:585-603. [PMID: 37173515 PMCID: PMC10176316 DOI: 10.1038/s41573-023-00692-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, a wave of rapid and collaborative drug discovery efforts took place in academia and industry, culminating in several therapeutics being discovered, approved and deployed in a 2-year time frame. This article summarizes the collective experience of several pharmaceutical companies and academic collaborations that were active in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral discovery. We outline our opinions and experiences on key stages in the small-molecule drug discovery process: target selection, medicinal chemistry, antiviral assays, animal efficacy and attempts to pre-empt resistance. We propose strategies that could accelerate future efforts and argue that a key bottleneck is the lack of quality chemical probes around understudied viral targets, which would serve as a starting point for drug discovery. Considering the small size of the viral proteome, comprehensively building an arsenal of probes for proteins in viruses of pandemic concern is a worthwhile and tractable challenge for the community.
Collapse
Affiliation(s)
- Annette von Delft
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, National Institute for Health Research, University of Oxford, Oxford, UK.
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | - Alpha A Lee
- PostEra, Inc., Cambridge, MA, USA.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Madewell ZJ, Yang Y, Longini IM, Halloran ME, Vespignani A, Dean NE. Rapid review and meta-analysis of serial intervals for SARS-CoV-2 Delta and Omicron variants. BMC Infect Dis 2023; 23:429. [PMID: 37365505 PMCID: PMC10291789 DOI: 10.1186/s12879-023-08407-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The serial interval is the period of time between symptom onset in the primary case and symptom onset in the secondary case. Understanding the serial interval is important for determining transmission dynamics of infectious diseases like COVID-19, including the reproduction number and secondary attack rates, which could influence control measures. Early meta-analyses of COVID-19 reported serial intervals of 5.2 days (95% CI: 4.9-5.5) for the original wild-type variant and 5.2 days (95% CI: 4.87-5.47) for Alpha variant. The serial interval has been shown to decrease over the course of an epidemic for other respiratory diseases, which may be due to accumulating viral mutations and implementation of more effective nonpharmaceutical interventions. We therefore aggregated the literature to estimate serial intervals for Delta and Omicron variants. METHODS This study followed Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. A systematic literature search was conducted of PubMed, Scopus, Cochrane Library, ScienceDirect, and preprint server medRxiv for articles published from April 4, 2021, through May 23, 2023. Search terms were: ("serial interval" or "generation time"), ("Omicron" or "Delta"), and ("SARS-CoV-2" or "COVID-19"). Meta-analyses were done for Delta and Omicron variants using a restricted maximum-likelihood estimator model with a random effect for each study. Pooled average estimates and 95% confidence intervals (95% CI) are reported. RESULTS There were 46,648 primary/secondary case pairs included for the meta-analysis of Delta and 18,324 for Omicron. Mean serial interval for included studies ranged from 2.3-5.8 days for Delta and 2.1-4.8 days for Omicron. The pooled mean serial interval for Delta was 3.9 days (95% CI: 3.4-4.3) (20 studies) and Omicron was 3.2 days (95% CI: 2.9-3.5) (20 studies). Mean estimated serial interval for BA.1 was 3.3 days (95% CI: 2.8-3.7) (11 studies), BA.2 was 2.9 days (95% CI: 2.7-3.1) (six studies), and BA.5 was 2.3 days (95% CI: 1.6-3.1) (three studies). CONCLUSIONS Serial interval estimates for Delta and Omicron were shorter than ancestral SARS-CoV-2 variants. More recent Omicron subvariants had even shorter serial intervals suggesting serial intervals may be shortening over time. This suggests more rapid transmission from one generation of cases to the next, consistent with the observed faster growth dynamic of these variants compared to their ancestors. Additional changes to the serial interval may occur as SARS-CoV-2 continues to circulate and evolve. Changes to population immunity (due to infection and/or vaccination) may further modify it.
Collapse
Affiliation(s)
- Zachary J Madewell
- Department of Biostatistics, University of Florida, Gainesville, FL, USA.
| | - Yang Yang
- Department of Statistics, University of Georgia, Athens, GA, USA
| | - Ira M Longini
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - M Elizabeth Halloran
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alessandro Vespignani
- Laboratory for the Modeling of Biological and Socio-Technical Systems, Northeastern University, Boston, MA, USA
| | - Natalie E Dean
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| |
Collapse
|
28
|
Foidart JM, Simon K, Utian WH, Mauvais-Jarvis F, Douxfils J, Dixon G, Barrington P. Estetrol Is Safe and Well Tolerated during Treatment of Hospitalized Men and Women with Moderate COVID-19 in a Randomized, Double-Blind Study. J Clin Med 2023; 12:3928. [PMID: 37373625 DOI: 10.3390/jcm12123928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Epidemiological data suggest that the severe acute respiratory syndrome coronavirus 2 infection rate is higher in women than in men, but the death rate is lower, while women (>50 years) on menopausal hormone therapy (MHT) have a higher survival rate than those not on MHT. Classical oral estrogen enhances the synthesis of coagulation markers and may increase the risk of thromboembolic events that are common in coronavirus disease 2019 (COVID-19). The favorable hemostatic profile of estetrol (E4) might be suitable for use in women who are receiving estrogen treatment and contract COVID-19. A multicenter, randomized, double-blind, placebo-controlled, phase 2 study (NCT04801836) investigated the efficacy, safety, and tolerability of E4 versus placebo in hospitalized patients with moderate COVID-19. Eligible postmenopausal women and men (aged ≥ 18 years old) were randomized to E4 15 mg or placebo, once daily for 21 days, in addition to the standard of care (SoC). The primary efficacy endpoint of improvement in COVID-19 (percentage of patients recovered at day 28) between the placebo and E4 arms was not met. E4 was well tolerated, with no safety signals or thromboembolic events, suggesting that postmenopausal women can safely continue E4-based therapy in cases of moderate COVID-19 managed with SoC.
Collapse
Affiliation(s)
- Jean Michel Foidart
- Mithra Pharmaceuticals, 4000 Liège, Belgium
- Department of Obstetrics and Gynecology, University of Liège, 4000 Liège, Belgium
| | - Krzysztof Simon
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51149 Wrocław, Poland
| | - Wulf H Utian
- Department of Reproductive Biology, Case Western Reserve Medical School, Cleveland, OH 44106, USA
| | - Franck Mauvais-Jarvis
- Department of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jonathan Douxfils
- Department of Pharmacy, Namur Thrombosis and Hemostasis Center, Faculty of Medicine, University of Namur, 5000 Namur, Belgium
- QUALIblood s.a., 5000 Namur, Belgium
| | | | | |
Collapse
|
29
|
Lea AN, Leyden WA, Sofrygin O, Marafino BJ, Skarbinski J, Napravnik S, Agil D, Augenbraun M, Benning L, Horberg MA, Jefferson C, Marconi VC, Park LS, Gordon KS, Bastarache L, Gangireddy S, Althoff KN, Coburn SB, Gebo KA, Lang R, Williams C, Silverberg MJ. Human Immunodeficiency Virus Status, Tenofovir Exposure, and the Risk of Poor Coronavirus Disease 19 Outcomes: Real-World Analysis From 6 United States Cohorts Before Vaccine Rollout. Clin Infect Dis 2023; 76:1727-1734. [PMID: 36861341 PMCID: PMC10209434 DOI: 10.1093/cid/ciad084] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND People with human immunodeficiency virus (HIV) (PWH) may be at increased risk for severe coronavirus disease 2019 (COVID-19) outcomes. We examined HIV status and COVID-19 severity, and whether tenofovir, used by PWH for HIV treatment and people without HIV (PWoH) for HIV prevention, was associated with protection. METHODS Within 6 cohorts of PWH and PWoH in the United States, we compared the 90-day risk of any hospitalization, COVID-19 hospitalization, and mechanical ventilation or death by HIV status and by prior exposure to tenofovir, among those with severe acute respiratory syndrome coronavirus 2 infection between 1 March and 30 November 2020. Adjusted risk ratios (aRRs) were estimated by targeted maximum likelihood estimation, with adjustment for demographics, cohort, smoking, body mass index, Charlson comorbidity index, calendar period of first infection, and CD4 cell counts and HIV RNA levels (in PWH only). RESULTS Among PWH (n = 1785), 15% were hospitalized for COVID-19 and 5% received mechanical ventilation or died, compared with 6% and 2%, respectively, for PWoH (n = 189 351). Outcome prevalence was lower for PWH and PWoH with prior tenofovir use. In adjusted analyses, PWH were at increased risk compared with PWoH for any hospitalization (aRR, 1.31 [95% confidence interval, 1.20-1.44]), COVID-19 hospitalizations (1.29 [1.15-1.45]), and mechanical ventilation or death (1.51 [1.19-1.92]). Prior tenofovir use was associated with reduced hospitalizations among PWH (aRR, 0.85 [95% confidence interval, .73-.99]) and PWoH (0.71 [.62-.81]). CONCLUSIONS Before COVID-19 vaccine availability, PWH were at greater risk for severe outcomes than PWoH. Tenofovir was associated with a significant reduction in clinical events for both PWH and PWoH.
Collapse
Affiliation(s)
- Alexandra N Lea
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Wendy A Leyden
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Oleg Sofrygin
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Ben J Marafino
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Jacek Skarbinski
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Oakland Medical Center, Kaiser Permanente Northern California, Oakland, California, USA
| | - Sonia Napravnik
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Deana Agil
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Augenbraun
- Division of Infectious Diseases, State University of New York Health Sciences University, Brooklyn, USA
| | - Lorie Benning
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Michael A Horberg
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente Mid-Atlantic States, Rockville, Maryland, USA
| | - Celeena Jefferson
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente Mid-Atlantic States, Rockville, Maryland, USA
| | - Vincent C Marconi
- Emory University School of Medicine and Rollins School of Public Health, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Lesley S Park
- Stanford Department of Epidemiology & Population Health, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kirsha S Gordon
- VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Srushti Gangireddy
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keri N Althoff
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sally B Coburn
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kelly A Gebo
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raynell Lang
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carolyn Williams
- Epidemiology Branch, Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Michael J Silverberg
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| |
Collapse
|
30
|
Franco EJ, Drusano GL, Hanrahan KC, Warfield KL, Brown AN. Combination Therapy with UV-4B and Molnupiravir Enhances SARS-CoV-2 Suppression. Viruses 2023; 15:1175. [PMID: 37243261 PMCID: PMC10224493 DOI: 10.3390/v15051175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The host targeting antiviral, UV-4B, and the RNA polymerase inhibitor, molnupiravir, are two orally available, broad-spectrum antivirals that have demonstrated potent activity against SARS-CoV-2 as monotherapy. In this work, we evaluated the effectiveness of UV-4B and EIDD-1931 (molnupiravir's main circulating metabolite) combination regimens against the SARS-CoV-2 beta, delta, and omicron BA.2 variants in a human lung cell line. Infected ACE2 transfected A549 (ACE2-A549) cells were treated with UV-4B and EIDD-1931 both as monotherapy and in combination. Viral supernatant was sampled on day three when viral titers peaked in the no-treatment control arm, and levels of infectious virus were measured by plaque assay. The drug-drug effect interaction between UV-4B and EIDD-1931 was also defined using the Greco Universal Response Surface Approach (URSA) model. Antiviral evaluations demonstrated that treatment with UV-4B plus EIDD-1931 enhanced antiviral activity against all three variants relative to monotherapy. These results were in accordance with those obtained from the Greco model, as these identified the interaction between UV-4B and EIDD-1931 as additive against the beta and omicron variants and synergistic against the delta variant. Our findings highlight the anti-SARS-CoV-2 potential of UV-4B and EIDD-1931 combination regimens, and present combination therapy as a promising therapeutic strategy against SARS-CoV-2.
Collapse
Affiliation(s)
- Evelyn J. Franco
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (G.L.D.); (K.C.H.)
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| | - George L. Drusano
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (G.L.D.); (K.C.H.)
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (G.L.D.); (K.C.H.)
| | | | - Ashley N. Brown
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (G.L.D.); (K.C.H.)
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| |
Collapse
|
31
|
Khalaji A, Behnoush AH, Alilou S, Rezaee M, Peiman S, Sahebkar A. Adjunctive therapy with lipid-lowering agents in COVID-19: a systematic review and meta-analysis of randomized controlled trials. Lipids Health Dis 2023; 22:61. [PMID: 37158917 PMCID: PMC10165571 DOI: 10.1186/s12944-023-01828-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Many commonly used drugs were evaluated as repurposed treatment options since the emergence of the COVID-19 pandemic. The benefit of lipid-lowering agents has been controversial in this regard. In this systematic review, we assessed the effect of these medications as adjunctive therapy in COVID-19 by the inclusion of randomized controlled trials (RCTs). METHODS We searched four international databases including PubMed, the Web of Science, Scopus, and Embase for RCTs in April 2023. The primary outcome was mortality, while other efficacy indices were considered secondary outcomes. In order to estimate the pooled effect size of the outcomes, considering the odds ratio (OR) or standardized mean difference (SMD) and 95% confidence interval (CI), random-effect meta-analyses was conducted. RESULTS Ten studies involving 2,167 COVID-19 patients using statins, omega-3 fatty acids, fenofibrate, PCSK9 inhibitors, and nicotinamide as intervention compared to control or placebo, were included. No significant difference was found in terms of mortality (OR 0.96, 95% CI 0.58 to 1.59, p-value = 0.86, I2 = 20.4%) or length of hospital stay (SMD -0.10, 95% CI -0.78 to 0.59, p-value = 0.78, I2 = 92.4%) by adding a statin to the standard of care. The trend was similar for fenofibrate and nicotinamide. PCSK9 inhibition, however, led to decreased mortality and an overall better prognosis. Omega-3 supplementation showed contradicting results in two trials, suggesting the need for further evaluation. CONCLUSION Although some observational studies found improved outcomes in patients using lipid-lowering agents, our study found no benefit in adding statins, fenofibrate, or nicotinamide to COVID-19 treatment. On the other hand, PCSK9 inhibitors can be a good candidate for further assessment. Finally, there are major limitations in the use of omega-3 supplements in treating COVID-19 and more trials are warranted to evaluate this efficacy.
Collapse
Affiliation(s)
- Amirmohammad Khalaji
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Non–Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Behnoush
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Non–Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Alilou
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheil Peiman
- Department of Internal Medicine, AdventHealth Orlando Hospital, Orlando, FL USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Rezapour A, Behroozi Z, Nasirzadeh M, Rezaeian M, Barzegar M, Tashakori-Miyanroudi M, Sayyad A, Souresrafil A. Cost-effectiveness of remdesivir for the treatment of hospitalized patients with COVID-19: a systematic review. Infect Dis Poverty 2023; 12:39. [PMID: 37081575 PMCID: PMC10116457 DOI: 10.1186/s40249-023-01092-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/04/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Remdesivir is being studied and used to treat coronavirus disease 2019 (COVID-19). This study aimed to systematically identify, critically evaluate, and summarize the findings of the studies on the cost-effectiveness of remdesivir in the treatment of hospitalized patients with COVID-19. METHODS In this systematic review, PubMed, EMBASE, Web of Science, SCOPUS, and the Cochrane Library were searched for studies published between 2019 and 2022. We included all full economic evaluations of remdesivir for the treatment of hospitalized patients with COVID-19. Data were summarized in a structured and narrative manner. RESULTS Out of 616 articles obtained in this literature search, 12 studies were included in the final analysis. The mean score of the Quality of Health Economic Studies (QHES) for the studies was 87.66 (high quality). All studies were conducted in high-income countries (eight studies in the USA and one study in England), except for three studies from middle-to-high-income countries (China, South Africa, and Turkey). Six studies conducted their economic analysis in terms of a health system perspective; five studies conducted their economic analysis from a payer perspective; three studies from the perspective of a health care provider. The results of five studies showed that remdesivir was cost-effective compared to standard treatment. Furthermore, the therapeutic strategy of combining remdesivir with baricitinib was cost-effective compared to remdesivir alone. CONCLUSIONS Based on the results of the present study, remdesivir appears to be cost-effective in comparison with the standard of care in China, Turkey, and South Africa. Studies conducted in the United States show conflicting results, and combining remdesivir with baricitinib is cost-effective compared with remdesivir alone. However, the cost-effectiveness of remdesivir in low-income countries remains unknown. Thus, more studies in different countries are required to determine the cost-effectiveness of this drug.
Collapse
Affiliation(s)
- Aziz Rezapour
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mostafa Nasirzadeh
- Department of Health Education and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohsen Rezaeian
- Department of Epidemiology and Biostatistics, School of Medicine, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Barzegar
- Department of English Language Teaching, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Tashakori-Miyanroudi
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdollah Sayyad
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Aghdas Souresrafil
- Department of Health Services and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
33
|
Astărăstoae V, Rogozea LM. Against Authority: The Bioethics of Ivermectin Use for COVID-19 Infection. Am J Ther 2023:00045391-990000000-00143. [PMID: 37068020 DOI: 10.1097/mjt.0000000000001629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND The COVID-19 pandemic has brought new ethical challenges to both health care professionals and the general public. Among the ethical problems amplified during this period were the making of medical decisions to quickly introduce some drugs into therapeutic practice with unproven or insufficiently proven effects (such as ivermectin), the validity of drug testing, and the allocation of limited resources. FIELDS OF UNCERTAINTY The COVID-19 pandemic brought to the attention of the entire scientific world a new problem, which exceeded the guidelines and rules known until then. Out of the desire to quickly solve this medical problem, a series of measures were taken, however not sufficiently validated in scientific terms; the recommendations regarding the use of drugs known for their properties to treat a greater number of conditions, such as ivermectin, was tried. DATA SOURCES A narrative review of the specialized literature was carried out using keywords such as COVID-19, ivermectin, ethics, and off-label medication from Scopus and Google Scholar but also of official documents developed at the international level (World Health Organization). ETHICS AND THERAPEUTIC ADVANCES The off-label use of ivermectin alone or in combination with other medications during COVID pandemic raised problems related to the demonstration of its effectiveness, but also to ethics, starting from the expectations that both the medical staff and the population had of it. Ivermectin therapy was also evaluated by analyzing the behavior of ivermectin based on ethical principles (nonmaleficence, beneficence, and respect for one's autonomy) or on justice. Even in times of pandemic, exceptionalism must not triumph, and finding an effective treatment must be done through studies that respect ethical standard. CONCLUSIONS The failures or rather lack of success in decision making during the pandemic showed that alongside scientific knowledge and the development of health policies, it is necessary to constantly evaluate the measures and decisions from an ethical point of view, and the prevention of slippages and abuses is not only necessary but even mandatory.
Collapse
Affiliation(s)
- Vasile Astărăstoae
- Faculty of Medicine, Grigore T Popa University of Medicine & Pharmacy, Iasi, Romania; and
| | - Liliana M Rogozea
- Basic, Preventive and Clinical Sciences Department, Transilvania University, Brasov, Romania
| |
Collapse
|
34
|
Xu P, Wang X, Shi J, Chen W, Lu ZJ, Jia H, Ye D, Li X. Functionally Collaborative Nanostructure for Direct Monitoring of Neurotransmitter Exocytosis in Living Cells. NANO LETTERS 2023; 23:2427-2435. [PMID: 36715488 DOI: 10.1021/acs.nanolett.2c04117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Neurotransmitter exocytosis of living cells plays a vital role in neuroscience. However, the available amperometric technique with carbon fiber electrodes typically measures exocytotic events from one cell during one procedure, which requires professional operations and takes time to produce statistical results of multiple cells. Here, we develop a functionally collaborative nanostructure to directly measure the neurotransmitter dopamine (DA) exocytosis from living rat pheochromocytoma (PC12) cells. The functionally collaborative nanostructure is constructed of metal-organic framework (MOF)-on-nanowires-on-graphene oxide, which is highly sensitive to DA molecules and enables direct detection of neurotransmitter exocytosis. Using the microsensor, the exocytosis from PC12 cells pretreated with the desired drugs (e.g., anticoronavirus drug, antiflu drug, or anti-inflammatory drug) has been successfully measured. Our achievements demonstrate the feasibility of the functionally collaborative nanostructure in the real-time detection of exocytosis and the potential applicability in the highly efficient assessment of the modulation effects of medications on exocytosis.
Collapse
Affiliation(s)
- Pengcheng Xu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- School of Microelectronics, University of Chinese Academy of Sciences, Beijing100049, China
| | - Xuefeng Wang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- School of Microelectronics, University of Chinese Academy of Sciences, Beijing100049, China
| | - Jiaci Shi
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Wei Chen
- Department of Emergency, Tongji Hospital, Tongji University School of Medicine, Shanghai200065, China
| | - Zhan-Jun Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200080, China
| | - Hao Jia
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- School of Microelectronics, University of Chinese Academy of Sciences, Beijing100049, China
| | - Daixin Ye
- Department of Chemistry, Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai200444, China
| | - Xinxin Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- School of Microelectronics, University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
35
|
Zhu B, Wei X, Narasimhan H, Qian W, Zhang R, Cheon IS, Wu Y, Li C, Jones RG, Kaplan MH, Vassallo RA, Braciale TJ, Somerville L, Colca JR, Pandey A, Jackson PEH, Mann BJ, Krawczyk CM, Sturek JM, Sun J. Inhibition of the mitochondrial pyruvate carrier simultaneously mitigates hyperinflammation and hyperglycemia in COVID-19. Sci Immunol 2023; 8:eadf0348. [PMID: 36821695 PMCID: PMC9972900 DOI: 10.1126/sciimmunol.adf0348] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The relationship between diabetes and COVID-19 is bi-directional: while individuals with diabetes and high blood glucose (hyperglycemia) are predisposed to severe COVID-19, SARS-CoV-2 infection can also cause hyperglycemia and exacerbate underlying metabolic syndrome. Therefore, interventions capable of breaking the network of SARS-CoV-2 infection, hyperglycemia, and hyper-inflammation, all factors that drive COVID-19 pathophysiology, are urgently needed. Here, we show that genetic ablation or pharmacological inhibition of mitochondrial pyruvate carrier (MPC) attenuates severe disease following influenza or SARS-CoV-2 pneumonia. MPC inhibition using a second-generation insulin sensitizer, MSDC-0602 K (MSDC), dampened pulmonary inflammation and promoted lung recovery, while concurrently reducing blood glucose levels and hyperlipidemia following viral pneumonia in obese mice. Mechanistically, MPC inhibition enhanced mitochondrial fitness and destabilized HIF-1α, leading to dampened virus-induced inflammatory responses in both murine and human lung macrophages. We further showed that MSDC enhanced responses to nirmatrelvir (the antiviral component of Paxlovid) to provide high levels of protection against severe host disease development following SARS-CoV-2 infection and suppressed cellular inflammation in human COVID-19 lung autopsies, demonstrating its translational potential for treating severe COVID-19. Collectively, we uncover a metabolic pathway that simultaneously modulates pulmonary inflammation, tissue recovery, and host metabolic health, presenting a synergistic therapeutic strategy to treat severe COVID-19, particularly in patients with underlying metabolic disease.
Collapse
Affiliation(s)
- Bibo Zhu
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaoqin Wei
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Harish Narasimhan
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ruixuan Zhang
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - In Su Cheon
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Yue Wu
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Chaofan Li
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University of School of Medicine, Indianapolis, IN 46202, USA
| | - Robert A Vassallo
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Thomas J Braciale
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Lindsay Somerville
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Patrick E H Jackson
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Barbara J Mann
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Connie M Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Jeffrey M Sturek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
36
|
Saqib K, Qureshi AS, Butt ZA. COVID-19, Mental Health, and Chronic Illnesses: A Syndemic Perspective. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3262. [PMID: 36833955 PMCID: PMC9962717 DOI: 10.3390/ijerph20043262] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The COVID-19 pandemic is an epidemiological and psychological crisis; what it does to the body is quite well known by now, and more research is underway, but the syndemic impact of COVID-19 and mental health on underlying chronic illnesses among the general population is not completely understood. METHODS We carried out a literature review to identify the potential impact of COVID-19 and related mental health issues on underlying comorbidities that could affect the overall health of the population. RESULTS Many available studies have highlighted the impact of COVID-19 on mental health only, but how complex their interaction is in patients with comorbidities and COVID-19, the absolute risks, and how they connect with the interrelated risks in the general population, remain unknown. The COVID-19 pandemic can be recognized as a syndemic due to; synergistic interactions among different diseases and other health conditions, increasing overall illness burden, emergence, spread, and interactions between infectious zoonotic diseases leading to new infectious zoonotic diseases; this is together with social and health interactions leading to increased risks in vulnerable populations and exacerbating clustering of multiple diseases. CONCLUSION There is a need to develop evidence to support appropriate and effective interventions for the overall improvement of health and psychosocial wellbeing of at-risk populations during this pandemic. The syndemic framework is an important framework that can be used to investigate and examine the potential benefits and impact of codesigning COVID-19/non-communicable diseases (NCDs)/mental health programming services which can tackle these epidemics concurrently.
Collapse
Affiliation(s)
- Kiran Saqib
- School of Public health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Afaf Saqib Qureshi
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zahid Ahmad Butt
- School of Public health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
37
|
Centofanti F, Buono A, Verboni M, Tomino C, Lucarini S, Duranti A, Pandolfi PP, Novelli G. Synthetic Methodologies and Therapeutic Potential of Indole-3-Carbinol (I3C) and Its Derivatives. Pharmaceuticals (Basel) 2023; 16:240. [PMID: 37259386 PMCID: PMC9960368 DOI: 10.3390/ph16020240] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 01/01/2025] Open
Abstract
Indole-3-carbinol (I3C) is a natural product contained in vegetables belonging to the Brassicaceae family and has been studied in recent decades for its biological and pharmacological properties. Herein, we will analyze: (1) the biosynthetic processes and synthetic procedures through which I3C and its main derivatives have been obtained; (2) the characteristics that lead to believe that both I3C and its derivatives are responsible for several important activities-in particular, antitumor and antiviral, through insights concerning in vitro assays and in vivo tests; (3) the mechanisms of action of the most important compounds considered; (4) the potential social impact that the enhancement of the discussed molecules can have in the prevention and treatment of the pathologies' examined field-first of all, those related to respiratory tract disorders and cancer.
Collapse
Affiliation(s)
- Federica Centofanti
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessandro Buono
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Verboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Carlo Tomino
- Scientific Direction—IRCCS San Raffaele Rome, 00166 Rome, Italy
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Pier Paolo Pandolfi
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV 89502, USA
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
38
|
Grundeis F, Ansems K, Dahms K, Thieme V, Metzendorf MI, Skoetz N, Benstoem C, Mikolajewska A, Griesel M, Fichtner F, Stegemann M. Remdesivir for the treatment of COVID-19. Cochrane Database Syst Rev 2023; 1:CD014962. [PMID: 36695483 PMCID: PMC9875553 DOI: 10.1002/14651858.cd014962.pub2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Remdesivir is an antiviral medicine approved for the treatment of mild-to-moderate coronavirus disease 2019 (COVID-19). This led to widespread implementation, although the available evidence remains inconsistent. This update aims to fill current knowledge gaps by identifying, describing, evaluating, and synthesising all evidence from randomised controlled trials (RCTs) on the effects of remdesivir on clinical outcomes in COVID-19. OBJECTIVES To assess the effects of remdesivir and standard care compared to standard care plus/minus placebo on clinical outcomes in patients treated for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. SEARCH METHODS We searched the Cochrane COVID-19 Study Register (which comprises the Cochrane Central Register of Controlled Trials (CENTRAL), PubMed, Embase, ClinicalTrials.gov, World Health Organization (WHO) International Clinical Trials Registry Platform, and medRxiv) as well as Web of Science (Science Citation Index Expanded and Emerging Sources Citation Index) and WHO COVID-19 Global literature on coronavirus disease to identify completed and ongoing studies, without language restrictions. We conducted the searches on 31 May 2022. SELECTION CRITERIA We followed standard Cochrane methodology. We included RCTs evaluating remdesivir and standard care for the treatment of SARS-CoV-2 infection compared to standard care plus/minus placebo irrespective of disease severity, gender, ethnicity, or setting. We excluded studies that evaluated remdesivir for the treatment of other coronavirus diseases. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess risk of bias in included studies, we used the Cochrane RoB 2 tool for RCTs. We rated the certainty of evidence using the GRADE (Grading of Recommendations, Assessment, Development and Evaluation) approach for outcomes that were reported according to our prioritised categories: all-cause mortality, in-hospital mortality, clinical improvement (being alive and ready for discharge up to day 28) or worsening (new need for invasive mechanical ventilation or death up to day 28), quality of life, serious adverse events, and adverse events (any grade). We differentiated between non-hospitalised individuals with asymptomatic SARS-CoV-2 infection or mild COVID-19 and hospitalised individuals with moderate to severe COVID-19. MAIN RESULTS We included nine RCTs with 11,218 participants diagnosed with SARS-CoV-2 infection and a mean age of 53.6 years, of whom 5982 participants were randomised to receive remdesivir. Most participants required low-flow oxygen at baseline. Studies were mainly conducted in high- and upper-middle-income countries. We identified two studies that are awaiting classification and five ongoing studies. Effects of remdesivir in hospitalised individuals with moderate to severe COVID-19 With moderate-certainty evidence, remdesivir probably makes little or no difference to all-cause mortality at up to day 28 (risk ratio (RR) 0.93, 95% confidence interval (CI) 0.81 to 1.06; risk difference (RD) 8 fewer per 1000, 95% CI 21 fewer to 6 more; 4 studies, 7142 participants), day 60 (RR 0.85, 95% CI 0.69 to 1.05; RD 35 fewer per 1000, 95% CI 73 fewer to 12 more; 1 study, 1281 participants), or in-hospital mortality at up to day 150 (RR 0.93, 95% CI 0.84 to 1.03; RD 11 fewer per 1000, 95% CI 25 fewer to 5 more; 1 study, 8275 participants). Remdesivir probably increases the chance of clinical improvement at up to day 28 slightly (RR 1.11, 95% CI 1.06 to 1.17; RD 68 more per 1000, 95% CI 37 more to 105 more; 4 studies, 2514 participants; moderate-certainty evidence). It probably decreases the risk of clinical worsening within 28 days (hazard ratio (HR) 0.67, 95% CI 0.54 to 0.82; RD 135 fewer per 1000, 95% CI 198 fewer to 69 fewer; 2 studies, 1734 participants, moderate-certainty evidence). Remdesivir may make little or no difference to the rate of adverse events of any grade (RR 1.04, 95% CI 0.92 to 1.18; RD 23 more per 1000, 95% CI 46 fewer to 104 more; 4 studies, 2498 participants; low-certainty evidence), or serious adverse events (RR 0.84, 95% CI 0.65 to 1.07; RD 44 fewer per 1000, 95% CI 96 fewer to 19 more; 4 studies, 2498 participants; low-certainty evidence). We considered risk of bias to be low, with some concerns for mortality and clinical course. We had some concerns for safety outcomes because participants who had died did not contribute information. Without adjustment, this leads to an uncertain amount of missing values and the potential for bias due to missing data. Effects of remdesivir in non-hospitalised individuals with mild COVID-19 One of the nine RCTs was conducted in the outpatient setting and included symptomatic people with a risk of progression. No deaths occurred within the 28 days observation period. We are uncertain about clinical improvement due to very low-certainty evidence. Remdesivir probably decreases the risk of clinical worsening (hospitalisation) at up to day 28 (RR 0.28, 95% CI 0.11 to 0.75; RD 46 fewer per 1000, 95% CI 57 fewer to 16 fewer; 562 participants; moderate-certainty evidence). We did not find any data for quality of life. Remdesivir may decrease the rate of serious adverse events at up to 28 days (RR 0.27, 95% CI 0.10 to 0.70; RD 49 fewer per 1000, 95% CI 60 fewer to 20 fewer; 562 participants; low-certainty evidence), but it probably makes little or no difference to the risk of adverse events of any grade (RR 0.91, 95% CI 0.76 to 1.10; RD 42 fewer per 1000, 95% CI 111 fewer to 46 more; 562 participants; moderate-certainty evidence). We considered risk of bias to be low for mortality, clinical improvement, and safety outcomes. We identified a high risk of bias for clinical worsening. AUTHORS' CONCLUSIONS Based on the available evidence up to 31 May 2022, remdesivir probably has little or no effect on all-cause mortality or in-hospital mortality of individuals with moderate to severe COVID-19. The hospitalisation rate was reduced with remdesivir in one study including participants with mild to moderate COVID-19. It may be beneficial in the clinical course for both hospitalised and non-hospitalised patients, but certainty remains limited. The applicability of the evidence to current practice may be limited by the recruitment of participants from mostly unvaccinated populations exposed to early variants of the SARS-CoV-2 virus at the time the studies were undertaken. Future studies should provide additional data on the efficacy and safety of remdesivir for defined core outcomes in COVID-19 research, especially for different population subgroups.
Collapse
Affiliation(s)
- Felicitas Grundeis
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Kelly Ansems
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Karolina Dahms
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Volker Thieme
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Maria-Inti Metzendorf
- Institute of General Practice, Medical Faculty of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Carina Benstoem
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Agata Mikolajewska
- Centre for Biological Threats and Special Pathogens (ZBS), Strategy and Incident Response, Clinical Management and Infection Control, Robert Koch Institute, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mirko Griesel
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Falk Fichtner
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
39
|
Prophylactic Administration of the Monoclonal Antibody Adintrevimab Protects against SARS-CoV-2 in Hamster and Non-Human Primate Models of COVID-19. Antimicrob Agents Chemother 2023; 67:e0135322. [PMID: 36519929 PMCID: PMC9872613 DOI: 10.1128/aac.01353-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Adintrevimab is a human immunoglobulin G1 monoclonal antibody engineered to have broad neutralization against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and other SARS-like coronaviruses with pandemic potential. In both Syrian golden hamster and rhesus macaque models, prophylactic administration of a single dose of adintrevimab provided protection against SARS-CoV-2/WA1/2020 infection in a dose-dependent manner, as measured by significant reductions in lung viral load and virus-induced lung pathology, and by inhibition of viral replication in the upper and lower respiratory tract.
Collapse
|
40
|
Baghela A, An A, Zhang P, Acton E, Gauthier J, Brunet-Ratnasingham E, Blimkie T, Freue GC, Kaufmann D, Lee AHY, Levesque RC, Hancock REW. Predicting severity in COVID-19 disease using sepsis blood gene expression signatures. Sci Rep 2023; 13:1247. [PMID: 36690713 PMCID: PMC9868505 DOI: 10.1038/s41598-023-28259-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Severely-afflicted COVID-19 patients can exhibit disease manifestations representative of sepsis, including acute respiratory distress syndrome and multiple organ failure. We hypothesized that diagnostic tools used in managing all-cause sepsis, such as clinical criteria, biomarkers, and gene expression signatures, should extend to COVID-19 patients. Here we analyzed the whole blood transcriptome of 124 early (1-5 days post-hospital admission) and late (6-20 days post-admission) sampled patients with confirmed COVID-19 infections from hospitals in Quebec, Canada. Mechanisms associated with COVID-19 severity were identified between severity groups (ranging from mild disease to the requirement for mechanical ventilation and mortality), and established sepsis signatures were assessed for dysregulation. Specifically, gene expression signatures representing pathophysiological events, namely cellular reprogramming, organ dysfunction, and mortality, were significantly enriched and predictive of severity and lethality in COVID-19 patients. Mechanistic endotypes reflective of distinct sepsis aetiologies and therapeutic opportunities were also identified in subsets of patients, enabling prediction of potentially-effective repurposed drugs. The expression of sepsis gene expression signatures in severely-afflicted COVID-19 patients indicates that these patients should be classified as having severe sepsis. Accordingly, in severe COVID-19 patients, these signatures should be strongly considered for the mechanistic characterization, diagnosis, and guidance of treatment using repurposed drugs.
Collapse
Affiliation(s)
- Arjun Baghela
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | - Andy An
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | | | - Erica Acton
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Jeff Gauthier
- Institut de Biologie Intégrative et des Systèmes (IBIS), Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Quebec, QC, Canada
| | - Elsa Brunet-Ratnasingham
- Département de Microbiologie, Infectiologie Et Immunologie, Université de Montréal, Montreal, Canada
- Centre de Recherche du CHUM, Montreal, QC, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | | | - Daniel Kaufmann
- Centre de Recherche du CHUM, Montreal, QC, Canada
- Département de Médecine, Université de Montréal, Montreal, Canada
| | - Amy H Y Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Roger C Levesque
- Institut de Biologie Intégrative et des Systèmes (IBIS), Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Quebec, QC, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada.
- Asep Medical, Vancouver, Canada.
| |
Collapse
|
41
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
42
|
Kapusta J, Chudzik M, Kałuzińska-Kołat Ż, Kołat D, Burzyńska M, Jankowski P, Babicki M. Do selected lifestyle parameters affect the severity and symptoms of COVID-19 among elderly patients? The retrospective evaluation of individuals from the STOP-COVID registry of the PoLoCOV study. J Infect Public Health 2023; 16:143-153. [PMID: 36521330 PMCID: PMC9743693 DOI: 10.1016/j.jiph.2022.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Older individuals tend to include less physical activity in their routine and are more prone to chronic diseases and severe medical complications, making them the most burdened group that is losing years of life due to pandemic-related premature mortality. This research aimed to assess the lifestyle factors that affect the COVID-19 course among patients ≥ 65 years old. METHODS The study included 568 convalescents (64.1% women and 35.9% men) with persistent clinical symptoms after isolation. The mean age was 70.41 ± 4.64 years (minimum: 65 years; maximum: 89 years). The patients completed the questionnaire during their in-person visit to the medical center. The survey included questions regarding their health status when suffering from COVID-19, basic sociodemographic data, and medical history concerning chronic conditions and lifestyle. RESULTS Physical inactivity (p < 0.001) and feeling nervous (p = 0.026) increased the risk of having a severe disease course. Coronary artery disease raised both the risk of a severe disease course (p = 0.002) and the number of present symptoms up to 4 weeks (p = 0.039). Sleep disturbances increased the number of symptoms during infection (p = 0.001). The occurrence of any symptoms was also associated with the female sex (p = 0.004). The severity of the course was associated with longer persistent symptoms (p < 0.001) and a greater number of symptoms (p = 0.004); those with a more severe course were also at a greater risk of persistent symptoms for up to 4 weeks (p = 0.006). Senior citizens in the third pandemic wave suffered with more severe disease (p = 0.004), while illness during the fourth (p = 0.001) and fifth (p < 0.001) waves was associated with a lower risk of persistent symptoms for up to 4 weeks. The disease duration was significantly shorter among vaccinated patients (p = 0.042). CONCLUSIONS Elderly COVID-19 patients should re-think their lifestyle habits to consider a physical activity level that is adjusted to their abilities, in order to decrease the risk of a severe disease course and to further limit both the number and duration of symptoms. The research was carried out in accordance with the Declaration of Helsinki, and approval from the Bioethics Committee of Lodz Regional Medical Chamber to conduct the study was obtained (approval number 0115/2021). The PoLoCOV-Study ClinicalTrials.gov identifier is NCT05018052.
Collapse
Affiliation(s)
- Joanna Kapusta
- Department of Internal Medicine and Cardiac Rehabilitation, Medical University of Lodz, 70-445 Lodz, Poland.
| | - Michał Chudzik
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, 01-813 Warsaw, Poland; Boruta Medical Center, 95-100 Zgierz, Poland.
| | - Żaneta Kałuzińska-Kołat
- Boruta Medical Center, 95-100 Zgierz, Poland; Department of Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Damian Kołat
- Boruta Medical Center, 95-100 Zgierz, Poland; Department of Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Monika Burzyńska
- Department of Epidemiology and Biostatistics, Social and Preventive Medicine of the Medical University of Lodz, 90-752 Lodz, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, 01-813 Warsaw, Poland
| | - Mateusz Babicki
- Department of Family Medicine, Wroclaw Medical University, 51-141 Wroclaw, Poland
| |
Collapse
|
43
|
Clinical outcomes of hospitalized COVID-19 patients treated with remdesivir: a retrospective analysis of a large tertiary care center in Germany. Infection 2023; 51:97-108. [PMID: 35553032 PMCID: PMC9098143 DOI: 10.1007/s15010-022-01841-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/26/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE The benefits of antiviral treatment with remdesivir in hospitalized patients with COVID-19 remain controversial. Clinical analyses are needed to demonstrate which patient populations are most likely to benefit. METHODS In a retrospective monocentric analysis, patients with COVID-19 treated between July 1, 2020 and June 30, 2021 at Hospital St. Georg, Leipzig, Germany were evaluated. The primary endpoint was time to clinical improvement, and the secondary endpoint was 28-day mortality. Propensity score matching was used for the endpoint analysis. RESULTS A total of 839 patients were fully evaluated, 68% of whom received specific COVID-19 drug therapy. Remdesivir was used in 31.3% of the patients, corticosteroids in 61.7%, and monoclonal antibodies in 2.3%. While dexamethasone administration was the most common therapeutic approach during the second pandemic wave, combination therapy with remdesivir and corticosteroids predominated during the third wave. Cox regression analysis revealed that combination therapy was not associated with faster clinical improvement (median: 13 days in both matched groups, HR 0.97 [95% CI 0.77-1.21], P = 0.762). By contrast, 28-day mortality was significantly lower in the corticosteroid-remdesivir group (14.8% versus 22.2% in the corticosteroid group, HR 0.60 [95% CI 0.39-0.95], P = 0.03) in the low-care setting. This effect was also demonstrated in a subgroup analysis of patients with remdesivir monotherapy (n = 44) versus standard of care (SOC). CONCLUSION In COVID-19 patients with only mild disease (low-flow oxygen therapy and treatment in a normal ward) who received corticosteroids and/or remdesivir in addition to SOC, early administration of remdesivir was associated with a measurable survival benefit.
Collapse
|
44
|
Xu K, Wang Z, Qin M, Gao Y, Luo N, Xie W, Zou Y, Wang J, Ma X. A systematic review and meta-analysis of the effectiveness and safety of COVID-19 vaccination in older adults. Front Immunol 2023; 14:1113156. [PMID: 36936964 PMCID: PMC10020204 DOI: 10.3389/fimmu.2023.1113156] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
In the coronavirus disease 2019 (COVID-19) pandemic, vaccinations were essential in preventing COVID-19 infections and related mortality in older adults. The objectives of this study were to evaluate the effectiveness and safety of the COVID-19 vaccines in older adults. We systematically searched the electronic bibliographic databases of PubMed, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, Research Square, and OpenGrey, as well as other sources of gray literature, for studies published between January 1, 2020, and October 1, 2022. We retrieved 22 randomized controlled trials (RCTs), with a total of 3,404,696 older adults (aged over 60 years) participating, that were included in the meta-analysis. No significant publication bias was found. In the cumulative meta-analysis, we found that the COVID-19 vaccines were effective in preventing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (OR = 0.38, 95% CI = 0.23-0.65, p = 0.0004) and in reducing the number of COVID-19-related deaths (OR = 0.16, 95% CI = 0.10-0.25, p < 0.00001) in elderly people. Antibody seroconversion (AS) and geometric mean titer (GMT) levels significantly increased in vaccinated older adults [OR = 24.42, 95% CI = 19.29-30.92; standardized mean difference (SMD) = 0.92, 95% CI = 0.64-1.20, respectively]. However, local and systemic adverse events after COVID-19 vaccine administration were found in older adults (OR = 2.57, 95% CI = 1.83-3.62, p < 0.00001). Although vaccination might induce certain adverse reactions in the elderly population, the available evidence showed that the COVID-19 vaccines are effective and tolerated, as shown by the decrease in COVID-19-related deaths in older adults. It needs to be made abundantly clear to elderly people that the advantages of vaccination far outweigh any potential risks. Therefore, COVID-19 vaccination should be considered as the recommended strategy for the control of this disease by preventing SARS-CoV-2 infection and related deaths in older adults. More RCTs are needed to increase the certainty of the evidence and to verify our conclusions. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022319698, identifier CRD42022319698.
Collapse
Affiliation(s)
- Kun Xu
- School of Health Management, Xihua University, Chengdu, China
| | - Zihan Wang
- Faculty of Geosciences and Environmental Engineering, Southwest Jiaotong University, Chengdu, China
| | - Maorong Qin
- School of Health Management, Xihua University, Chengdu, China
| | - Yangyu Gao
- School of Health Management, Xihua University, Chengdu, China
| | - Na Luo
- School of Health Management, Xihua University, Chengdu, China
| | - Wanting Xie
- School of Health Management, Xihua University, Chengdu, China
| | - Yihan Zou
- School of Health Management, Xihua University, Chengdu, China
| | - Jie Wang
- School of Food and Biological Engineering, Xihua University, Chengdu, China
| | - Xingming Ma
- School of Health Management, Xihua University, Chengdu, China
- Health Promotion Center, Xihua University, Chengdu, China
- *Correspondence: Xingming Ma,
| |
Collapse
|
45
|
Jeon J, Chin B. Treatment Options for Patients With Mild-to-Moderate Coronavirus Disease 2019 in Korea. J Korean Med Sci 2022; 37:e352. [PMID: 36513054 PMCID: PMC9745683 DOI: 10.3346/jkms.2022.37.e352] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) continues to threaten public health in Korea although several surges have passed in the past 3 years since 2019. Although patients with mild-to-moderate COVID-19 can usually recover at home, antiviral therapy to prevent disease progression and hospitalization is beneficial for those at high risk of progressing to severe COVID-19. The purpose of this article was to review how antivirals have been rolled out for the treatment of COVID-19 and how domestic and international guidelines for their use have evolved. Several evidence-based treatment guidelines have been developed in Korea, including those derived from domestic studies. Although many different antiviral agents were nominated as promising therapeutics at the onset of the pandemic, most failed to show efficacy in clinical trials. Currently, three types of antiviral agents-nirmatrelvir-ritonavir, molnupiravir, and remdesivir-are available in Korea to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Each antiviral has its advantages and disadvantages. For most individuals, nirmatrelvir/ritonavir is preferred because of its high efficacy and convenience of administration. When serious drug interactions occur or are expected with nirmatrelvir/ritonavir administration, 3 days of remdesivir treatment is shown to be a reasonable alternative. Molnupiravir may be prescribed with caution only if no other therapeutic options are available or acceptable.
Collapse
Affiliation(s)
- Jaehyun Jeon
- Research Institute for Public Healthcare, National Medical Center, Seoul, Korea
- Department of Infectious Diseases, Clinical Infectious Disease Research Center, National Medical Center, Seoul, Korea.
| | - BumSik Chin
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
| |
Collapse
|
46
|
Sen S, Singh B, Biswas G. Corticosteroids: A boon or bane for COVID-19 patients? Steroids 2022; 188:109102. [PMID: 36029810 PMCID: PMC9400384 DOI: 10.1016/j.steroids.2022.109102] [Citation(s) in RCA: 7] [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: 12/26/2021] [Revised: 07/29/2022] [Accepted: 08/19/2022] [Indexed: 01/11/2023]
Abstract
Several drugs and antibodies have been repurposed to treat COVID-19. Since the outcome of the drugs and antibodies clinical studies have been mostly inconclusive or with lesser effects, therefore the need for alternative treatments has become unavoidable. However, corticosteroids, which have a history of therapeutic efficacy against coronaviruses (SARS and MERS), might emerge into one of the pandemic's heroic characters. Corticosteroids serve an immunomodulatory function in the post-viral hyper-inflammatory condition (the cytokine storm, or release syndrome), suppressing the excessive immunological response and preventing multi-organ failure and death. Therefore, corticosteroids have been used to treat COVID-19 patients for more than last two years. According to recent clinical trials and the results of observational studies, corticosteroids can be administered to patients with severe and critical COVID-19 symptoms with a favorable risk-benefit ratio. Corticosteroids like Hydrocortisone, dexamethasone, Prednisolone and Methylprednisolone has been reported to be effective against SARS-CoV-2 virus in comparison to that of non-steroid drugs, by using non-genomic and genomic effects to prevent and reduce inflammation in tissues and the circulation. Clinical trials also show that inhaled budesonide (a synthetic corticosteroid) increases time to recovery and has the potential to reduce hospitalizations or fatalities in persons with COVID-19. There is also a brief overview of the industrial preparation of common glucocorticoids.
Collapse
Affiliation(s)
- Subhadeep Sen
- Department of Chemistry, Cooch Behar Panchanan Barma University, Panchanan Nagar, Cooch Behar 736101, West Bengal, India
| | - Bhagat Singh
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Goutam Biswas
- Department of Chemistry, Cooch Behar Panchanan Barma University, Panchanan Nagar, Cooch Behar 736101, West Bengal, India.
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW To describe the burden of post-COVID respiratory sequelae in posthospital and nonhospitalized COVID-19 survivors and to describe the priorities of clinical management. RECENT FINDINGS Due to varying definitions of 'Long COVID' or 'Post-COVID', the prevalence of post-COVID sequelae or persisting symptoms is challenging to estimate but ranges from 2.3 to 51%. Risk factors for persistent post-COVID symptoms include age, female sex, deprivation, presence of comorbidities; and in posthospital COVID-19 survivors, the severity of acute infection. Common post-COVID respiratory symptoms include breathlessness, cough and chest pain and many individuals also experience exercise intolerance. The most common pulmonary function test abnormality is impaired diffusing capacity for carbon monoxide. In posthospital COVID-19 survivors, the prevalence of interstitial lung damage is 5-11%. Disordered breathing is common in all post-COVID patients and respiratory physiotherapy is helpful. SUMMARY The vast numbers of COVID-19 infections globally implies that a large number of people will be affected by post-COVID sequelae even with conservative estimates. A significant number of people are affected for several months and up to years following acute infection. Post-COVID sequelae have a detrimental impact on quality of life and ability to work.
Collapse
|
48
|
Hengyotmark A, Kusoom W. Physical as Well as Psychological Distress and Coping with Situational Dilemmas Experienced by People Infected with COVID-19: A Mixed Method Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14657. [PMID: 36429368 PMCID: PMC9691110 DOI: 10.3390/ijerph192214657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 06/16/2023]
Abstract
The COVID-19 pandemic caused serious health problems that affected people around the globe. This study aims to understand the physical distress (PhyD), psychological distress (PsyD), and coping experiences among people infected with COVID-19, develop a grounded theory, and examine PhyD, PsyD, and coping among people infected with COVID-19. A sequential exploratory mixed methods strategy is employed. A qualitative procedure is based on a grounded theory; data collection includes observation and in-depth interviews with 25 participants, aged 18 years and above. The quantitative one included 180 participants. Content analysis was applied using the Strauss and Corbin method, and ATLAS.ti software. Descriptive statistics, Pearson's correlation, and the independent t-test were used. Results: The six major themes, including (1) severity of COVID-19 symptoms, (2) death anxiety, (3) uncertainty, (4) barrier to healthcare access, (5) compliance and self-regulation coping (6) post-COVID-19 effects. PhyD, PsyD, and coping were all at a moderate level. The relationship between PhyD, PsyD, and coping was positive. The prevalence in post-COVID-19 effects was 70% (95% CI 63.3-76.4%). There were higher amounts in women than men. The most frequent residual symptoms were decreased activity tolerance (40%), fatigue (33.3%), anxiety and fear of abnormal lungs (33.3%), dyspnea (27.8%), allergy (24.4%), and lung impairment (22.2%). Moreover, the prevalence of more than two symptoms was 54% (95% CI 47.2-61.7%). This study considers that the healthcare providers should be concerned with sufficient healthcare services. Interventions are needed for supporting their recovery from COVID-19 effects.
Collapse
Affiliation(s)
- Arunee Hengyotmark
- Kuakarun Faculty of Nursing, Navamindradhiraj University, Bangkok 10300, Thailand
| | - Wichitra Kusoom
- Faculty of Nursing Science, Bangkokthonburi University, Bangkok 10170, Thailand
| |
Collapse
|
49
|
Poulakou G, Barakat M, Israel RJ, Bacci MR, Álvarez SN, Fonseca FLA, Kainis I, Kalomoiri S, Leontis K, Metallidis S, Panagopoulos P, Papastamopoulos V, Ragognete HG, Ramacciotti E, Rapti V, Sakka V, Syrigos KN, Tsoukalas G, Xynogalas I. Ribavirin aerosol in hospitalized adults with respiratory distress and COVID-19: An open-label trial. Clin Transl Sci 2022; 16:165-174. [PMID: 36326174 PMCID: PMC9841304 DOI: 10.1111/cts.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
There is an unmet medical need for effective treatments for hospitalized patients with coronavirus disease 2019 (COVID-19). Ribavirin is a broad-spectrum antiviral with demonstrated in vitro activity against multiple viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This trial evaluated the potential of ribavirin inhalation solution (ribavirin aerosol) to reduce COVID-19 disease severity in adults with confirmed SARS-CoV-2 infection and a diagnosis of respiratory distress. This phase I, multicenter, open-label, nonrandomized trial was conducted from February 2021 through August 2021. Patients received ribavirin aerosol (100 mg/ml for 30 min or 50 mg/ml for 60 min) twice daily for up to 6 days. The primary end point was change from baseline in clinical status severity, rated on a 7-point scale (1 [death]; 7 [not hospitalized; no limitations on activities]), at day 7 (or end-of-treatment/early termination) and day 30 (follow-up). Fifty-one patients were treated with ribavirin aerosol (mean age, 51.5 years; 78.4% men); mean number of doses was 9.7 (range, 1-12). Improvement of ≥1 level in clinical status severity was observed in 31.4% (16/51) and 78.4% (40/51) of patients at end-of-treatment and day 30, respectively. Of 21 patients who required a ventilator, 16 (76.2%) were able to discontinue ventilator use. Five patients (9.8%) died between end-of-treatment and day 30. Three patients (5.9%) discontinued study treatment due to adverse events. No deaths were considered related to study treatment. These data provide preliminary evidence that ribavirin aerosol may be an efficacious treatment for respiratory distress in adults with COVID-19.
Collapse
Affiliation(s)
- Garyfallia Poulakou
- Third Department of Internal Medicine, “Sotiria” General Hospital of Chest DiseasesNational and Kapodistrian University of Athens School of MedicineAthensGreece
| | | | | | - Marcelo R. Bacci
- Clinical Analysis Division, Praxis Pesquisa MedicaCentro Universitário Faculdade de Medicina do ABCSanto AndreSão PauloBrazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Righi E, Lambertenghi L, Gorska A, Sciammarella C, Ivaldi F, Mirandola M, Sartor A, Tacconelli E. Impact of COVID-19 and Antibiotic Treatments on Gut Microbiome: A Role for Enterococcus spp. Biomedicines 2022; 10:2786. [PMID: 36359311 PMCID: PMC9687172 DOI: 10.3390/biomedicines10112786] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 08/29/2023] Open
Abstract
OBJECTIVE Several studies showed the substantial use of antibiotics and increased risk of antimicrobial resistant infections in patients with COVID-19. The impact of COVID-19-related treatments and antibiotics on gut dysbiosis has not been clarified. DESIGN The prospective cohort study included hospitalized COVID-19 patients (April-December 2020). The gut microbiome composition was analysed by 16S sequencing. The gut diversity and changes in opportunistic bacteria (OBs) or symbionts were analysed according to clinical parameters, laboratory markers of disease progression, type of non-antibiotic COVID-19 treatments (NACT) and type, WHO AWaRe group, and duration of antibiotic therapy (AT). RESULTS A total of 82 patients (mean age 66 ± 13 years, 70% males) were enrolled. The relative abundance of Enterococcus was significantly correlated with duration of hospitalization, intensive care unit stay, O2 needs, and D-dimer, ferritin, and IL-6 blood levels. The presence of Enterococcus showed the highest number of correlations with NACT, AT, and AT + NACT (e.g., hydroxychloroquine ± lopinavir/ritonavir) and increased relative abundance with AWaRe Watch/Reserve antibiotics, AT duration, and combinations. Abundance of Dorea, Agathobacter, Roseburia, and Barnesiella was negatively correlated with AT and corticosteroids use. Patients with increased IL-6, D-dimer, and ferritin levels receiving AT were more likely to show dysbiosis with increased abundance of Enterococcus and Bilophila bacteria and decreased abundance of Roseburia compared with those not receiving AT. CONCLUSION Microbiome diversity is affected by COVID-19 severity. In this context, antibiotic treatment may shift the gut microbiome composition towards OBs, particularly Enterococcus. The impact of treatment-driven dysbiosis on OBs infections and long-term consequences needs further study to define the role of gut homeostasis in COVID-19 recovery and inform targeted interventions.
Collapse
Affiliation(s)
- Elda Righi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Lorenza Lambertenghi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Anna Gorska
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Concetta Sciammarella
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Federico Ivaldi
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Massimo Mirandola
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Assunta Sartor
- Microbiology Unit, Udine University Hospital, 33100 Udine, Italy
| | - Evelina Tacconelli
- Infectious Diseases Unit, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| |
Collapse
|